Subscribe to RSS

DOI: 10.1055/a-2566-4073
Sleep as the Foundation of Brain Health
- Abstract
- Sleep Physiology and Brain Health: Mechanisms, Homeostasis, and Implications
- Circadian Rhythms and Brain Health
- Sleep Architecture Across the Lifespan
- Memory Consolidation and Synaptic Homeostasis During Sleep
- Sleep and Waste Clearance
- Consequences of Sleep Deprivation on Brain Health
- Sleep in Neurodegenerative and Neurovascular Diseases
- Therapeutic Implications and Future Directions
- Role of Wearable/Nearable Devices and Artificial Intelligence
- Take-home Messages and Conclusion
- References
Abstract
Sleep is a vital function, taking about one-third of a human lifetime, and is essential for achieving and maintaining brain health. From homeostatic neurophysiology to emotional and procedural memory processing to clearance of brain waste, sleep and circadian alignment remain paramount. Yet modern lifestyles and clinical practice often dismiss sleep, resulting in profound long-term repercussions. This chapter examines the roles of sleep and circadian rhythms in memory consolidation, synaptic plasticity, and clearance of metabolic waste, highlighting recent advances in neuroscience research. We explore how insufficient and disordered sleep—a public health concern—can impair cognition, escalate neurodegenerative risks, and compromise neurovascular integrity, thereby impacting brain health. These findings underscore the need for comprehensive screening for disturbed sleep and targeted interventions in clinical practice. Emerging interventions and AI-driven technologies may allow early detection and personalized and individualized treatments and improve outcomes. Overall, this chapter reaffirms that healthy sleep is indispensable at any level of neurological disease prevention—on par with the role of diet and exercise in cardiovascular health—and represents the foundation of brain health.
#
Keywords
neurodegeneration - stroke - sleep deprivation - circadian rhythm - glymphatic system - memorySleep health and circadian rhythm regulation are fundamental to human health, playing a critical role in optimizing brain function, mental well-being, immune resilience, and cardiometabolic health. Sleep is an active process facilitating several functions essential for brain health, including memory consolidation,[1] synaptic plasticity,[2] and the clearance of neurotoxic waste products through the glymphatic system.[3] Despite its vital role, over half of the world's adult population falls short of the recommended 7 to 9 hours of nightly sleep—with profound consequences for brain health[4] [5] [6] ([Fig. 1]).


Sleep serves as a cornerstone for preventing and better managing various neurological disorders ([Fig. 2]). Sleep–wake disturbances are closely associated with major neurodegenerative diseases (NDDs), with sleep disorders, alterations in sleep architecture, and circadian rhythm disruptions often preceding their clinical onset.[7] Similarly, sleep disturbances are highly prevalent among post-stroke patients.[8] Notably, even during the developmental stages, sleep loss reduces brain size and produces long-lasting behavioral, morphological, and biochemical abnormalities in later life.[9] Therefore, not only is sleep research paramount in neuroscience, but clinicians also have a unique opportunity to transform patient outcomes and enhance quality of life by making sleep assessment a core component of their practice.


Sleep Physiology and Brain Health: Mechanisms, Homeostasis, and Implications
Two-Process Model and Neuronal Circuits Regulating Sleep
The two-process model proposed by Borbély more than four decades ago still offers an important conceptual framework for understanding sleep–wake regulation.[10] The model proposes that sleep regulation is governed by the interaction of a homeostatic process, which builds up during wakefulness and decreases during sleep (Process S), and a circadian process driven by the body's circadian pacemaker, aligning sleep and wakefulness with the 24-hour light–dark cycle (Process C).[11] Brain nuclei involved in sleep–wake regulation are located within the brainstem and the hypothalamus.
The accumulation of endogenous sleep-promoting substances during the day, such as adenosine, activates the ventrolateral preoptic area (VLPO), which is well described in rodents, and the median preoptic nucleus (MnPO).[12] The rodent VLPO and MnPO, which mainly produce gamma-aminobutyric acid (GABA), act on different neurotransmitters and circuits, inhibiting wake-promoting regions ([Fig. 1]).[12] Other neuronal circuits promoting non-rapid eye movement (NREM) sleep include the parafacial zone in the brainstem (GABA/glycine) and the nNOS neurons in the cortex (nitric oxide),[13] whereas neurons involved in regulating rapid eye movement (REM) sleep are mainly located in the pons.[12] [14] Additionally, at night, the absence of light reduces suprachiasmatic nucleus (SCN) activity, lifting the inhibition on melatonin production that the SCN imposes during the day, allowing melatonin synthesis to occur.[15] Melatonin, in turn, enhances the activity of the VLPO and reduces the activity of arousal-promoting brain regions, which promotes sleep.
#
#
Circadian Rhythms and Brain Health
Circadian rhythms, the intrinsic 24-hour cycles that regulate various physiological processes, play a crucial role in maintaining brain health and function. These rhythms are orchestrated by the SCN in the hypothalamus, which serves as the master clock, synchronizing circadian rhythms throughout the body, including those in the brain. The circadian rhythms are self-sustained, meaning that they exist in the absence of any exogenous signals. However, they can be entrained by environmental factors and are influenced by genetics.[16] Light is the primary stimulus (Zeitgeber) for entraining the SCN rhythm period, along with other environmental factors such as physical activity and temperature. Additionally, light suppresses the secretion of melatonin, a hormone that facilitates the transition to sleep, synthesized in the pineal gland with its receptors located in the SCN, as mentioned earlier.[15] The collective work of Jeffrey Hall, Michael Rosbash, and Michael Young demonstrated that individual cells also have a circadian molecular rhythm, which is regulated via a transcriptional–translational feedback loop. Transcription factors such as circadian locomotor output cycles kaput (CLOCK) or brain and muscle ARNT-like protein 1 (BMALI1) regulate the expression of genes such as the encoding period (PER) and the cryptochrome (CRY) that, once translated, inhibit their own transcription.[17] Furthermore, circadian rhythms also experience changes with aging; the timing of sleep onset is relatively earlier in childhood compared with adulthood and shifts later during adolescence, returning to being earlier again in older adults.[17] These changes are also observed at a molecular level.[18] Aging is also associated with a decrease in sensitivity to environmental cues (e.g., reduced response to light due to increased prevalence of ophthalmological conditions[19]), aligning the circadian rhythms to the natural day/night cycle. Disruptions in circadian rhythms have been linked to neurological disorders, highlighting their significance in brain health, as we discuss in the following sections. Perhaps the most directly affected group is shift workers, as 10% suffer from shift work sleep disorder and are at risk of significant health consequences.[20] Furthermore, a recent meta-analysis showed that shift work was associated with an increased incidence of dementia.[21] Of note, the implications of circadian rhythms disturbances extend beyond cognitive decline; they also encompass metabolic and immune dysregulation, which can further compromise brain health. For example, the interplay between circadian rhythms and the gut microbiome has been identified as a critical factor influencing overall health, including mental well-being.[22]
Melatonin, beyond its role in regulating circadian rhythms, has been suggested to have neuroprotective properties, particularly through its interactions with mitochondrial dynamics and cytoprotection.[23] It acts as an antioxidant, scavenging reactive oxygen species (ROS) and enhancing the activity of antioxidant enzymes. Melatonin and its metabolites reduce oxidative damage in particular in the mitochondria, reducing also mitochondria-related apoptosis, contrasting aging-related mitochondrial dysfunction.[24] In line with that, age-related declines in melatonin levels are associated not only with mitochondrial dysfunction but also with increased neuroinflammation, contributing to the pathophysiology of neurodegenerative diseases.[23] This makes melatonin a potential therapeutic candidate for age-related brain disorders.
#
Sleep Architecture Across the Lifespan
The structure of sleep is broadly categorized into two primary states: non-rapid eye movement (NREM) sleep and rapid eye movement (REM) sleep. NREM sleep is further subdivided into three distinct stages: N1, N2, and N3, with N3 being characterized as slow-wave sleep. Each stage of sleep exhibits unique electroencephalographic (EEG) patterns, eye movement characteristics, and variations in muscle tone. The progression between NREM and REM sleep occurs in a cyclical manner throughout the sleep period.[25] The time course of process S is characterized by an exponential decline in slow-wave activity during both baseline sleep and recovery sleep following sleep deprivation. REM sleep, on the other hand, is mainly regulated by the circadian pacemaker.[11]
Sleep architecture changes across the lifespan; for example, REM sleep is predominant in infants and subtly decreases with age, while the percentage of N3 sleep decreases linearly by approximately 2% per decade of life, plateauing after the age of 60. Consequently, there is an increase in N1 and N2 sleep and wake after sleep onset.[26] Furthermore, sex differences exist, as the decline in N3 sleep is slower in women compared with men.[27] These changes in sleep architecture are accompanied by hormonal changes, as the decline of N3 sleep from early adulthood to midlife is paralleled by a major decline in growth hormone secretion, and increased sleep fragmentation related to aging is associated with higher cortisol levels.[28]
Adenosine: A Key Regulator of Sleep and Cognitive Health
Adenosine plays a crucial role in the regulation of sleep, acting as a homeostatic sleep factor that accumulates during wakefulness (thereby contributing to factor S) and promotes sleep through its interaction with specific adenosine receptors, primarily A1 and A2A receptors.[29] Adenosine mediates a glial–neuronal circuit linking glial metabolic state (modulated by adenosine kinase in astrocytes) to neural-expressed sleep homeostasis.[30] The extracellular accumulation of adenosine in the basal forebrain and preoptic areas during prolonged wakefulness enhances sleep pressure, leading to increased sleep propensity. The sleep-promoting effects of adenosine are further evidenced by the observation that its receptor agonists can increase NREM sleep in rats[31] and cats.[32] The stimulation of A1 adenosine receptors induces EEG changes that are similar to EEG profiles observed after sleep deprivation.[33] On the other hand, antagonists like caffeine, the most consumed stimulant, counteract these effects by blocking adenosine receptors (non-selectively), thereby increasing alertness and reducing the drive for sleep.[34] The mean half-life of caffeine is about 5 hours,[35] and when humans consume approximately 200 mg of caffeine in the early evening, the endogenous melatonin rhythm is delayed by around 40 minutes. Additionally, one study indicated that caffeine lengthens the circadian rhythms and causes a phase delay ex vivo.[36] Therefore, antagonizing the effect of adenosine could also affect sleep through changes in circadian rhythms (Process C). Furthermore, adenosine plays a role in memory performance. For example, the short-term memory impairment induced by scopolamine could be prevented by A1 and A2A antagonists, while the activation of A2A receptors is sufficient to trigger memory impairment in mice.[37] Additionally, one study showed that administration of caffeine enhanced memory consolidation but not retrieval for up to 24 hours according to an inverted U-shaped dose–response curve.[38] Moreover, chronic caffeine consumption was associated with a reduced risk of dementia progression in patients with mild cognitive impairment (MCI), and epidemiological studies found an association between coffee consumption and a lower incidence of Alzheimer's disease (AD).[39] [40] These effects are primarily attributed to the antagonism of adenosine receptors.[41] In line with these findings, a recent study showed that blockade of A2A receptors could reverse early spatial memory defects in transgenic mouse AD models by promoting synaptic plasticity.[42] Although the precise mechanism by which adenosine receptor antagonism might prevent dementia progression remains unclear, a recent study has shown that A2A receptors are upregulated in brain samples from patients with various tauopathies (including frontotemporal dementia, corticobasal degeneration, and Pick's disease), potentially accelerating neurodegeneration.[43] This observation suggests that caffeine, through its antagonistic action on these receptors, may help counteract such effects and slow the neurodegenerative processes. However, current evidence on the relationship between caffeine and brain health is inconsistent across various studies due to factors such as gender, age, genetic predisposition, and caffeine dosage. In patients with severe hypertension, drinking more than two cups of coffee a day was associated with an increased risk of cardiovascular mortality.[44] Additionally, coffee consumption in higher dosages (≥300 mg) is associated with an increased risk of neuropsychiatric symptoms in individuals with mental disorders or dementia.[45] [46] Daytime caffeine consumption was associated with reduced apathy and aberrant motor behavior but more frequent nighttime awakenings in elderly dementia patients, with caffeine intake after 6 pm significantly contributing to sleep disturbances[47] and reported improved sleep after eliminating caffeine.[48] A study on habitual caffeine consumers found that 10 days of caffeine intake led to reduced gray matter (GM) volume in the medial temporal lobe, independent of sleep depth, suggesting that daily caffeine consumption may induce neural plasticity in this region.[49] Similarly, another study on 36 healthy adults found that 5 days of sleep restriction led to increased gray matter in several brain regions, while caffeine consumption during this period resulted in GM reductions, particularly in the thalamus and prefrontal cortex. The findings suggest that sleep restriction alone may trigger adaptive GM changes, whereas caffeine interferes with this process, potentially through mechanisms independent of A1 adenosine receptor availability.[50]
In summary, adenosine regulates sleep, circadian rhythms, and cognition, while caffeine, as its antagonist, enhances alertness but can also disrupt sleep and alter brain structure, with still unclear effects on neurodegenerative processes.
#
#
Memory Consolidation and Synaptic Homeostasis During Sleep
Several studies showed that memory performance improves significantly after a whole 8-hour sleep at night[51] [52] [53]; moreover, 1- to 2-hour naps[54] [55] or even 6-minute naps were shown to provide memory benefits,[56] with longer durations yielding more benefits.[57] Interestingly, for the largest benefits, sleep should happen relatively close to the learning process, as better results on declarative memories have been reported if sleep happens 3 hours compared with 10 hours after learning.[58] [59] Not only the duration of sleep but also the different sleep stages play a role in memory consolidation, with different sleep stages affecting different types of memory. NREM sleep is thought to facilitate declarative, hippocampus-dependent memories (e.g., learning facts), whereas REM sleep is thought to facilitate consolidation of memories that are non-declarative (procedural and emotional aspects, e.g., driving a bike).[60] Notably, some studies showed that declarative and non-declarative memory consolidation can occur at any sleep stage.[61] [62]
Mechanisms involved in memory reinforcement during sleep include memory reactivation and synaptic consolidation.[57] Compelling evidence of the former emerged from a study where participants learned spatial locations paired with an odor. When the odor was reintroduced during N3 but not during REM sleep, it led to a notable enhancement of spatial memory.[63] Moreover, in rats, it was shown that spatial–temporal patterns of neuronal firing occurring during learning are reactivated in the same order during subsequent slow-wave sleep.[64] Slow-wave sleep and sleep spindles are thought to initialize long-term potentiation (LTP)[65] and aid in synaptic consolidation during subsequent REM sleep. Some studies showed a positive correlation between spindle density and post-sleep memory improvement.[66]
Moreover, specific neurotransmitters are essential for memory processes during sleep. For instance, acetylcholine has been shown to facilitate memory consolidation during NREM sleep by stabilizing neural network dynamics and enhancing synaptic plasticity.[67] Additionally, the expression of clock genes, such as Per1, has been implicated in regulating memory consolidation during the day, suggesting that the timing of sleep relative to the circadian cycle can influence memory outcomes.[68] Thus, NREM and REM sleep seem to play complementary roles, interacting with circadian cycles, which ultimately optimize memory consolidation.
#
Sleep and Waste Clearance
The glymphatic system is a recently discovered waste clearance pathway in the brain, primarily active during sleep.[69] It is a functional system analogous to the lymphatic system but unique to the central nervous system, as the brain lacks conventional lymphatic vessels. This system operates through a network of perivascular spaces that allow cerebrospinal fluid (CSF) to flow through the brain, effectively flushing out waste products that accumulate during wakefulness. The glymphatic system's perivascular tunnels connect directly to the brain's subarachnoid spaces, where cardiac rhythm–driven arterial pulsations propel CFS flow.[70] The organizational units of this system are astrocytes and the water channel Aquaporin 4 (AQP4), which face the vessel wall. The observation that sleep and the glymphatic system are related came initially from animal models, whereby sleep was associated with a 60% increase in the interstitial space, resulting in a significant increase in the convective exchange of CSF with interstitial fluid.[71] Indeed, CSF tracer influx was found to be correlated with EEG slow-wave activity, which means that the glymphatic clearance is most active during N3 sleep (which physiologically corresponds to the first hours of sleep).[72] Moreover, glymphatic flow was shown to be regulated by circadian rhythmicity.[73] In humans, it was found that even a single night's sleep deprivation significantly impairs cerebral clearance.[74] The implications of glymphatic dysfunction are particularly interesting in the context of neurodegenerative diseases. By analyzing multimodal data from the AD Neuroimaging Initiative project, it was shown that coupling between the global functional magnetic resonance image (fMRI) signal and CSF influx is correlated with AD-related pathology.[75] Furthermore, interstitial fluid's convective fluxes increase the amyloid-β clearance rate during sleep.[71] However, one recent study has reported findings that challenge the notion of consistently enhanced glymphatic flow during sleep. This study showed that in mice, brain clearance is markedly reduced, not increased, during sleep and anesthesia.[76] This might, however, be related to methodological aspects, e.g., the dye injection technique used, which can affect brain clearance, impacting the findings and methods applied to measure brain clearance. Therefore, although the concept of the glymphatic system is very promising to improve the understanding of neurodegeneration pathophysiology, no methodology has been established to clearly and directly assess its function, and more research is needed in this area.
#
Consequences of Sleep Deprivation on Brain Health
The optimal sleep amount recommended by the American Academy of Sleep Medicine is 7 to 9 hours per night.[5] There is an inverted U-shaped relationship between health outcomes and sleep durations, with sleep duration of less than 6 hours or more than 9 hours being associated with all-cause mortality, older phenotypic age, and increased likelihood of depression.[77] [78] [79] Sleep deprivation, broadly defined as an insufficient amount of sleep, can manifest in two main forms: acute and chronic. Acute sleep deprivation involves a short-term period—usually 24 to 48 hours—of severely restricted or entirely missed sleep, such as pulling an “all-nighter” to meet a deadline. By contrast, chronic sleep deprivation arises from a longer-term pattern of consistently limited sleep over weeks or months. Both forms can yield profound negative consequences on brain health. Numerous studies showed a decrease in attention and working memory after acute sleep deprivation; these include findings such as slower reaction time, reduced vigilance, and lower performance in serial addition/subtraction tasks.[80] [81] Sleep deprivation increases adenosine levels in the basal forebrain, thereby inhibiting the cholinergic system, which plays a role in memory modulation, as mentioned earlier.[82] [83] Regarding clearance of brain waste products, one study showed that sleep deprivation increased overnight amyloid-β38, amyloid-β40, and amyloid-β42 levels by 25 to 30% as measured in the CSF.[81] Using positron emission tomography (PET), another study showed that sleep deprivation increases amyloid-β in the hippocampus and thalamus, an increase which was associated with mood worsening; moreover, the study found an inverse association between amyloid-β burden in subcortical areas and reported night sleep hours.[84] Beyond the acute effect of sleep deprivation, a recent study showed that persistent short sleep duration at ages 50, 60, and 70 compared with persistent normal sleep duration was associated with a 30% increased dementia risk.[85] Moreover, reduced sleep efficiency (defined as the percentage of the time in bed spent sleeping) was associated with incident risk of all neurodegenerative diseases.[86] Another recent study showed that sleep regularity seems to be a stronger predictor of mortality than sleep duration.[87] Although more data are needed to disentangle the complex relationship between sleep duration and brain health, these studies highlight the critical role of healthy sleep in maintaining optimal brain health.
#
Sleep in Neurodegenerative and Neurovascular Diseases
Sleep Disorders and Neurodegenerative Diseases
As mentioned earlier, since sleep and circadian rhythms rely on coordinated functions across multiple regions, nuclei, and neurotransmitters in the central nervous system, it follows that neurodegenerative conditions can easily disrupt these interconnected pathways, thereby contributing to sleep disturbances. In AD, for instance, a reduction in the number of neurons within the SCN,[88] combined with diminished synchrony among various circadian oscillators in the brain,[89] is frequently linked to daytime sleepiness, sleep apnea, insomnia, and fragmented sleep patterns.[90] Likewise, very early in the course of Parkinson's disease (PD) the centers responsible for sleep–wake regulation, as well as the expression of clock genes, are impacted, leading to sleep disturbances in nearly all patients.[91] The most common disorders include rapid eye movement sleep behavior disorder (RBD), insomnia, restless legs syndrome (RLS), and sleep-related breathing disorders. Moreover, excessive daytime sleepiness is common in patients with PD.[91] Sleep problems are not confined to these two prevalent neurodegenerative disorders but also affect patients suffering from other neurodegenerative diseases, such as dementia with Lewy bodies, multiple system atrophy, amyotrophic lateral sclerosis, progressive supranuclear palsy, Huntington's disease, and spinocerebellar ataxias.[7]
Importantly, sleep disturbances are among the critical early markers indicating the onset of neurodegeneration. The best example is RBD, which is characterized by abnormal behaviors, jerks, and/or vocalizations during REM sleep, and the absence of physiological muscle atonia during REM sleep. Longitudinal studies have reported that the vast majority of patients with isolated RBD (i.e., RBD not secondary to a manifest NDD, other neurological diseaser or other sleep disorders such as narcolepsy) eventually develop an α-synuclein-related neurodegenerative disorder (i.e., PD, dementia with Lewy body and multiple system atrophy), whereby the risk for developing neurodegenerative diseases was 33.5% at 5 years follow-up, 82.4% at 10.5 years, and 96.6% at 14 years.[92] Additionally, in patients with isolated RBD, there is a frequent occurrence of subtle prodromal neurodegenerative abnormalities such as hyposmia, subtle motor deficits, constipation, and orthostatic hypotension, along with abnormalities detected on various neurophysiological and autonomic tests as well as on neuroimaging and biofluids (including the presence of pathological α-synuclein aggregates).[93] [94]
Another well-investigated example of the link between sleep and neurodegeneration is sleep-disordered breathing (SDB), characterized by recurrent apneas and hypopneas frequently leading to recurrent arousals from sleep, which has been associated with dementia. The earliest longitudinal evidence came from a community study, where women with SDB were more likely to develop MCI (adjusted HR 1.85; 95% confidence interval (CI) 1.11–3.08) after 5 years follow-up.[95] Similarly, in community-dwelling men participating in the Osteoporotic Fractures in Men Study (MrOS), nocturnal hypoxemia was associated with global cognitive decline both cross-sectionally and after 3.4 years.[96] [97] In the AD Neuroimaging Initiative cohort, patients with SDB were younger at MCI onset (72–77 versus 82–89 years), or AD dementia onset (83 versus 88 years) after 3 years follow-up, compared with participants without SDB. Moreover, continuous positive airway pressure (CPAP) use was associated with older age at MCI onset.[98] In the Atherosclerosis Risk in Communities Study, initially no association was found between SDB and incident risk of dementia after 15 years. However, a later study from the same cohort, including more participants, indicated that the relationship might not be linear as severe OSA (≥30 apnea–hypopnea events/hour) versus no OSA (<5 apnea–hypopnea events/hour) was associated with a higher risk of all-cause dementia (risk ratio 2.35).[99] A meta-analysis covering 4.3 million individuals indicated that those with SDB were 26% (risk ratio, 1.26; 95% CI, 1.05–1.50) more likely to develop cognitive impairment.[100] In a sample of 53,000 Medicare beneficiaries with SDB, PAP treatment and adherence were associated with lower odds of incident diagnosis of AD (odds ratio [OR] 0.78, 95% CI, 0.69–0.8).[101] Furthermore, imaging studies in cognitively asymptomatic individuals with SDB showed medial temporal lobe atrophy, which may increase the risk of developing memory impairment.[102] Recently, metrics other than the classical apnea–hypopnea index, such as hypoxic burden, were shown to predict cognitive dysfunction in sleep apnea patients.[103]
Besides SDB, other sleep disorders and disturbances, such as insomnia, excessive daytime sleepiness, and sleep fragmentation, predate the diagnosis of NDDs.[104] [105] [106] Furthermore, beyond manifest sleep disturbances, we found that early sleep architecture changes, as measured by polysomnography, are present up to 12 years before the diagnosis of NDDs. These include reduced REM sleep, N3 sleep, sleep efficiency, as well as increased wake in sleep period time.[86] Similarly, in a subset of the Framingham Heart Study (FHS), lower REM sleep percentage and longer REM sleep latency were both associated with a higher risk of incident dementia after 12 years.[107] Additionally, in the MrOS study lower REM sleep percentage and lower α/theta ratio in sleep EEG were associated with incident PD after a median follow-up of 9.8 years.[108]
Circadian disturbances are also prevalent in NDDs and are associated with their incidence. For example, these patients may exhibit increased nighttime activity and reduced daytime activity, and in some cases even experience a complete reversal or disruption of their normal 24-hour rest–activity cycle. AD patients also experience increased neuropsychiatric symptoms around sunset, referred to as “sundowning,” which is partly attributed to circadian misalignment.[109] To understand circadian changes in patients with NDD, it is important to distinguish circadian phase from circadian amplitude. The former refers to the timing of specific points within the roughly 24-hour cycle—such as when melatonin levels begin to rise or when core body temperature reaches its lowest point.[110] Circadian amplitude, on the other hand, reflects the intensity of the rhythmic variation—often quantified as the difference between the maximum and minimum levels of a measured circadian parameter (like melatonin concentration or body temperature).[110] PD is characterized by a reduction in the circadian amplitude without shifts in circadian phases.[111] In AD, rest–activity rhythm fragmentation[112] and circadian shifts prevail. Regarding the latter, literature findings are mixed, with one study showing phase delay among AD patients[79] and others finding phase advance.[113] Of note, it is still unclear whether neurodegeneration is a consequence or a cause of circadian disturbances, or both. Using data from the UK Biobank and MrOS, variables reflecting circadian rhythm disruption were derived from the accelerometry data and showed that disturbed circadian rhythm is associated with all-cause dementia, PD, and anxiety disorders 6 to 11 years before their clinical onset.[114] [115] Another study found that preclinical AD was associated with rest–activity rhythm fragmentation, and that aging was associated with circadian dysfunction independently of preclinical AD pathology (assessed with PET imaging and amyloid-β pathology).[116] Additionally, a randomized controlled double-blinded study showed that long-term application of bright light treatment (+/− 10,000 lux) reduced cognitive decline by a modest 5% and improved depressive symptoms, while the combination of bright light treatment with melatonin improved sleep efficiency and reduced nocturnal restlessness.[117] In PD patients, bright light exposure (10,000 lux) improved daily activity and reduced daytime sleepiness.[118] Blue light exposure early in the day not only suppresses daytime melatonin but also enhances serotonin levels via the retino-raphe pathway, where photoreceptor signals from the retina stimulate the dorsal raphe nucleus. This process enhances tryptophan metabolism, leading to increased serotonin availability, which is later converted into melatonin in the pineal gland.[119] As a result, natural sunlight exposure during the day ensures optimal serotonin levels, directly influencing nighttime melatonin production. Furthermore, infrared light has been identified as a promising intervention for neurodegenerative diseases, as it stimulates cytochrome c oxidase (complex IV) in the electron transport chain, enhancing ATP production and cellular energy metabolism.[120] This process also facilitates the reduction of oxidative stress and supports mitochondrial resilience, reinforcing its potential therapeutic role in aging and neuroprotection.
Insights from animal models also provide evidence of circadian disruption being linked to NDDs pathology. For instance, a mouse model of amyloidosis-β with deletion of the core clock gene Bmal1 has demonstrated that loss of central circadian rhythms leads to disruption of daily hippocampal interstitial fluid amyloid-β oscillations and accelerates plaque accumulation.[121] Another study found that orexin regulates the hippocampal clock and circadian oscillation of AD risk genes.[122] Overall, circadian science is evolving and provides new insights into early detection and modulation of NDDs.
#
Sleep and Stroke
Cardiovascular diseases remain the leading cause of all-cause mortality.[123] In the context of sleep and neurovascular health, the most relevant sleep disorder is SDB, as it affects around 20% of the general population. SDB is related to wake-up stroke through mechanisms including non-dipping blood pressure and impaired cerebral hemodynamics.[124] On the other hand, SDB is common in stroke patients; a meta-analysis of 2,343 ischemic or hemorrhagic stroke and TIA patients found the frequency of SDB with AHI > 5 to be 72% and with AHI > 20 to be 38%.[125] In both the Wisconsin Sleep Cohort Study (1,189 subjects followed up for 4 years) and the Sleep Heart Health Study (5,422 participants followed up for 8.7 years), there was a 2- to 3-fold increase in the risk of stroke for subjects with untreated SDB.[126] [127] Silent brain infarctions on MRI were more common among patients with moderate to severe sleep apnea than among control subjects or patients with mild sleep apnea.[128] Importantly, this is an easily modifiable risk factor, as CPAP therapy with good adherence, which is the gold standard for SDB, is associated with stroke risk reduction.[129] Notably, poor compliance with PAP therapy negates benefits concerning stroke risk reduction. Beyond SDB, other sleep disorders, such as RLS, seem to possibly occur post-stroke in the setting of lesions in the basal ganglia/corona radiata.[130] RLS is also likely linked to a higher prevalence of coronary artery disease and cardiovascular disease, and this relationship appears to be stronger in individuals who experience more frequent or severe RLS symptoms.[131] Furthermore, a recent study showed an association between insomnia and objective (<6 hours confirmed by polysomnography) but not subjective short-sleep duration and incident cardiovascular diseases after a median follow-up of 11.4 years.[132] Moreover, persistent insomnia symptoms were associated with an increased risk of stroke in younger adults.[133]
Sleep changes are also frequent post-stroke, with poor sleep quality affecting up to 53% of stroke patients.[134] Sleep architecture was shown to be significantly affected in 104 post-stroke patients compared with 162 controls; changes included reduced total sleep time and sleep efficiency, and increased wakefulness. Additionally, the percentage of REM sleep was shown to be negatively associated with stroke severity. Of note, in the same study, sleep quality and sleep architecture improved but did not normalize during the 3 months after stroke.[135]
In 437 stroke patients over 2 years follow-up (after one to seven days, one month, three months, twelve months and twenty-four months), the frequencies of excessive daytime sleepiness (EDS), fatigue, and insomnia varied between 10 and 14% for EDS, 22 and 28% for fatigue, and 20 and 28% for insomnia over the five follow-up visits.[136] Additionally, depending on the infarct area, not only RLS but also other sleep disorders such as RBD or hypersomnolence disease can occur, albeit more rarely as small strategic areas need to be affected by the stroke to cause these symptoms.[8] In conclusion, the complex bidirectional relationship between stroke and sleep requires further investigation, given the high prevalence of these diseases and the potential implications of treating sleep disorders in stroke prevention and management.
#
#
Therapeutic Implications and Future Directions
The most straightforward and broadly applicable intervention to safeguard against the deleterious effects of sleep disturbances is maintaining adequate sleep duration and rhythmicity and practicing sound sleep hygiene. Fundamental measures include establishing a consistent bedtime and rise time, avoiding stimulants like caffeine in the late afternoon or evening, and ensuring an environment conducive to rest—cool, dark, and free from excessive noise. Such non-pharmacological strategies can substantially improve sleep quality, reduce nocturnal awakenings, and bolster cognitive function, especially in individuals experiencing early, mild sleep problems.[5] [137] Moreover, by raising public awareness of sleep disorders, early detection and intervention become more feasible, reducing the risk of serious complications. Strengthening sleep education and implementing targeted healthcare strategies are, therefore, critical steps toward improving overall sleep health and preventing long-term adverse outcomes impacting brain health.[6]
When these general interventions fail to sufficiently address persistent sleep issues, targeted treatments become necessary. For example, those suffering from insomnia can often benefit from Cognitive Behavioral Therapy for Insomnia (CBT-I). This evidence-based and currently first-line approach combines behavioral techniques (e.g., stimulus control, sleep restriction) with cognitive strategies aimed at alleviating anxiety or maladaptive thoughts about sleep.[138] Several studies have shown that CBT-I not only improves sleep continuity and reduces insomnia severity but may also have a protective effect against cognitive decline and reduce the rate of amyloid-β deposition in older adults.[139] Additional interventions, such as melatonin supplementation (to recalibrate the sleep–wake cycle), emerge as promising candidates for mitigating AD pathology.[140] A meta-analysis of 22 randomized controlled trials in patients with AD who received more than 12 weeks of melatonin treatment showed improvements in Mini-Mental State Examination scores.[141] Additionally, higher melatonin levels in community-dwelling adults correlated with lower cognitive impairment.[142] The orexin (hypocretin) signaling pathway offers a compelling frontier for novel therapeutic strategies aimed at optimizing sleep architecture. Orexins regulate wakefulness and stabilize sleep–wake transitions; thus, selective antagonists (orexin receptor blockers) have been effective and are currently available for treating insomnia,[143] with emerging research suggesting, beyond insomnia treatment, potential roles in modulating disease course in AD.[144] PD patients might instead benefit from orexin agonists,[145] as PD is associated with lower orexin levels.[146] As discussed above, treatment of obstructive sleep apnea reduces the risk of neurovascular and neurodegenerative disorders. Newer interventions aiming at directly improving the sleep structure, such as acoustic stimulation (to enhance slow-wave sleep), are promising as they could improve glymphatic clearance and memory consolidation, but research on this topic is still ongoing,[136] [137] and longitudinal data to demonstrate such benefits are needed.
#
Role of Wearable/Nearable Devices and Artificial Intelligence
In parallel with pharmacologic advances, the steadily increasing availability of wearable and nearable devices and AI-driven technologies for early identification and monitoring of sleep alterations are starting to reshape both clinical practice and large-scale screening. Automated analyses of polysomnography, actigraphy, other devices used in the research field, or even consumer sleep-tracker data can detect subtle, prodromal sleep pattern disruptions—such as sleep fragmentation or microarousals—years before overt neurological symptoms appear.[147] [148] [149] Additionally, these technologies not only provide continuous measures of multiple parameters, but they also allow applying a new way to score traditional sleep, introducing the concept of “hypodensity” to score sleep in a probabilistic way instead of relying on the traditional sleep stages.[150] Moreover, these technologies are widely available now and are getting more diverse (wearables, nearables, and airables), offering multifaceted and continuous observation for different groups (e.g., those who do not tolerate sleeping with a smart-watch could use a smart-mattress or a smartphone which can detect movement or snoring) with the potential to unlock new knowledge in preventing, diagnosing early, and managing neurological diseases.[151] Furthermore, the emergence of commercial Large Language Models (LLM; e.g., GPT [OpenAI], LLAMA [Meta], Gemini [Google]) offers new ways in which patients can interact with the technology and get direct answers to their complaints,[152] but the thorough validation of these tools is still needed. By integrating these AI insights into personalized and individualized sleep management plans, clinicians may intervene earlier, tailoring therapies that could meaningfully delay disease onset and progression.
#
Take-home Messages and Conclusion
Sleep is a cornerstone of brain health and should be prioritized accordingly. Subtle sleep disturbances may precede neurological disease by years, providing a critical window for early intervention. Prompt management of obstructive sleep apnea, insomnia, sleep deprivation, or circadian disruptions reduces the risk of occurrence and disability after the onset of stroke and neurodegenerative diseases. Meanwhile, AI-driven sleep trackers and automated analyses will empower clinicians to detect prodromal and preclinical changes, personalize treatments, and optimize outcomes, helping at the same time raising awareness of the relevance of sleep health in the general population. Maintaining a healthy sleep, through consistency in sleep–wake schedules and adequate sleep duration, underpins brain health and overall well-being.
#
#
Conflict of Interest
None declared.
-
References
- 1 Klinzing JG, Niethard N, Born J. Mechanisms of systems memory consolidation during sleep. Nat Neurosci 2019; 22 (10) 1598-1610
- 2 Wang G, Grone B, Colas D, Appelbaum L, Mourrain P. Synaptic plasticity in sleep: learning, homeostasis and disease. Trends Neurosci 2011; 34 (09) 452-463
- 3 Nedergaard M, Goldman SA. Glymphatic failure as a final common pathway to dementia. Science 2020; 370 (6512) 50-56
- 4 Golombek DA, Booi L, Campbell D. et al. Sleep diplomacy: an approach to boosting global brain health. Lancet Healthy Longev 2023; 4 (08) e368-e370
- 5 Watson NF, Badr MS, Belenky G. et al; Consensus Conference Panel, Non-Participating Observers, American Academy of Sleep Medicine Staff. 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 2015; 11 (06) 591-592
- 6 Lim DC, Najafi A, Afifi L. et al; World Sleep Society Global Sleep Health Taskforce. The need to promote sleep health in public health agendas across the globe. Lancet Public Health 2023; 8 (10) e820-e826
- 7 Ibrahim A, Högl B, Stefani A. The bidirectional relationship between sleep and neurodegeneration: actionability to improve brain health. Clinical and Translational Neuroscience 2024; 8: 11
- 8 Mayer-Suess L, Ibrahim A, Moelgg K. et al. Sleep disorders as both risk factors for, and a consequence of, stroke: a narrative review. Int J Stroke 2024; 19 (05) 490-498
- 9 Mirmiran M, Scholtens J, van de Poll NE, Uylings HB, van der Gugten J, Boer GJ. Effects of experimental suppression of active (REM) sleep during early development upon adult brain and behavior in the rat. Brain Res 1983; 283 (2-3): 277-286
- 10 Borbély AA. Circadian rhythm of vigilance in rats: modulation by short light-dark cycles. Neurosci Lett 1975; 1 (01) 67-71
- 11 Borbély A. The two-process model of sleep regulation: beginnings and outlook. J Sleep Res 2022; 31 (04) e13598
- 12 Scammell TE, Arrigoni E, Lipton JO. Neural circuitry of wakefulness and sleep. Neuron 2017; 93 (04) 747-765
- 13 Morairty SR, Dittrich L, Pasumarthi RK. et al. A role for cortical nNOS/NK1 neurons in coupling homeostatic sleep drive to EEG slow wave activity. Proc Natl Acad Sci U S A 2013; 110 (50) 20272-20277
- 14 Eban-Rothschild A, Appelbaum L, de Lecea L. Neuronal mechanisms for sleep/wake regulation and modulatory drive. Neuropsychopharmacology 2018; 43 (05) 937-952
- 15 Zisapel N. New perspectives on the role of melatonin in human sleep, circadian rhythms and their regulation. Br J Pharmacol 2018; 175 (16) 3190-3199
- 16 Leng Y, Musiek ES, Hu K, Cappuccio FP, Yaffe K. Association between circadian rhythms and neurodegenerative diseases. Lancet Neurol 2019; 18 (03) 307-318
- 17 Logan RW, McClung CA. Rhythms of life: circadian disruption and brain disorders across the lifespan. Nat Rev Neurosci 2019; 20 (01) 49-65
- 18 Chen CY, Logan RW, Ma T. et al. Effects of aging on circadian patterns of gene expression in the human prefrontal cortex. Proc Natl Acad Sci U S A 2016; 113 (01) 206-211
- 19 Klein R, Klein BE. The prevalence of age-related eye diseases and visual impairment in aging: current estimates. Invest Ophthalmol Vis Sci 2013; 54 (14) F5 , F13
- 20 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 (08) 1453-1462
- 21 Lee KW, Yang CC, Chen CH, Hung CH, Chuang HY. Shift work is significantly and positively associated with dementia: a meta-analysis study. Front Public Health 2023; 11: 998464
- 22 Teichman EM, O'Riordan KJ, Gahan CGM, Dinan TG, Cryan JF. When rhythms meet the blues: circadian interactions with the microbiota-gut-brain axis. Cell Metab 2020; 31 (03) 448-471
- 23 Verma AK, Singh S, Rizvi SI. Therapeutic potential of melatonin and its derivatives in aging and neurodegenerative diseases. Biogerontology 2023; 24 (02) 183-206
- 24 Bocheva G, Bakalov D, Iliev P, Tafradjiiska-Hadjiolova R. The vital role of melatonin and its metabolites in the neuroprotection and retardation of brain aging. Int J Mol Sci 2024; 25 (10) 25
- 25 Troester MQuan SF, Berry RB. et al, for the American Academy of Sleep Medicine; The AASM Manual for the Scoring of Sleep and Associated Events: Rules, Terminology and Technical Specifications. Version 3:. Dariel, IL: American Academy of Sleep Medicine; 2023
- 26 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 (07) 1255-1273
- 27 Taporoski TP, Beijamini F, Alexandria S. et al. Gender differences in the relationship between sleep and age in a Brazilian cohort: the Baependi Heart Study. J Sleep Res 2024; e14154 . Epub ahead of print.
- 28 Van Cauter E, Leproult R, Plat L. Age-related changes in slow wave sleep and REM sleep and relationship with growth hormone and cortisol levels in healthy men. JAMA 2000; 284 (07) 861-868
- 29 Porkka-Heiskanen T, Strecker RE, Thakkar M, Bjorkum AA, Greene RW, McCarley RW. Adenosine: a mediator of the sleep-inducing effects of prolonged wakefulness. Science 1997; 276 (5316) 1265-1268
- 30 Bjorness TE, Dale N, Mettlach G. et al. An adenosine-mediated glial-neuronal circuit for homeostatic sleep. J Neurosci 2016; 36 (13) 3709-3721
- 31 Hu Z, Lee CI, Shah VK. et al. Cordycepin increases nonrapid eye movement sleep via adenosine receptors in rats. Evid Based Complement Alternat Med 2013; 2013: 840134
- 32 Feldberg W, Sherwood SL. Behaviour of cats after intraventricular injections of eserine and DFP. J Physiol 1954; 125 (03) 488-500
- 33 Benington JH, Kodali SK, Heller HC. Stimulation of A1 adenosine receptors mimics the electroencephalographic effects of sleep deprivation. Brain Res 1995; 692 (1-2): 79-85
- 34 Nehlig A, Daval JL, Debry G. Caffeine and the central nervous system: mechanisms of action, biochemical, metabolic and psychostimulant effects. Brain Res Brain Res Rev 1992; 17 (02) 139-170
- 35 Arnaud MJ. The pharmacology of caffeine. Prog Drug Res 1987; 31: 273-313
- 36 Oike H, Kobori M, Suzuki T, Ishida N. Caffeine lengthens circadian rhythms in mice. Biochem Biophys Res Commun 2011; 410 (03) 654-658
- 37 Pagnussat N, Almeida AS, Marques DM. et al. Adenosine A(2A) receptors are necessary and sufficient to trigger memory impairment in adult mice. Br J Pharmacol 2015; 172 (15) 3831-3845
- 38 Borota D, Murray E, Keceli G. et al. Post-study caffeine administration enhances memory consolidation in humans. Nat Neurosci 2014; 17 (02) 201-203
- 39 Cao C, Loewenstein DA, Lin X. et al. High blood caffeine levels in MCI linked to lack of progression to dementia. J Alzheimers Dis 2012; 30 (03) 559-572
- 40 Santos C, Costa J, Santos J, Vaz-Carneiro A, Lunet N. Caffeine intake and dementia: systematic review and meta-analysis. J Alzheimers Dis 2010; 20 (Suppl. 01) S187-S204
- 41 Lopes JP, Pliássova A, Cunha RA. The physiological effects of caffeine on synaptic transmission and plasticity in the mouse hippocampus selectively depend on adenosine A1 and A2A receptors. Biochem Pharmacol 2019; 166: 313-321
- 42 Ji Q, Yang Y, Xiong Y. et al. Blockade of adenosine A2A receptors reverses early spatial memory defects in the APP/PS1 mouse model of Alzheimer's disease by promoting synaptic plasticity of adult-born granule cells. Alzheimers Res Ther 2023; 15 (01) 187
- 43 Carvalho K, Faivre E, Pietrowski MJ. et al; NeuroCEB Brain Bank. Exacerbation of C1q dysregulation, synaptic loss and memory deficits in tau pathology linked to neuronal adenosine A2A receptor. Brain 2019; 142 (11) 3636-3654
- 44 Teramoto M, Yamagishi K, Muraki I, Tamakoshi A, Iso H. Coffee and green tea consumption and cardiovascular disease mortality among people with and without hypertension. J Am Heart Assoc 2023; 12 (02) e026477
- 45 Kim E, Robinson NM, Newman BM. A brewed awakening: neuropsychiatric effects of caffeine in older adults. Clin Geriatr Med 2022; 38 (01) 133-144
- 46 Kromhout MA, Rius Ottenheim N, Giltay E, Numans ME, Achterberg WP. Caffeine and neuropsychiatric symptoms in patients with dementia: a systematic review. Exp Gerontol 2019; 122: 85-91
- 47 Kromhout MA, Jongerling J, Achterberg WP. Relation between caffeine and behavioral symptoms in elderly patients with dementia: an observational study. J Nutr Health Aging 2014; 18 (04) 407-410
- 48 de Pooter-Stijnman LMM, Vrijkotte S, Smalbrugge M. Effect of caffeine on sleep and behaviour in nursing home residents with dementia. Eur Geriatr Med 2018; 9 (06) 829-835
- 49 Lin YS, Weibel J, Landolt HP. et al. Daily caffeine intake induces concentration-dependent medial temporal plasticity in humans: a multimodal double-blind randomized controlled trial. Cereb Cortex 2021; 31 (06) 3096-3106
- 50 Lin YS, Lange D, Baur DM. et al. Repeated caffeine intake suppresses cerebral grey matter responses to chronic sleep restriction in an A1 adenosine receptor-dependent manner: a double-blind randomized controlled study with PET-MRI. Sci Rep 2024; 14 (01) 12724
- 51 Korman M, Doyon J, Doljansky J, Carrier J, Dagan Y, Karni A. Daytime sleep condenses the time course of motor memory consolidation. Nat Neurosci 2007; 10 (09) 1206-1213
- 52 Baena D, Cantero JL, Fuentemilla L, Atienza M. Weakly encoded memories due to acute sleep restriction can be rescued after one night of recovery sleep. Sci Rep 2020; 10 (01) 1449
- 53 Ribeiro N, Gounden Y, Quaglino V. A full night's sleep at home improves memory performance in an associative and relational learning task. Dreaming 2020; 30 (02) 171-188
- 54 Tucker MA, Hirota Y, Wamsley EJ, Lau H, Chaklader A, Fishbein W. A daytime nap containing solely non-REM sleep enhances declarative but not procedural memory. Neurobiol Learn Mem 2006; 86 (02) 241-247
- 55 Mednick S, Nakayama K, Stickgold R. Sleep-dependent learning: a nap is as good as a night. Nat Neurosci 2003; 6 (07) 697-698
- 56 Lahl O, Wispel C, Willigens B, Pietrowsky R. An ultra short episode of sleep is sufficient to promote declarative memory performance. J Sleep Res 2008; 17 (01) 3-10
- 57 Diekelmann S, Born J. The memory function of sleep. Nat Rev Neurosci 2010; 11 (02) 114-126
- 58 Gais S, Lucas B, Born J. Sleep after learning aids memory recall. Learn Mem 2006; 13 (03) 259-262
- 59 Holz J, Piosczyk H, Landmann N. et al. The timing of learning before night-time sleep differentially affects declarative and procedural long-term memory consolidation in adolescents. PLoS One 2012; 7 (07) e40963
- 60 Maquet P. The role of sleep in learning and memory. Science 2001; 294 (5544) 1048-1052
- 61 Gais S, Plihal W, Wagner U, Born J. Early sleep triggers memory for early visual discrimination skills. Nat Neurosci 2000; 3 (12) 1335-1339
- 62 Fogel SM, Smith CT, Cote KA. Dissociable learning-dependent changes in REM and non-REM sleep in declarative and procedural memory systems. Behav Brain Res 2007; 180 (01) 48-61
- 63 Rasch B, Büchel C, Gais S, Born J. Odor cues during slow-wave sleep prompt declarative memory consolidation. Science 2007; 315 (5817) 1426-1429
- 64 Wilson MA, McNaughton BL. Reactivation of hippocampal ensemble memories during sleep. Science 1994; 265 (5172) 676-679
- 65 Rosanova M, Ulrich D. Pattern-specific associative long-term potentiation induced by a sleep spindle-related spike train. J Neurosci 2005; 25 (41) 9398-9405
- 66 Morin A, Doyon J, Dostie V. et al. Motor sequence learning increases sleep spindles and fast frequencies in post-training sleep. Sleep 2008; 31 (08) 1149-1156
- 67 Skilling QM, Eniwaye B, Clawson BC. et al. Acetylcholine-gated current translates wake neuronal firing rate information into a spike timing-based code in Non-REM sleep, stabilizing neural network dynamics during memory consolidation. PLOS Comput Biol 2021; 17 (09) e1009424
- 68 Bellfy L, Smies CW, Bernhardt AR. et al. The clock gene Per1 may exert diurnal control over hippocampal memory consolidation. Neuropsychopharmacology 2023; 48 (12) 1789-1797
- 69 Iliff JJ, Wang M, Liao Y. et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci Transl Med 2012; 4 (147) 147ra111
- 70 Mestre H, Tithof J, Du T. et al. Flow of cerebrospinal fluid is driven by arterial pulsations and is reduced in hypertension. Nat Commun 2018; 9 (01) 4878
- 71 Xie L, Kang H, Xu Q. et al. Sleep drives metabolite clearance from the adult brain. Science 2013; 342 (6156) 373-377
- 72 Hablitz LM, Vinitsky HS, Sun Q. et al. Increased glymphatic influx is correlated with high EEG delta power and low heart rate in mice under anesthesia. Sci Adv 2019; 5 (02) eaav5447
- 73 Hablitz LM, Plá V, Giannetto M. et al. Circadian control of brain glymphatic and lymphatic fluid flow. Nat Commun 2020; 11 (01) 4411
- 74 Eide PK, Vinje V, Pripp AH, Mardal KA, Ringstad G. Sleep deprivation impairs molecular clearance from the human brain. Brain 2021; 144 (03) 863-874
- 75 Han F, Chen J, Belkin-Rosen A. et al; Alzheimer's Disease Neuroimaging Initiative. Reduced coupling between cerebrospinal fluid flow and global brain activity is linked to Alzheimer disease-related pathology. PLoS Biol 2021; 19 (06) e3001233
- 76 Miao A, Luo T, Hsieh B. et al. Brain clearance is reduced during sleep and anesthesia. Nat Neurosci 2024; 27 (06) 1046-1050
- 77 Henríquez-Beltrán M, Dreyse J, Jorquera J. et al. The U-shaped association between sleep duration, all-cause mortality and cardiovascular risk in a Hispanic/Latino clinically based cohort. J Clin Med 2023; 12 (15) 4961
- 78 You Y, Chen Y, Liu R. et al. Inverted U-shaped relationship between sleep duration and phenotypic age in US adults: a population-based study. Sci Rep 2024; 14 (01) 6247
- 79 Dong L, Xie Y, Zou X. Association between sleep duration and depression in US adults: a cross-sectional study. J Affect Disord 2022; 296: 183-188
- 80 Alhola P, Polo-Kantola P. Sleep deprivation: impact on cognitive performance. Neuropsychiatr Dis Treat 2007; 3 (05) 553-567
- 81 Lucey BP, Hicks TJ, McLeland JS. et al. Effect of sleep on overnight cerebrospinal fluid amyloid β kinetics. Ann Neurol 2018; 83 (01) 197-204
- 82 Arrigoni E, Chamberlin NL, Saper CB, McCarley RW. Adenosine inhibits basal forebrain cholinergic and noncholinergic neurons in vitro. Neuroscience 2006; 140 (02) 403-413
- 83 Maurer SV, Williams CL. The cholinergic system modulates memory and hippocampal plasticity via its interactions with non-neuronal cells. Front Immunol 2017; 8: 1489
- 84 Shokri-Kojori E, Wang GJ, Wiers CE. et al. β-Amyloid accumulation in the human brain after one night of sleep deprivation. Proc Natl Acad Sci U S A 2018; 115 (17) 4483-4488
- 85 Sabia S, Fayosse A, Dumurgier J. et al. Association of sleep duration in middle and old age with incidence of dementia. Nat Commun 2021; 12 (01) 2289
- 86 Ibrahim A, Cesari M, Heidbreder A. et al. Sleep features and long-term incident neurodegeneration: a polysomnographic study. Sleep 2024; 47 (03) 47
- 87 Windred DP, Burns AC, Lane JM. et al. Sleep regularity is a stronger predictor of mortality risk than sleep duration: a prospective cohort study. Sleep 2024; 47 (01) 253
- 88 Wang JL, Lim AS, Chiang WY. et al. Suprachiasmatic neuron numbers and rest-activity circadian rhythms in older humans. Ann Neurol 2015; 78 (02) 317-322
- 89 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 Rhythms 2011; 26 (02) 160-170
- 90 Iranzo A. Sleep in neurodegenerative diseases. Sleep Med Clin 2016; 11 (01) 1-18
- 91 Stefani A, Högl B. Sleep in Parkinson's disease. Neuropsychopharmacology 2020; 45 (01) 121-128
- 92 Galbiati A, Verga L, Giora E, Zucconi M, Ferini-Strambi L. The risk of neurodegeneration in REM sleep behavior disorder: a systematic review and meta-analysis of longitudinal studies. Sleep Med Rev 2019; 43: 37-46
- 93 Postuma RB, Iranzo A, Hu M. et al. Risk and predictors of dementia and parkinsonism in idiopathic REM sleep behaviour disorder: a multicentre study. Brain 2019; 142 (03) 744-759
- 94 Iranzo A, Stefani A, Serradell M. et al; SINBAR (Sleep Innsbruck Barcelona) group. Characterization of patients with longstanding idiopathic REM sleep behavior disorder. Neurology 2017; 89 (03) 242-248
- 95 Yaffe K, Laffan AM, Harrison SL. et al. Sleep-disordered breathing, hypoxia, and risk of mild cognitive impairment and dementia in older women. JAMA 2011; 306 (06) 613-619
- 96 Blackwell T, Yaffe K, Ancoli-Israel S. et al; Osteoporotic Fractures in Men Study Group. Associations between sleep architecture and sleep-disordered breathing and cognition in older community-dwelling men: the Osteoporotic Fractures in Men Sleep Study. J Am Geriatr Soc 2011; 59 (12) 2217-2225
- 97 Blackwell T, Yaffe K, Laffan A. et al; Osteoporotic Fractures in Men (MrOS) Study Group. Associations of objectively and subjectively measured sleep quality with subsequent cognitive decline in older community-dwelling men: the MrOS sleep study. Sleep 2014; 37 (04) 655-663
- 98 Osorio RS, Gumb T, Pirraglia E. et al; Alzheimer's Disease Neuroimaging Initiative. Sleep-disordered breathing advances cognitive decline in the elderly. Neurology 2015; 84 (19) 1964-1971
- 99 Lutsey PL, Misialek JR, Mosley TH. et al. Sleep characteristics and risk of dementia and Alzheimer's disease: The Atherosclerosis Risk in Communities Study. Alzheimers Dement 2018; 14 (02) 157-166
- 100 Leng Y, McEvoy CT, Allen IE, Yaffe K. Association of sleep-disordered breathing with cognitive function and risk of cognitive impairment: a systematic review and meta-analysis. JAMA Neurol 2017; 74 (10) 1237-1245
- 101 Dunietz GL, Chervin RD, Burke JF, Conceicao AS, Braley TJ. Obstructive sleep apnea treatment and dementia risk in older adults. Sleep 2021; 44 (09) 44
- 102 André C, Kuhn E, Rehel S. et al; Medit-Ageing Research Group. Association of sleep-disordered breathing and medial temporal lobe atrophy in cognitively unimpaired amyloid-positive older adults. Neurology 2023; 101 (04) e370-e385
- 103 Leonhard AG, Kapur VK. Beyond traditional hypoxemia metrics: hypoxic burden as a predictor of cognitive dysfunction in sleep apnea. Sleep (Basel) 2025; 48 (03) zsae292
- 104 Breen DP, Williams-Gray CH, Mason SL, Foltynie T, Barker RA. Excessive daytime sleepiness and its risk factors in incident Parkinson's disease. J Neurol Neurosurg Psychiatry 2013; 84 (02) 233-234
- 105 Sohail S, Yu L, Schneider JA, Bennett DA, Buchman AS, Lim ASP. Sleep fragmentation and Parkinson's disease pathology in older adults without Parkinson's disease. Mov Disord 2017; 32 (12) 1729-1737
- 106 Osorio RS, Pirraglia E, Agüera-Ortiz LF. et al. Greater risk of Alzheimer's disease in older adults with insomnia. J Am Geriatr Soc 2011; 59 (03) 559-562
- 107 Pase MP, Himali JJ, Grima NA. et al. Sleep architecture and the risk of incident dementia in the community. Neurology 2017; 89 (12) 1244-1250
- 108 Otaiku AI. Association of sleep abnormalities in older adults with risk of developing Parkinson's disease. Sleep 2022; 45 (11) 45
- 109 Volicer L, Harper DG, Manning BC, Goldstein R, Satlin A. Sundowning and circadian rhythms in Alzheimer's disease. Am J Psychiatry 2001; 158 (05) 704-711
- 110 Del Olmo M, Schmal C, Mizaikoff C. et al. Are circadian amplitudes and periods correlated? A new twist in the story. F1000 Res 2024; 12: 1077
- 111 Breen DP, Vuono R, Nawarathna U. et al. Sleep and circadian rhythm regulation in early Parkinson disease. JAMA Neurol 2014; 71 (05) 589-595
- 112 Hooghiemstra AM, Eggermont LH, Scheltens P, van der Flier WM, Scherder EJ. The rest-activity rhythm and physical activity in early-onset dementia. Alzheimer Dis Assoc Disord 2015; 29 (01) 45-49
- 113 Naismith SL, Hickie IB, Terpening Z. et al. Circadian misalignment and sleep disruption in mild cognitive impairment. J Alzheimers Dis 2014; 38 (04) 857-866
- 114 Chen S-J, Deng Y-T, Li Y-Z. et al. Association of circadian rhythms with brain disorder incidents: a prospective cohort study of 72242 participants. Transl Psychiatry 2022; 12 (01) 514
- 115 Leng Y, Blackwell T, Cawthon PM, Ancoli-Israel S, Stone KL, Yaffe K. Association of circadian abnormalities in older adults with an increased risk of developing Parkinson disease. JAMA Neurol 2020; 77 (10) 1270-1278
- 116 Musiek ES, Bhimasani M, Zangrilli MA, Morris JC, Holtzman DM, Ju YS. Circadian rest-activity pattern changes in aging and preclinical Alzheimer disease. JAMA Neurol 2018; 75 (05) 582-590
- 117 Riemersma-van der Lek RF, Swaab DF, Twisk J, Hol EM, Hoogendijk WJ, Van Someren EJ. Effect of bright light and melatonin on cognitive and noncognitive function in elderly residents of group care facilities: a randomized controlled trial. JAMA 2008; 299 (22) 2642-2655
- 118 Videnovic A, Klerman EB, Wang W, Marconi A, Kuhta T, Zee PC. Timed light therapy for sleep and daytime sleepiness associated with Parkinson disease: a randomized clinical trial. JAMA Neurol 2017; 74 (04) 411-418
- 119 Lee BH, Hille B, Koh DS. Serotonin modulates melatonin synthesis as an autocrine neurotransmitter in the pineal gland. Proc Natl Acad Sci U S A 2021; 118 (43) 118
- 120 Cardoso FDS, Barrett DW, Wade Z, Gomes da Silva S, Gonzalez-Lima F. Photobiomodulation of cytochrome c oxidase by chronic transcranial laser in young and aged brains. Front Neurosci 2022; 16: 818005
- 121 Kress GJ, Liao F, Dimitry J. et al. Regulation of amyloid-β dynamics and pathology by the circadian clock. J Exp Med 2018; 215 (04) 1059-1068
- 122 Ma Z, Jiang W, Zhang EE. Orexin signaling regulates both the hippocampal clock and the circadian oscillation of Alzheimer's disease-risk genes. Sci Rep 2016; 6: 36035
- 123 GBD 2019 Stroke Collaborators. Global, regional, and national burden of stroke and its risk factors, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Neurol 2021; 20 (10) 795-820
- 124 Baillieul S, Dekkers M, Brill AK. et al. Sleep apnoea and ischaemic stroke: current knowledge and future directions. Lancet Neurol 2022; 21 (01) 78-88
- 125 Johnson KG, Johnson DC. Frequency of sleep apnea in stroke and TIA patients: a meta-analysis. J Clin Sleep Med 2010; 6 (02) 131-137
- 126 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) 1447-1451
- 127 Redline S, Yenokyan G, Gottlieb DJ. et al. Obstructive sleep apnea-hypopnea and incident stroke: the sleep heart health study. Am J Respir Crit Care Med 2010; 182 (02) 269-277
- 128 Minoguchi K, Yokoe T, Tazaki T. et al. Silent brain infarction and platelet activation in obstructive sleep apnea. Am J Respir Crit Care Med 2007; 175 (06) 612-617
- 129 Lin HJ, Yeh JH, Hsieh MT, Hsu CY. Continuous positive airway pressure with good adherence can reduce risk of stroke in patients with moderate to severe obstructive sleep apnea: an updated systematic review and meta-analysis. Sleep Med Rev 2020; 54: 101354
- 130 Lee SJ, Kim JS, Song IU, An JY, Kim YI, Lee KS. Poststroke restless legs syndrome and lesion location: anatomical considerations. Mov Disord 2009; 24 (01) 77-84
- 131 Winkelman JW, Shahar E, Sharief I, Gottlieb DJ. Association of restless legs syndrome and cardiovascular disease in the Sleep Heart Health Study. Neurology 2008; 70 (01) 35-42
- 132 Bertisch SM, Pollock BD, Mittleman MA. et al. Insomnia with objective short sleep duration and risk of incident cardiovascular disease and all-cause mortality: Sleep Heart Health Study. Sleep 2018; 41 (06) 41
- 133 Sawadogo W, Adera T, Alattar M, Perera R, Burch JB. Association between insomnia symptoms and trajectory with the risk of stroke in the Health and Retirement Study. Neurology 2023; 101 (05) e475-e488
- 134 Luo Y, Yu G, Liu Y, Zhuge C, Zhu Y. Sleep quality after stroke: a systematic review and meta-analysis. Medicine (Baltimore) 2023; 102 (20) e33777
- 135 Miano S, Fanfulla F, Nobili L. et al. SAS CARE 1: sleep architecture changes in a cohort of patients with ischemic stroke/TIA. Sleep Med 2022; 98: 106-113
- 136 Duss SB, Bauer-Gambelli SA, Bernasconi C. et al. Frequency and evolution of sleep-wake disturbances after ischemic stroke: a 2-year prospective study of 437 patients. Sleep Med 2023; 101: 244-251
- 137 Varadharasu S, Das N. Sleep hygiene efficacy on quality of sleep and mental ability among insomniac patients. J Family Med Prim Care 2024; 13 (10) 4693-4698
- 138 Riemann D, Baglioni C, Bassetti C. et al. European guideline for the diagnosis and treatment of insomnia. J Sleep Res 2017; 26 (06) 675-700
- 139 Siengsukon CF, Nelson E, Williams-Cooke C. et al. Cognitive behavioral therapy for insomnia to enhance cognitive function and reduce the rate of Aβ deposition in older adults with symptoms of insomnia: a single-site randomized pilot clinical trial protocol. Contemp Clin Trials 2020; 99: 106190
- 140 Zhang Z, Xue P, Bendlin BB. et al. Melatonin: a potential nighttime guardian against Alzheimer's. Mol Psychiatry 2025; 30 (01) 237-250
- 141 Sumsuzzman DM, Choi J, Jin Y, Hong Y. Neurocognitive effects of melatonin treatment in healthy adults and individuals with Alzheimer's disease and insomnia: a systematic review and meta-analysis of randomized controlled trials. Neurosci Biobehav Rev 2021; 127: 459-473
- 142 Obayashi K, Saeki K, Iwamoto J. et al. Physiological levels of melatonin relate to cognitive function and depressive symptoms: the HEIJO-KYO cohort. J Clin Endocrinol Metab 2015; 100 (08) 3090-3096
- 143 Mignot E, Mayleben D, Fietze I. et al; investigators. Safety and efficacy of daridorexant in patients with insomnia disorder: results from two multicentre, randomised, double-blind, placebo-controlled, phase 3 trials. Lancet Neurol 2022; 21 (02) 125-139
- 144 Zhou M, Tang S. Effect of a dual orexin receptor antagonist on Alzheimer's disease: sleep disorders and cognition. Front Med (Lausanne) 2023; 9: 984227
- 145 Evans R, Kimura H, Alexander R. et al. Orexin 2 receptor-selective agonist danavorexton improves narcolepsy phenotype in a mouse model and in human patients. Proc Natl Acad Sci U S A 2022; 119 (35) e2207531119
- 146 Suresh V, Bardhan M, Ghosh S. et al. Exploring the role of Orexin-A neuropeptide in Parkinson's disease: a systematic review and meta-analysis. Clin Neurol Neurosurg 2024; 242: 108320
- 147 Cesari M, Stefani A, Penzel T. et al. Interrater sleep stage scoring reliability between manual scoring from two European sleep centers and automatic scoring performed by the artificial intelligence-based Stanford-STAGES algorithm. J Clin Sleep Med 2021; 17 (06) 1237-1247
- 148 Cesari M, Portscher A, Stefani A. et al. Machine learning predicts phenoconversion from polysomnography in isolated REM sleep behavior disorder. Brain Sci 2024; 14 (09) 871
- 149 Zhao H, Cao J, Xie J. et al. Wearable sensors and features for diagnosis of neurodegenerative diseases: a systematic review. Digit Health 2023;9:20552076231173569
- 150 Stephansen JB, Olesen AN, Olsen M. et al. Neural network analysis of sleep stages enables efficient diagnosis of narcolepsy. Nat Commun 2018; 9 (01) 5229
- 151 de Zambotti M, Goldstein C, Cook J. et al. State of the science and recommendations for using wearable technology in sleep and circadian research. Sleep 2024; 47 (04) 47
- 152 Thirunavukarasu AJ, Ting DSJ, Elangovan K, Gutierrez L, Tan TF, Ting DSW. Large language models in medicine. Nat Med 2023; 29 (08) 1930-1940
Address for correspondence
Publication History
Accepted Manuscript online:
26 March 2025
Article published online:
25 April 2025
© 2025. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)
Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA
-
References
- 1 Klinzing JG, Niethard N, Born J. Mechanisms of systems memory consolidation during sleep. Nat Neurosci 2019; 22 (10) 1598-1610
- 2 Wang G, Grone B, Colas D, Appelbaum L, Mourrain P. Synaptic plasticity in sleep: learning, homeostasis and disease. Trends Neurosci 2011; 34 (09) 452-463
- 3 Nedergaard M, Goldman SA. Glymphatic failure as a final common pathway to dementia. Science 2020; 370 (6512) 50-56
- 4 Golombek DA, Booi L, Campbell D. et al. Sleep diplomacy: an approach to boosting global brain health. Lancet Healthy Longev 2023; 4 (08) e368-e370
- 5 Watson NF, Badr MS, Belenky G. et al; Consensus Conference Panel, Non-Participating Observers, American Academy of Sleep Medicine Staff. 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 2015; 11 (06) 591-592
- 6 Lim DC, Najafi A, Afifi L. et al; World Sleep Society Global Sleep Health Taskforce. The need to promote sleep health in public health agendas across the globe. Lancet Public Health 2023; 8 (10) e820-e826
- 7 Ibrahim A, Högl B, Stefani A. The bidirectional relationship between sleep and neurodegeneration: actionability to improve brain health. Clinical and Translational Neuroscience 2024; 8: 11
- 8 Mayer-Suess L, Ibrahim A, Moelgg K. et al. Sleep disorders as both risk factors for, and a consequence of, stroke: a narrative review. Int J Stroke 2024; 19 (05) 490-498
- 9 Mirmiran M, Scholtens J, van de Poll NE, Uylings HB, van der Gugten J, Boer GJ. Effects of experimental suppression of active (REM) sleep during early development upon adult brain and behavior in the rat. Brain Res 1983; 283 (2-3): 277-286
- 10 Borbély AA. Circadian rhythm of vigilance in rats: modulation by short light-dark cycles. Neurosci Lett 1975; 1 (01) 67-71
- 11 Borbély A. The two-process model of sleep regulation: beginnings and outlook. J Sleep Res 2022; 31 (04) e13598
- 12 Scammell TE, Arrigoni E, Lipton JO. Neural circuitry of wakefulness and sleep. Neuron 2017; 93 (04) 747-765
- 13 Morairty SR, Dittrich L, Pasumarthi RK. et al. A role for cortical nNOS/NK1 neurons in coupling homeostatic sleep drive to EEG slow wave activity. Proc Natl Acad Sci U S A 2013; 110 (50) 20272-20277
- 14 Eban-Rothschild A, Appelbaum L, de Lecea L. Neuronal mechanisms for sleep/wake regulation and modulatory drive. Neuropsychopharmacology 2018; 43 (05) 937-952
- 15 Zisapel N. New perspectives on the role of melatonin in human sleep, circadian rhythms and their regulation. Br J Pharmacol 2018; 175 (16) 3190-3199
- 16 Leng Y, Musiek ES, Hu K, Cappuccio FP, Yaffe K. Association between circadian rhythms and neurodegenerative diseases. Lancet Neurol 2019; 18 (03) 307-318
- 17 Logan RW, McClung CA. Rhythms of life: circadian disruption and brain disorders across the lifespan. Nat Rev Neurosci 2019; 20 (01) 49-65
- 18 Chen CY, Logan RW, Ma T. et al. Effects of aging on circadian patterns of gene expression in the human prefrontal cortex. Proc Natl Acad Sci U S A 2016; 113 (01) 206-211
- 19 Klein R, Klein BE. The prevalence of age-related eye diseases and visual impairment in aging: current estimates. Invest Ophthalmol Vis Sci 2013; 54 (14) F5 , F13
- 20 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 (08) 1453-1462
- 21 Lee KW, Yang CC, Chen CH, Hung CH, Chuang HY. Shift work is significantly and positively associated with dementia: a meta-analysis study. Front Public Health 2023; 11: 998464
- 22 Teichman EM, O'Riordan KJ, Gahan CGM, Dinan TG, Cryan JF. When rhythms meet the blues: circadian interactions with the microbiota-gut-brain axis. Cell Metab 2020; 31 (03) 448-471
- 23 Verma AK, Singh S, Rizvi SI. Therapeutic potential of melatonin and its derivatives in aging and neurodegenerative diseases. Biogerontology 2023; 24 (02) 183-206
- 24 Bocheva G, Bakalov D, Iliev P, Tafradjiiska-Hadjiolova R. The vital role of melatonin and its metabolites in the neuroprotection and retardation of brain aging. Int J Mol Sci 2024; 25 (10) 25
- 25 Troester MQuan SF, Berry RB. et al, for the American Academy of Sleep Medicine; The AASM Manual for the Scoring of Sleep and Associated Events: Rules, Terminology and Technical Specifications. Version 3:. Dariel, IL: American Academy of Sleep Medicine; 2023
- 26 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 (07) 1255-1273
- 27 Taporoski TP, Beijamini F, Alexandria S. et al. Gender differences in the relationship between sleep and age in a Brazilian cohort: the Baependi Heart Study. J Sleep Res 2024; e14154 . Epub ahead of print.
- 28 Van Cauter E, Leproult R, Plat L. Age-related changes in slow wave sleep and REM sleep and relationship with growth hormone and cortisol levels in healthy men. JAMA 2000; 284 (07) 861-868
- 29 Porkka-Heiskanen T, Strecker RE, Thakkar M, Bjorkum AA, Greene RW, McCarley RW. Adenosine: a mediator of the sleep-inducing effects of prolonged wakefulness. Science 1997; 276 (5316) 1265-1268
- 30 Bjorness TE, Dale N, Mettlach G. et al. An adenosine-mediated glial-neuronal circuit for homeostatic sleep. J Neurosci 2016; 36 (13) 3709-3721
- 31 Hu Z, Lee CI, Shah VK. et al. Cordycepin increases nonrapid eye movement sleep via adenosine receptors in rats. Evid Based Complement Alternat Med 2013; 2013: 840134
- 32 Feldberg W, Sherwood SL. Behaviour of cats after intraventricular injections of eserine and DFP. J Physiol 1954; 125 (03) 488-500
- 33 Benington JH, Kodali SK, Heller HC. Stimulation of A1 adenosine receptors mimics the electroencephalographic effects of sleep deprivation. Brain Res 1995; 692 (1-2): 79-85
- 34 Nehlig A, Daval JL, Debry G. Caffeine and the central nervous system: mechanisms of action, biochemical, metabolic and psychostimulant effects. Brain Res Brain Res Rev 1992; 17 (02) 139-170
- 35 Arnaud MJ. The pharmacology of caffeine. Prog Drug Res 1987; 31: 273-313
- 36 Oike H, Kobori M, Suzuki T, Ishida N. Caffeine lengthens circadian rhythms in mice. Biochem Biophys Res Commun 2011; 410 (03) 654-658
- 37 Pagnussat N, Almeida AS, Marques DM. et al. Adenosine A(2A) receptors are necessary and sufficient to trigger memory impairment in adult mice. Br J Pharmacol 2015; 172 (15) 3831-3845
- 38 Borota D, Murray E, Keceli G. et al. Post-study caffeine administration enhances memory consolidation in humans. Nat Neurosci 2014; 17 (02) 201-203
- 39 Cao C, Loewenstein DA, Lin X. et al. High blood caffeine levels in MCI linked to lack of progression to dementia. J Alzheimers Dis 2012; 30 (03) 559-572
- 40 Santos C, Costa J, Santos J, Vaz-Carneiro A, Lunet N. Caffeine intake and dementia: systematic review and meta-analysis. J Alzheimers Dis 2010; 20 (Suppl. 01) S187-S204
- 41 Lopes JP, Pliássova A, Cunha RA. The physiological effects of caffeine on synaptic transmission and plasticity in the mouse hippocampus selectively depend on adenosine A1 and A2A receptors. Biochem Pharmacol 2019; 166: 313-321
- 42 Ji Q, Yang Y, Xiong Y. et al. Blockade of adenosine A2A receptors reverses early spatial memory defects in the APP/PS1 mouse model of Alzheimer's disease by promoting synaptic plasticity of adult-born granule cells. Alzheimers Res Ther 2023; 15 (01) 187
- 43 Carvalho K, Faivre E, Pietrowski MJ. et al; NeuroCEB Brain Bank. Exacerbation of C1q dysregulation, synaptic loss and memory deficits in tau pathology linked to neuronal adenosine A2A receptor. Brain 2019; 142 (11) 3636-3654
- 44 Teramoto M, Yamagishi K, Muraki I, Tamakoshi A, Iso H. Coffee and green tea consumption and cardiovascular disease mortality among people with and without hypertension. J Am Heart Assoc 2023; 12 (02) e026477
- 45 Kim E, Robinson NM, Newman BM. A brewed awakening: neuropsychiatric effects of caffeine in older adults. Clin Geriatr Med 2022; 38 (01) 133-144
- 46 Kromhout MA, Rius Ottenheim N, Giltay E, Numans ME, Achterberg WP. Caffeine and neuropsychiatric symptoms in patients with dementia: a systematic review. Exp Gerontol 2019; 122: 85-91
- 47 Kromhout MA, Jongerling J, Achterberg WP. Relation between caffeine and behavioral symptoms in elderly patients with dementia: an observational study. J Nutr Health Aging 2014; 18 (04) 407-410
- 48 de Pooter-Stijnman LMM, Vrijkotte S, Smalbrugge M. Effect of caffeine on sleep and behaviour in nursing home residents with dementia. Eur Geriatr Med 2018; 9 (06) 829-835
- 49 Lin YS, Weibel J, Landolt HP. et al. Daily caffeine intake induces concentration-dependent medial temporal plasticity in humans: a multimodal double-blind randomized controlled trial. Cereb Cortex 2021; 31 (06) 3096-3106
- 50 Lin YS, Lange D, Baur DM. et al. Repeated caffeine intake suppresses cerebral grey matter responses to chronic sleep restriction in an A1 adenosine receptor-dependent manner: a double-blind randomized controlled study with PET-MRI. Sci Rep 2024; 14 (01) 12724
- 51 Korman M, Doyon J, Doljansky J, Carrier J, Dagan Y, Karni A. Daytime sleep condenses the time course of motor memory consolidation. Nat Neurosci 2007; 10 (09) 1206-1213
- 52 Baena D, Cantero JL, Fuentemilla L, Atienza M. Weakly encoded memories due to acute sleep restriction can be rescued after one night of recovery sleep. Sci Rep 2020; 10 (01) 1449
- 53 Ribeiro N, Gounden Y, Quaglino V. A full night's sleep at home improves memory performance in an associative and relational learning task. Dreaming 2020; 30 (02) 171-188
- 54 Tucker MA, Hirota Y, Wamsley EJ, Lau H, Chaklader A, Fishbein W. A daytime nap containing solely non-REM sleep enhances declarative but not procedural memory. Neurobiol Learn Mem 2006; 86 (02) 241-247
- 55 Mednick S, Nakayama K, Stickgold R. Sleep-dependent learning: a nap is as good as a night. Nat Neurosci 2003; 6 (07) 697-698
- 56 Lahl O, Wispel C, Willigens B, Pietrowsky R. An ultra short episode of sleep is sufficient to promote declarative memory performance. J Sleep Res 2008; 17 (01) 3-10
- 57 Diekelmann S, Born J. The memory function of sleep. Nat Rev Neurosci 2010; 11 (02) 114-126
- 58 Gais S, Lucas B, Born J. Sleep after learning aids memory recall. Learn Mem 2006; 13 (03) 259-262
- 59 Holz J, Piosczyk H, Landmann N. et al. The timing of learning before night-time sleep differentially affects declarative and procedural long-term memory consolidation in adolescents. PLoS One 2012; 7 (07) e40963
- 60 Maquet P. The role of sleep in learning and memory. Science 2001; 294 (5544) 1048-1052
- 61 Gais S, Plihal W, Wagner U, Born J. Early sleep triggers memory for early visual discrimination skills. Nat Neurosci 2000; 3 (12) 1335-1339
- 62 Fogel SM, Smith CT, Cote KA. Dissociable learning-dependent changes in REM and non-REM sleep in declarative and procedural memory systems. Behav Brain Res 2007; 180 (01) 48-61
- 63 Rasch B, Büchel C, Gais S, Born J. Odor cues during slow-wave sleep prompt declarative memory consolidation. Science 2007; 315 (5817) 1426-1429
- 64 Wilson MA, McNaughton BL. Reactivation of hippocampal ensemble memories during sleep. Science 1994; 265 (5172) 676-679
- 65 Rosanova M, Ulrich D. Pattern-specific associative long-term potentiation induced by a sleep spindle-related spike train. J Neurosci 2005; 25 (41) 9398-9405
- 66 Morin A, Doyon J, Dostie V. et al. Motor sequence learning increases sleep spindles and fast frequencies in post-training sleep. Sleep 2008; 31 (08) 1149-1156
- 67 Skilling QM, Eniwaye B, Clawson BC. et al. Acetylcholine-gated current translates wake neuronal firing rate information into a spike timing-based code in Non-REM sleep, stabilizing neural network dynamics during memory consolidation. PLOS Comput Biol 2021; 17 (09) e1009424
- 68 Bellfy L, Smies CW, Bernhardt AR. et al. The clock gene Per1 may exert diurnal control over hippocampal memory consolidation. Neuropsychopharmacology 2023; 48 (12) 1789-1797
- 69 Iliff JJ, Wang M, Liao Y. et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci Transl Med 2012; 4 (147) 147ra111
- 70 Mestre H, Tithof J, Du T. et al. Flow of cerebrospinal fluid is driven by arterial pulsations and is reduced in hypertension. Nat Commun 2018; 9 (01) 4878
- 71 Xie L, Kang H, Xu Q. et al. Sleep drives metabolite clearance from the adult brain. Science 2013; 342 (6156) 373-377
- 72 Hablitz LM, Vinitsky HS, Sun Q. et al. Increased glymphatic influx is correlated with high EEG delta power and low heart rate in mice under anesthesia. Sci Adv 2019; 5 (02) eaav5447
- 73 Hablitz LM, Plá V, Giannetto M. et al. Circadian control of brain glymphatic and lymphatic fluid flow. Nat Commun 2020; 11 (01) 4411
- 74 Eide PK, Vinje V, Pripp AH, Mardal KA, Ringstad G. Sleep deprivation impairs molecular clearance from the human brain. Brain 2021; 144 (03) 863-874
- 75 Han F, Chen J, Belkin-Rosen A. et al; Alzheimer's Disease Neuroimaging Initiative. Reduced coupling between cerebrospinal fluid flow and global brain activity is linked to Alzheimer disease-related pathology. PLoS Biol 2021; 19 (06) e3001233
- 76 Miao A, Luo T, Hsieh B. et al. Brain clearance is reduced during sleep and anesthesia. Nat Neurosci 2024; 27 (06) 1046-1050
- 77 Henríquez-Beltrán M, Dreyse J, Jorquera J. et al. The U-shaped association between sleep duration, all-cause mortality and cardiovascular risk in a Hispanic/Latino clinically based cohort. J Clin Med 2023; 12 (15) 4961
- 78 You Y, Chen Y, Liu R. et al. Inverted U-shaped relationship between sleep duration and phenotypic age in US adults: a population-based study. Sci Rep 2024; 14 (01) 6247
- 79 Dong L, Xie Y, Zou X. Association between sleep duration and depression in US adults: a cross-sectional study. J Affect Disord 2022; 296: 183-188
- 80 Alhola P, Polo-Kantola P. Sleep deprivation: impact on cognitive performance. Neuropsychiatr Dis Treat 2007; 3 (05) 553-567
- 81 Lucey BP, Hicks TJ, McLeland JS. et al. Effect of sleep on overnight cerebrospinal fluid amyloid β kinetics. Ann Neurol 2018; 83 (01) 197-204
- 82 Arrigoni E, Chamberlin NL, Saper CB, McCarley RW. Adenosine inhibits basal forebrain cholinergic and noncholinergic neurons in vitro. Neuroscience 2006; 140 (02) 403-413
- 83 Maurer SV, Williams CL. The cholinergic system modulates memory and hippocampal plasticity via its interactions with non-neuronal cells. Front Immunol 2017; 8: 1489
- 84 Shokri-Kojori E, Wang GJ, Wiers CE. et al. β-Amyloid accumulation in the human brain after one night of sleep deprivation. Proc Natl Acad Sci U S A 2018; 115 (17) 4483-4488
- 85 Sabia S, Fayosse A, Dumurgier J. et al. Association of sleep duration in middle and old age with incidence of dementia. Nat Commun 2021; 12 (01) 2289
- 86 Ibrahim A, Cesari M, Heidbreder A. et al. Sleep features and long-term incident neurodegeneration: a polysomnographic study. Sleep 2024; 47 (03) 47
- 87 Windred DP, Burns AC, Lane JM. et al. Sleep regularity is a stronger predictor of mortality risk than sleep duration: a prospective cohort study. Sleep 2024; 47 (01) 253
- 88 Wang JL, Lim AS, Chiang WY. et al. Suprachiasmatic neuron numbers and rest-activity circadian rhythms in older humans. Ann Neurol 2015; 78 (02) 317-322
- 89 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 Rhythms 2011; 26 (02) 160-170
- 90 Iranzo A. Sleep in neurodegenerative diseases. Sleep Med Clin 2016; 11 (01) 1-18
- 91 Stefani A, Högl B. Sleep in Parkinson's disease. Neuropsychopharmacology 2020; 45 (01) 121-128
- 92 Galbiati A, Verga L, Giora E, Zucconi M, Ferini-Strambi L. The risk of neurodegeneration in REM sleep behavior disorder: a systematic review and meta-analysis of longitudinal studies. Sleep Med Rev 2019; 43: 37-46
- 93 Postuma RB, Iranzo A, Hu M. et al. Risk and predictors of dementia and parkinsonism in idiopathic REM sleep behaviour disorder: a multicentre study. Brain 2019; 142 (03) 744-759
- 94 Iranzo A, Stefani A, Serradell M. et al; SINBAR (Sleep Innsbruck Barcelona) group. Characterization of patients with longstanding idiopathic REM sleep behavior disorder. Neurology 2017; 89 (03) 242-248
- 95 Yaffe K, Laffan AM, Harrison SL. et al. Sleep-disordered breathing, hypoxia, and risk of mild cognitive impairment and dementia in older women. JAMA 2011; 306 (06) 613-619
- 96 Blackwell T, Yaffe K, Ancoli-Israel S. et al; Osteoporotic Fractures in Men Study Group. Associations between sleep architecture and sleep-disordered breathing and cognition in older community-dwelling men: the Osteoporotic Fractures in Men Sleep Study. J Am Geriatr Soc 2011; 59 (12) 2217-2225
- 97 Blackwell T, Yaffe K, Laffan A. et al; Osteoporotic Fractures in Men (MrOS) Study Group. Associations of objectively and subjectively measured sleep quality with subsequent cognitive decline in older community-dwelling men: the MrOS sleep study. Sleep 2014; 37 (04) 655-663
- 98 Osorio RS, Gumb T, Pirraglia E. et al; Alzheimer's Disease Neuroimaging Initiative. Sleep-disordered breathing advances cognitive decline in the elderly. Neurology 2015; 84 (19) 1964-1971
- 99 Lutsey PL, Misialek JR, Mosley TH. et al. Sleep characteristics and risk of dementia and Alzheimer's disease: The Atherosclerosis Risk in Communities Study. Alzheimers Dement 2018; 14 (02) 157-166
- 100 Leng Y, McEvoy CT, Allen IE, Yaffe K. Association of sleep-disordered breathing with cognitive function and risk of cognitive impairment: a systematic review and meta-analysis. JAMA Neurol 2017; 74 (10) 1237-1245
- 101 Dunietz GL, Chervin RD, Burke JF, Conceicao AS, Braley TJ. Obstructive sleep apnea treatment and dementia risk in older adults. Sleep 2021; 44 (09) 44
- 102 André C, Kuhn E, Rehel S. et al; Medit-Ageing Research Group. Association of sleep-disordered breathing and medial temporal lobe atrophy in cognitively unimpaired amyloid-positive older adults. Neurology 2023; 101 (04) e370-e385
- 103 Leonhard AG, Kapur VK. Beyond traditional hypoxemia metrics: hypoxic burden as a predictor of cognitive dysfunction in sleep apnea. Sleep (Basel) 2025; 48 (03) zsae292
- 104 Breen DP, Williams-Gray CH, Mason SL, Foltynie T, Barker RA. Excessive daytime sleepiness and its risk factors in incident Parkinson's disease. J Neurol Neurosurg Psychiatry 2013; 84 (02) 233-234
- 105 Sohail S, Yu L, Schneider JA, Bennett DA, Buchman AS, Lim ASP. Sleep fragmentation and Parkinson's disease pathology in older adults without Parkinson's disease. Mov Disord 2017; 32 (12) 1729-1737
- 106 Osorio RS, Pirraglia E, Agüera-Ortiz LF. et al. Greater risk of Alzheimer's disease in older adults with insomnia. J Am Geriatr Soc 2011; 59 (03) 559-562
- 107 Pase MP, Himali JJ, Grima NA. et al. Sleep architecture and the risk of incident dementia in the community. Neurology 2017; 89 (12) 1244-1250
- 108 Otaiku AI. Association of sleep abnormalities in older adults with risk of developing Parkinson's disease. Sleep 2022; 45 (11) 45
- 109 Volicer L, Harper DG, Manning BC, Goldstein R, Satlin A. Sundowning and circadian rhythms in Alzheimer's disease. Am J Psychiatry 2001; 158 (05) 704-711
- 110 Del Olmo M, Schmal C, Mizaikoff C. et al. Are circadian amplitudes and periods correlated? A new twist in the story. F1000 Res 2024; 12: 1077
- 111 Breen DP, Vuono R, Nawarathna U. et al. Sleep and circadian rhythm regulation in early Parkinson disease. JAMA Neurol 2014; 71 (05) 589-595
- 112 Hooghiemstra AM, Eggermont LH, Scheltens P, van der Flier WM, Scherder EJ. The rest-activity rhythm and physical activity in early-onset dementia. Alzheimer Dis Assoc Disord 2015; 29 (01) 45-49
- 113 Naismith SL, Hickie IB, Terpening Z. et al. Circadian misalignment and sleep disruption in mild cognitive impairment. J Alzheimers Dis 2014; 38 (04) 857-866
- 114 Chen S-J, Deng Y-T, Li Y-Z. et al. Association of circadian rhythms with brain disorder incidents: a prospective cohort study of 72242 participants. Transl Psychiatry 2022; 12 (01) 514
- 115 Leng Y, Blackwell T, Cawthon PM, Ancoli-Israel S, Stone KL, Yaffe K. Association of circadian abnormalities in older adults with an increased risk of developing Parkinson disease. JAMA Neurol 2020; 77 (10) 1270-1278
- 116 Musiek ES, Bhimasani M, Zangrilli MA, Morris JC, Holtzman DM, Ju YS. Circadian rest-activity pattern changes in aging and preclinical Alzheimer disease. JAMA Neurol 2018; 75 (05) 582-590
- 117 Riemersma-van der Lek RF, Swaab DF, Twisk J, Hol EM, Hoogendijk WJ, Van Someren EJ. Effect of bright light and melatonin on cognitive and noncognitive function in elderly residents of group care facilities: a randomized controlled trial. JAMA 2008; 299 (22) 2642-2655
- 118 Videnovic A, Klerman EB, Wang W, Marconi A, Kuhta T, Zee PC. Timed light therapy for sleep and daytime sleepiness associated with Parkinson disease: a randomized clinical trial. JAMA Neurol 2017; 74 (04) 411-418
- 119 Lee BH, Hille B, Koh DS. Serotonin modulates melatonin synthesis as an autocrine neurotransmitter in the pineal gland. Proc Natl Acad Sci U S A 2021; 118 (43) 118
- 120 Cardoso FDS, Barrett DW, Wade Z, Gomes da Silva S, Gonzalez-Lima F. Photobiomodulation of cytochrome c oxidase by chronic transcranial laser in young and aged brains. Front Neurosci 2022; 16: 818005
- 121 Kress GJ, Liao F, Dimitry J. et al. Regulation of amyloid-β dynamics and pathology by the circadian clock. J Exp Med 2018; 215 (04) 1059-1068
- 122 Ma Z, Jiang W, Zhang EE. Orexin signaling regulates both the hippocampal clock and the circadian oscillation of Alzheimer's disease-risk genes. Sci Rep 2016; 6: 36035
- 123 GBD 2019 Stroke Collaborators. Global, regional, and national burden of stroke and its risk factors, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Neurol 2021; 20 (10) 795-820
- 124 Baillieul S, Dekkers M, Brill AK. et al. Sleep apnoea and ischaemic stroke: current knowledge and future directions. Lancet Neurol 2022; 21 (01) 78-88
- 125 Johnson KG, Johnson DC. Frequency of sleep apnea in stroke and TIA patients: a meta-analysis. J Clin Sleep Med 2010; 6 (02) 131-137
- 126 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) 1447-1451
- 127 Redline S, Yenokyan G, Gottlieb DJ. et al. Obstructive sleep apnea-hypopnea and incident stroke: the sleep heart health study. Am J Respir Crit Care Med 2010; 182 (02) 269-277
- 128 Minoguchi K, Yokoe T, Tazaki T. et al. Silent brain infarction and platelet activation in obstructive sleep apnea. Am J Respir Crit Care Med 2007; 175 (06) 612-617
- 129 Lin HJ, Yeh JH, Hsieh MT, Hsu CY. Continuous positive airway pressure with good adherence can reduce risk of stroke in patients with moderate to severe obstructive sleep apnea: an updated systematic review and meta-analysis. Sleep Med Rev 2020; 54: 101354
- 130 Lee SJ, Kim JS, Song IU, An JY, Kim YI, Lee KS. Poststroke restless legs syndrome and lesion location: anatomical considerations. Mov Disord 2009; 24 (01) 77-84
- 131 Winkelman JW, Shahar E, Sharief I, Gottlieb DJ. Association of restless legs syndrome and cardiovascular disease in the Sleep Heart Health Study. Neurology 2008; 70 (01) 35-42
- 132 Bertisch SM, Pollock BD, Mittleman MA. et al. Insomnia with objective short sleep duration and risk of incident cardiovascular disease and all-cause mortality: Sleep Heart Health Study. Sleep 2018; 41 (06) 41
- 133 Sawadogo W, Adera T, Alattar M, Perera R, Burch JB. Association between insomnia symptoms and trajectory with the risk of stroke in the Health and Retirement Study. Neurology 2023; 101 (05) e475-e488
- 134 Luo Y, Yu G, Liu Y, Zhuge C, Zhu Y. Sleep quality after stroke: a systematic review and meta-analysis. Medicine (Baltimore) 2023; 102 (20) e33777
- 135 Miano S, Fanfulla F, Nobili L. et al. SAS CARE 1: sleep architecture changes in a cohort of patients with ischemic stroke/TIA. Sleep Med 2022; 98: 106-113
- 136 Duss SB, Bauer-Gambelli SA, Bernasconi C. et al. Frequency and evolution of sleep-wake disturbances after ischemic stroke: a 2-year prospective study of 437 patients. Sleep Med 2023; 101: 244-251
- 137 Varadharasu S, Das N. Sleep hygiene efficacy on quality of sleep and mental ability among insomniac patients. J Family Med Prim Care 2024; 13 (10) 4693-4698
- 138 Riemann D, Baglioni C, Bassetti C. et al. European guideline for the diagnosis and treatment of insomnia. J Sleep Res 2017; 26 (06) 675-700
- 139 Siengsukon CF, Nelson E, Williams-Cooke C. et al. Cognitive behavioral therapy for insomnia to enhance cognitive function and reduce the rate of Aβ deposition in older adults with symptoms of insomnia: a single-site randomized pilot clinical trial protocol. Contemp Clin Trials 2020; 99: 106190
- 140 Zhang Z, Xue P, Bendlin BB. et al. Melatonin: a potential nighttime guardian against Alzheimer's. Mol Psychiatry 2025; 30 (01) 237-250
- 141 Sumsuzzman DM, Choi J, Jin Y, Hong Y. Neurocognitive effects of melatonin treatment in healthy adults and individuals with Alzheimer's disease and insomnia: a systematic review and meta-analysis of randomized controlled trials. Neurosci Biobehav Rev 2021; 127: 459-473
- 142 Obayashi K, Saeki K, Iwamoto J. et al. Physiological levels of melatonin relate to cognitive function and depressive symptoms: the HEIJO-KYO cohort. J Clin Endocrinol Metab 2015; 100 (08) 3090-3096
- 143 Mignot E, Mayleben D, Fietze I. et al; investigators. Safety and efficacy of daridorexant in patients with insomnia disorder: results from two multicentre, randomised, double-blind, placebo-controlled, phase 3 trials. Lancet Neurol 2022; 21 (02) 125-139
- 144 Zhou M, Tang S. Effect of a dual orexin receptor antagonist on Alzheimer's disease: sleep disorders and cognition. Front Med (Lausanne) 2023; 9: 984227
- 145 Evans R, Kimura H, Alexander R. et al. Orexin 2 receptor-selective agonist danavorexton improves narcolepsy phenotype in a mouse model and in human patients. Proc Natl Acad Sci U S A 2022; 119 (35) e2207531119
- 146 Suresh V, Bardhan M, Ghosh S. et al. Exploring the role of Orexin-A neuropeptide in Parkinson's disease: a systematic review and meta-analysis. Clin Neurol Neurosurg 2024; 242: 108320
- 147 Cesari M, Stefani A, Penzel T. et al. Interrater sleep stage scoring reliability between manual scoring from two European sleep centers and automatic scoring performed by the artificial intelligence-based Stanford-STAGES algorithm. J Clin Sleep Med 2021; 17 (06) 1237-1247
- 148 Cesari M, Portscher A, Stefani A. et al. Machine learning predicts phenoconversion from polysomnography in isolated REM sleep behavior disorder. Brain Sci 2024; 14 (09) 871
- 149 Zhao H, Cao J, Xie J. et al. Wearable sensors and features for diagnosis of neurodegenerative diseases: a systematic review. Digit Health 2023;9:20552076231173569
- 150 Stephansen JB, Olesen AN, Olsen M. et al. Neural network analysis of sleep stages enables efficient diagnosis of narcolepsy. Nat Commun 2018; 9 (01) 5229
- 151 de Zambotti M, Goldstein C, Cook J. et al. State of the science and recommendations for using wearable technology in sleep and circadian research. Sleep 2024; 47 (04) 47
- 152 Thirunavukarasu AJ, Ting DSJ, Elangovan K, Gutierrez L, Tan TF, Ting DSW. Large language models in medicine. Nat Med 2023; 29 (08) 1930-1940



