Abstract
Sleep disorders, particularly insomnia and obstructive sleep apnea, are increasingly implicated as significant contributors to cognitive decline, dementia, and neurodegenerative diseases such as Alzheimer’s disease (AD) and vascular cognitive impairment and dementia (VCID). However, the extent and specificity of these associations remain uncertain. This meta-analysis evaluates the impact of common sleep disorders on the risk of developing dementia and cognitive decline. A comprehensive search of the literature was conducted to identify prospective cohort studies assessing sleep disorders and dementia risk. Studies reporting risk estimates for dementia, AD, or cognitive decline associated with obstructive sleep apnea, insomnia, and other sleep disorders (e.g., restless legs syndrome, circadian rhythm sleep disorders, excessive daytime sleepiness) were included. Meta-analyses were performed using a random-effects model to calculate pooled hazard ratios (HRs) and 95% confidence intervals (CIs). Thirty-nine cohort studies were included, with subgroup analyses showing significant associations between all-cause dementia and obstructive sleep apnea (HR 1.33, 95% CI 1.09–1.61), insomnia (HR 1.36, 95% CI 1.19–1.55), and other sleep disorders (HR 1.33, 95% CI 1.24–1.43). Obstructive sleep apnea increased the risk for AD (HR 1.45, 95% CI 1.24–1.69), though its association with vascular dementia did not reach statistical significance (HR 1.35, 95% CI 0.99–1.84). Insomnia was significantly associated with increased risk for both vascular dementia (HR 1.59, 95% CI 1.01–2.51) and AD (HR 1.49, 95% CI 1.27–1.74). This meta-analysis highlights the critical role of sleep disorders in dementia risk, emphasizing the need for early detection and management of sleep disturbances. Targeted interventions could play a pivotal role in reducing dementia risk, particularly among high-risk populations.
Keywords: Inadequate sleep, Apnoe, Sleep deficit, Cognitive decline, Aging, Circadian rhythms, Neurodegeneration, Stroke, Semmelweis Study, Sleep-disordered breathing
Introduction
Sleep is a fundamental biological process essential for maintaining brain health, memory consolidation, and overall well-being [1, 2]. Disruptions in sleep, particularly those associated with common sleep disorders, have increasingly been recognized as key contributors to neurodegeneration and cognitive decline [3, 4]. As global populations age, dementia—including Alzheimer’s disease (AD) and vascular cognitive impairment and dementia (VCID)—has emerged as a major public health challenge, underlining the urgency of identifying modifiable risk factors to inform prevention strategies.
A growing body of evidence implicates a range of sleep disorders in the pathogenesis of dementia [5–9]. Sleep disorders are highly prevalent across populations [10] and represent a significant public health concern due to their widespread impact on physical and cognitive health. Insomnia, the most commonly reported sleep disorder, affects approximately 10–30% of the global population, with higher prevalence among older adults and women [10–13]. Sleep-disordered breathing, including obstructive sleep apnea, affects an estimated 17–22% of men and 9–17% of women in the general population, with rates increasing in individuals with obesity and other comorbidities [14–19]. Other sleep disorders include excessive daytime sleepiness, circadian rhythm sleep disorders and sleep-related movement disorders. Excessive daytime sleepiness, which is associated with poor nighttime sleep quality and is reported by up to 20% of adults [20–25]. Circadian rhythm sleep disorders, which involve misalignment between an individual’s internal circadian clock and external environmental cues, are particularly prevalent in shift workers, affecting an estimated 20–30% of this occupational group [26–28]. Among healthcare workers, the prevalence of circadian rhythm sleep disorders is especially concerning due to the demanding and irregular schedules associated with shift work, extended hours, and overnight duties. Studies indicate that up to 40% of healthcare workers experience significant circadian disruption, which not only impairs sleep quality and duration but also impacts cognitive performance, decision-making, and overall well-being. Sleep-related movement disorders, including restless legs syndrome, are reported by 5–15% of adults, with prevalence increasing with age [29, 30]. These disorders not only impact daily functioning and quality of life but are also increasingly recognized as risk factors for neurodegenerative diseases [5–9], cardiovascular conditions [31–33], and other age-related health outcomes, underscoring the need for effective management and prevention strategies.
Despite the wealth of studies investigating the strength of the relationship between sleep disorders and the risk of dementia, Alzheimer’s disease, and cognitive decline, significant inconsistencies in study design, sleep disorder classifications, and diagnostic methodologies have resulted in conflicting conclusions. Some evidence suggests that sleep disorders might serve as early biomarkers of underlying neurodegeneration, reflecting prodromal stages of Alzheimer’s disease or vascular cognitive impairment, while others highlight their potential causal role in accelerating dementia onset [34–38] through mechanisms such as inflammation [39–42], glymphatic dysfunction, and oxidative stress [43]. Besides the many links between insomnia and Alzheimer’s disease, some research suggests common genetic and psychosocial risk factors [44–46].
Building on the framework of prior meta-analyses linking sleep duration to all-cause mortality and stroke risk, this study synthesizes data from prospective cohort studies to evaluate the predictive role of specific sleep disorders—including obstructive sleep apnea, insomnia and other sleep disorders—in dementia risk. Particular emphasis is placed on Alzheimer’s disease and vascular dementia, the most prevalent subtypes of dementia. A meta-analysis offers a powerful approach to addressing these gaps by pooling data from diverse cohorts, increasing statistical power, and identifying consistent patterns across varied populations. This methodology not only clarifies the strength and direction of associations but also allows for the exploration of subgroup effects and potential modifiers. By consolidating findings across a range of sleep disorders, this study aims to resolve key uncertainties and inform public health and clinical strategies for dementia prevention.
Methods
Definitions
In this meta-analysis, sleep disorders examined encompassed obstructive sleep apnea, insomnia, and other sleep disorders, including excessive daytime sleepiness, sleep-related movement disorders, and circadian rhythm sleep disorders. Insomnia is defined as difficulty initiating or maintaining sleep or returning to sleep after premature awakening, while obstructive sleep apnea is characterized by interruptions in breathing during sleep. Circadian rhythm sleep disorders and other non-specific sleep issues were grouped together due to limited specific classification. In the studies analyzed, sleep disorders were identified using self-reports, clinical diagnoses, or objective measurements. Dementia diagnoses were based on recognized international criteria, such as the Diagnostic and Statistical Manual of Mental Disorders, or documented in medical records. Our analysis focused on the predictive role of these sleep disorders in the development of dementia, with Alzheimer’s disease and vascular dementia being the primary subtypes of interest.
Search strategy and selection criteria
To identify relevant research, we performed comprehensive searches across the PubMed, Web of Science, and Cochrane Central Register of Controlled Trials (CENTRAL) databases. These databases were chosen to provide extensive coverage of biomedical and clinical literature. Our search was restricted to publications in the English language and focused on various sleep-related terms in combination with dementia and its subtypes. We also collected studies from previous meta-analyses [5–9].
The search terms included “sleep,” “sleep disorders,” “sleep quality,” “insomnia,” “obstructive sleep apnea,” (or “apnoe”, preferred in British English), “snoring,” “restless legs syndrome,” “circadian rhythm sleep disorders,” “excessive daytime sleepiness,” “dementia,” “Alzheimer’s disease,” and “vascular dementia.” These terms were systematically combined using Boolean operators to identify studies addressing the relationship between sleep disturbances and dementia. For example, we searched for combinations such as “sleep and dementia,” “insomnia and dementia,” “ssleep apnea and dementia,” “circadian rhythm sleep disorder and dementia,” “sleep disturbances and Alzheimer’s disease,” and “sleep disorders and vascular dementia”.
This iterative search strategy ensured the inclusion of studies that explored a broad spectrum of sleep-related issues and their potential associations with dementia, including both Alzheimer’s disease and vascular dementia.
Two researchers independently assessed the articles for eligibility, selecting studies for inclusion in the meta-analysis based on predefined criteria. Eligible studies examined the associations between sleep disorders and dementia, with dementia diagnosed according to international diagnostic standards. Only longitudinal studies were included, provided they evaluated symptoms through self-reports, questionnaires, clinical diagnoses, or objectively measured sleep parameters, and reported effect estimates such as odds ratios (ORs), relative risks (RRs), or hazard ratios (HRs). Studies that were case reports, comments, conference abstracts, cross-sectional designs, lacked recognized diagnostic criteria for dementia, or did not provide relevant effect estimates were excluded. A detailed overview of the selection process is presented in Fig. 1.
Fig. 1.
Flow diagram showing the study selection process
Data extraction
Data were independently extracted from the selected documents by two researchers (MF and AU), who then cross-verified the information. Any discrepancies were resolved through discussion until consensus was reached. The following data were collected from each study: (1) name of the first author, (2) year of publication, (3) type of research, (4) total sample size, and (5) prevalence of dementia cases during follow-up. If adjusted ORs, RRs, or HRs were reported at different levels of adjustment, the most strongly adjusted level was selected.
Statistical analysis
The statistical analyses were conducted using the web-based platform MetaAnalysisOnline.com. To estimate pooled risk metrics, including hazard ratios (HRs) with their corresponding 95% confidence intervals (CIs), we applied a random-effects model. This approach accounts for variations across studies, enhancing the generalizability of the results. Forest plots were generated to visually represent individual study findings alongside the overall pooled estimate, facilitating interpretation of the data and identification of heterogeneity across studies.
To assess inter-study variability, we employed Cochran’s Q test and the I2 statistic. Cochran’s Q test, based on a chi-square framework, evaluated whether the observed variation in effect sizes exceeded what would be expected by chance alone. The I2 metric quantified the proportion of total variation attributable to true heterogeneity rather than random error.
Assessment of publication bias
Potential publication bias was evaluated through visual inspection of funnel plots, which graphically display effect sizes against measures of study precision to detect asymmetrical distributions indicative of bias. For quantitative assessment, Egger’s regression analysis was performed to examine the relationship between effect sizes and their standard errors.
Subgroup analyses
Subgroup analyses were performed separately for apnea, insomnia, and other sleep disorders. For each subgroup, we calculated pooled effect estimates and heterogeneity metrics to evaluate the specific effects on each disorder. The analyses were also performed for the combined cohort to assess aggregated effects across all sleep disorders.
Results
Sleep disorders and all-cause dementia
This analysis included a total of 39 cohorts, of which eight study specifically examined the association between apnoe and the risk of all-cause dementia (see Fig. 2, subgroup = apnoe) [47–53]. The pooled hazard ratio (HR) was 1.33 (95% confidence interval [CI] 1.09–1.61), indicating that individuals with apnoe have a statistically significant 33% higher risk of developing dementia compared to those without it. Substantial heterogeneity was observed among the studies (p < 0.01, I2 = 93%), showing that the effect sizes across the included cohorts were not consistent. Publication bias was evaluated using a funnel plot, which appeared symmetrical, suggesting no evidence of bias (Fig. 3A). This was corroborated by Egger’s test, which produced a non-significant intercept of 0.96 (95% CI − 4.04 to 5.96, p = 0.72).
Fig. 2.
Summary of hazard ratios (HRs) for the association between apnoe, insomnia, or other sleep disorders and all-cause dementia risk across multiple studies. Each row represents an individual study, displaying the study author(s) and year of publication, the log hazard ratio (logHR), standard error (SE), weight assigned to the study in the random-effects model, and HR with 95% confidence intervals (CI). The red squares denote the HR for each study, with square size reflecting study weight. Horizontal lines indicate the 95% CI for each study’s HR. The pooled HR estimate (diamond shape) for all sleep disorders shows a significant association with an increased risk of dementia. Heterogeneity across studies is indicated by I2. Abbreviations: CI, confidence interval; HR, hazard ratio; IV, inverse variance; SE, standard error
Fig. 3.
Funnel plots depicting the relationship between hazard ratios (HRs) and standard error (SE) for the association between various sleep disorders and different cognitive outcomes: all-cause dementia (A–C), vascular dementia (D–F), Alzheimer’s disease (G–I), and cognitive decline (J–L). The plots are organized into these four cognitive outcome categories, with each section examining the relationship with apnea, insomnia, and other sleep disorders. The funnel plot shape and symmetry can provide insights into potential publication bias, with asymmetrical plots suggesting the possibility of selective reporting or publication of studies
Twelve cohorts were included to investigate the association between insomnia and the risk of all-cause dementia [47, 49, 50, 54–61]. The meta-analysis, using a random-effects model, revealed a statistically significant pooled HR of 1.36 (95% CI 1.19–1.55), indicating a 36% increased risk of dementia in those with insomnia. Notable heterogeneity was present across studies (p < 0.01, I2 = 77%), implying variability in effect size was influenced by differences in study design, populations, or definitions of insomnia (Fig. 2, subgroup = insomnia). The funnel plot showed no evidence of asymmetry, indicating a low likelihood of publication bias (intercept: 0.33, 95% CI − 1.58 to 2.25, p = 0.74, as depicted in Fig. 3B).
The third analysis in this cohort included 19 studies focusing on other sleep disorders [47, 49, 50, 55, 62–75]. The pooled HR was 1.33 (95% CI 1.24–1.43), demonstrating a significant association between other sleep disorders and dementia risk (Fig. 2, subgroup = other). Moderate heterogeneity was observed among the studies (I2 = 48%), suggesting that approximately half of the variability was due to differences in study methodologies or populations rather than chance. Publication bias was checked through a funnel plot, which revealed asymmetry, suggesting a potential bias (Fig. 3C). This was supported by Egger’s test, which yielded a significant intercept of 1.41 (95% CI 0.66–2.16, t = 3.681, p = 0.002), indicating a possible underrepresentation of studies with non-significant or negative results.
Inadequate sleep and vascular dementia
Altogether 12 studies were analyzed concerning vascular dementia (Fig. 4). The meta-analysis incorporated three studies examining the relationship between apnoe and vascular dementia (Fig. 4, subgroup = apnoe) [48, 50, 55]. Applying a random-effects model with inverse variance methodology yielded a pooled HR of 1.35 (95% CI 0.99–1.84). These findings suggest no statistically meaningful correlation, as the confidence interval encompassed values below unity. Heterogeneity assessment revealed minimal inter-study variability, with a low I2 value (24%) confirming consistency in both magnitude and directionality of effects. Publication bias evaluation demonstrated symmetrical distribution in the funnel plot, while Egger’s test showed no significant asymmetry (intercept: 1.51, CI 0.6–2.41; p = 0.189) (Fig. 3D).
Fig. 4.
Meta-analysis exploring the relationship between sleep disorders and vascular dementia, with subgroup analyses for apnoe, insomnia, and other sleep disorders. Hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated using a random-effects model. The combined analysis demonstrates a significant link between sleep disorders and vascular dementia (HR = 1.71, 95% CI 1.33–2.20). Substantial heterogeneity is reflected by an I2 value of 93%. Abbreviations: CI, confidence interval; HR, hazard ratio; IV, inverse variance; SE, standard error
Four cohort studies were analyzed to determine the relationship between insomnia and vascular cognitive impairment. Calculations produced a pooled HR of 1.59 (95% CI 1.01–2.51), revealing a significant link between sleep disruption and elevated risk (Fig. 4, subgroup = insomnia) [50, 55, 57, 76]. The analysis yielded statistical significance, though marked heterogeneity emerged (p < 0.01), suggesting considerable methodological or population-based variations across studies. The I2 value of 93% indicated that inter-study differences, rather than chance, accounted for most observed variation. The funnel plot examination (Fig. 3E) showed symmetrical distribution, while Egger’s test results (intercept: − 2.6, 95% CI: − 6.76–1.56; t = − 1.224; p = 0.346) confirmed absence of publication bias.
Five investigations encompassing other sleep disorders were also evaluated (Fig. 4, subgroup = other) [50, 55, 64, 73, 74]. Random-effects modeling produced a pooled HR of 1.97 (95% CI 1.28–3.02), demonstrating a significant correlation. The effect test confirmed statistical significance (p < 0.05). Considerable heterogeneity was observed (p < 0.01), with an I2 value of 85%, suggesting that methodological differences primarily explained the effect estimate variability. Publication bias assessment revealed funnel plot asymmetry, confirmed by Egger’s test (intercept: 3.94, 95% CI 2.7–5.19; t = 6.196; p = 0.008), suggesting potential reporting bias among included studies (Fig. 3F).
Insufficient sleep and Alzheimer’s disease
A total of 32 trials were examined related to Alzheimer’s disease (see Fig. 5). Seven studies yielded evidence for a significant correlation between apnoe and Alzheimer’s pathology (Fig. 5, subgroup = apnoe) [48, 50, 51, 53, 77–79]. The analysis produced a combined HR of 1.45 (95% CI 1.24–1.69; p < 0.05). Notable heterogeneity emerged (p = 0.03), with an I2 value of 57% indicating that methodological variations, rather than sampling error, accounted for the majority of inter-study differences. The funnel plot analysis revealed asymmetrical distribution (Fig. 3G), suggesting potential underrepresentation of smaller studies with non-significant results. Egger’s test confirmed this observation (intercept: 1.42, 95% CI 0.59–2.25; t = 3.348; p = 0.02).
Fig. 5.
Forest plot depicting hazard ratios (HRs) for the association between sleep disorders and Alzheimer’s disease risk. The analysis includes 32 studies stratified into three subgroups: apnoe (n = 7), insomnia (n = 8), and other sleep disorders (n = 17). The overall pooled effect shows a significant increased risk (HR = 1.45, 95% CI 1.28–1.63). Diamond markers represent pooled estimates; squares represent individual study effects with size proportional to study weight. Abbreviations: CI, confidence interval; HR, hazard ratio; IV, inverse variance; SE, standard error
Analysis of eight studies demonstrated a markedly elevated Alzheimer’s risk among individuals with insomnia (HR 1.49; 95% CI 1.27–1.74; p < 0.05). Substantial heterogeneity was detected (p < 0.01), with 74% of inter-study variability attributable to methodological differences rather than chance (Fig. 5, subgroup = insomnia) [50, 57, 58, 76, 80–82]. Publication bias assessment (Fig. 3H) revealed symmetrical distribution, supported by Egger’s test results (intercept: − 1.24, CI − 2.53–0.06; t = − 1.866; p = 0.111), suggesting negligible reporting bias.
Examination of 17 studies revealed that subjects with various sleep disturbances exhibited 42% higher Alzheimer’s risk compared to controls (Fig. 5, subgroup = other) [50, 64, 65, 68–74, 77, 83–89]. The meta-analysis generated an HR of 1.42 (95% CI 1.11–1.83; p < 0.05). Marked heterogeneity was observed (p < 0.01), with an I2 value of 99% indicating that study characteristics explained nearly all effect variations. Publication bias evaluation (Fig. 3I) demonstrated asymmetrical distribution, confirmed by Egger’s test (intercept: − 8.16, 95% CI − 11.71 to − 4.61; t = − 4.506; p < 0.01), suggesting potential underreporting of null or minimal effect findings.
Sleep disturbance and cognitive decline
Altogether 29 cohorts were analyzed about cognitive functions (Fig. 6). Of these, 13 cohort studies examined the relationship between apnoe and cognitive decline. The meta-analysis, conducted using a random-effects model with the inverse variance method, identified a statistically significant association, with a pooled HR of 1.44 (95% confidence interval [CI] 1.24–1.68), indicating that sleep-disordered breathing is linked to an elevated risk of cognitive decline (Fig. 6, subgroup = apnoe) [47, 48, 50, 51, 55, 90–97]. Substantial heterogeneity was detected among the included studies (p < 0.01; I2 = 66%). A funnel plot evaluation, supported by Egger’s test (intercept: 0.87, 95% CI: − 0.74 to 2.49; t = 1.059; p = 0.312) showed no signs of asymmetry, suggesting a low likelihood of publication bias (Fig. 3J).
Fig. 6.
Meta-analysis results showing associations between sleep disorders and risk of cognitive decline across three categories: apnoe (n = 13), insomnia (n = 8), and other sleep disturbances (n = 9). The combined analysis of 30 studies demonstrates an elevated risk (HR = 1.51, 95% CI 1.37–1.67), with moderate heterogeneity across studies. Individual study estimates are shown as squares, with pooled estimates represented by diamonds. Abbreviations: CI, confidence interval; HR, hazard ratio; IV, inverse variance; SE, standard error
Eight studies were included in the meta-analysis to evaluate the link between insomnia and cognitive decline (Fig. 6, subgroup = insomnia) [55, 63, 98–103]. The pooled HR was 1.63 (95% CI 1.36–1.95), indicating a statistically significant increase in risk. Unlike the previous subgroup, this analysis exhibited low heterogeneity (I2 = 29%; p = 0.19), suggesting consistency in effect sizes across the studies. However, the funnel plot revealed asymmetry, raising concerns about potential publication bias (Fig. 3K). Egger’s test further confirmed this, demonstrating significant small-study effects (intercept: 2.18, 95% CI 1.05–3.32; t = 3.772; p = 0.009).
Finally, eight studies assessing the association between other sleep disorders and cognitive decline were analyzed (Fig. 6, subgroup = other) [55, 66, 71, 104–107]. The meta-analysis, using a random-effects model, identified a significant association, with a pooled HR of 1.55 (95% CI 1.27–1.90). Notable heterogeneity was observed (p = 0.01; I2 = 64%), indicating that the effect sizes across cohorts were not uniform in both magnitude. Funnel plot analysis suggested potential publication bias, which was further corroborated by Egger’s test (intercept: 3.01, 95% CI 1.70–4.33; t = 4.495; p = 0.004; Fig. 3L).
Discussion
The findings of this meta-analysis provide robust evidence for an association between a range of sleep disorders—including obstructive apnea, insomnia, and other sleep disorders —and an elevated risk of dementia. These results build upon the growing recognition that sleep disturbances are not only symptoms of neurodegeneration but may also contribute causally to its onset and progression [5–9, 47–107]. This relationship underscores the critical role of sleep health in maintaining cognitive function and preventing age-related brain decline, with significant implications for both clinical practice and public health strategies.
The biological mechanisms underpinning the observed associations between sleep disorders and dementia are complex and multifactorial. Insomnia, characterized by difficulty initiating or maintaining sleep, is associated with chronic activation of the hypothalamic–pituitary–adrenal (HPA) axis [108], leading to elevated cortisol levels, systemic inflammation [39–42], and chronic oxidative stress [43], all of which exacerbate neurodegeneration [41–43, 108]. Sleep deprivation further impairs glymphatic clearance of neurotoxic proteins [109], such as amyloid-β and tau, both of which are central to Alzheimer’s disease pathology [1, 34–36, 109–111]. Obstructive sleep apnea is marked by intermittent hypoxia, hypercapnia, and sleep fragmentation. Other sleep disturbances, such as excessive daytime sleepiness and circadian rhythm sleep disorders, highlight the importance of circadian rhythm integrity for brain health. Disruption of circadian rhythms, commonly seen in shift workers, impairs restorative processes, deregulates inflammatory pathways, and disrupts the timing of critical metabolic and cognitive functions. These disturbances can lead to endothelial dysfunction [33, 37, 38, 112–114], increased blood–brain barrier permeability [115–118], impaired neurovascular coupling [119] and functional connectivity [120], and amyloid-β deposition [34, 35]—mechanisms that contribute to both Alzheimer’s disease [121] and VCID. Sleep disturbances have also been linked to systemic inflammation [39, 117, 122, 123] and metabolic dysregulation [124–126], both of which increase cerebrovascular and neuronal vulnerability. The impact of sleep disturbances in shift workers may be compounded by occupational stress, leading to chronic sleep restriction and cognitive impairment over time [127]. Collectively, these mechanistic insights demonstrate that sleep disturbances likely act through multiple, overlapping pathways to accelerate neurodegeneration and contribute to the development of dementia.
Sleep disorders may also impact the cellular mechanisms of aging [128], contributing to accelerated biological aging [129–132] and thereby increasing the risk of a range of age-related diseases, including Alzheimer’s disease and VCID. Accordingly, chronic sleep disturbances are associated with epigenetic alterations [130–133], increased oxidative stress [43, 134], systemic inflammation [39, 122], and impaired mitochondrial function [135, 136], all of which are hallmarks of aging. Supporting this concept, sleep disorders have been linked to several age-related diseases and conditions, including increased risks of stroke [137, 138], cardiovascular disease [31], and mortality [139, 140]. Furthermore, emerging evidence suggests a potential link between sleep disruption and cancer development [141–143], with mechanisms involving impaired DNA repair and enhanced tumor growth driven by chronic inflammation and hormonal imbalances. These findings underscore the broader impact of sleep disorders as accelerators of aging and highlight their role in the pathogenesis of multiple age-related diseases.
The differential effects of sleep disorders on dementia subtypes warrant further investigation. For example, the strong association of obstructive sleep apnea with vascular dementia may stem from intermittent hypoxia and its impact on cerebrovascular health, whereas insomnia’s relationship with Alzheimer’s disease is more closely tied to inflammation and impaired glymphatic clearance. Disentangling these subtype-specific effects is critical for guiding tailored prevention strategies.
The findings of this meta-analysis have significant implications for ongoing population-based research, including the Semmelweis Study, a prospective workplace cohort conducted at one of Central Europe’s largest health sciences universities [144]. This study focuses on identifying determinants of unhealthy aging, with particular emphasis on modifiable lifestyle factors [145–150], including sleep. Given its unique occupational setting [151], the Semmelweis Study provides an unparalleled opportunity to investigate the role of sleep disorders in cognitive decline and dementia risk, particularly among healthcare professionals—a group disproportionately affected by sleep disturbances due to shift work, workplace stress, and digital device overuse [152–155]. Healthcare professionals, such as doctors and nurses, are often exposed to circadian misalignment caused by rotating shifts and irregular work schedules [152–155]. These disruptions are well-documented contributors to insomnia, excessive daytime sleepiness, and poor sleep quality, which, as shown in this meta-analysis, significantly elevate dementia risk. By incorporating comprehensive assessments of sleep duration, quality, and disorders, the Semmelweis Study is uniquely positioned to provide longitudinal insights into the cumulative effects of sleep disturbances on cognitive health in a high-risk population.
Emerging evidence suggests that sleep disturbances may also serve as early biomarkers of neurodegeneration [156]. Conditions such as insomnia and disrupted circadian rhythms are often observed in the prodromal stages of Alzheimer’s disease, potentially reflecting underlying amyloid-β or tau pathology [157]. Identifying whether specific sleep disorders consistently precede cognitive decline could establish them as preclinical indicators of dementia, enabling earlier diagnosis and intervention [157]. The Semmelweis Study is well-positioned to explore these dynamics longitudinally, shedding light on the temporal relationships between sleep disturbances and neurodegeneration.
Sleep disorders frequently coexist with other chronic conditions, such as cardiovascular disease, diabetes, and depression, which independently increase dementia risk. Understanding how these comorbidities interact with sleep disturbances to accelerate cognitive decline is a critical area for future research. Furthermore, integrating genetic and epigenetic analyses—such as the role of circadian regulation genes (e.g., CLOCK and BMAL1) or APOE variations—could elucidate individual variability in sleep-dementia pathways and support the development of personalized interventions.
Moreover, the Semmelweis Study can examine how occupational stress and lifestyle factors, such as physical inactivity or prolonged screen exposure, interact with sleep disturbances to amplify dementia risk. For instance, by evaluating circadian rhythm disruption in shift workers, the study can elucidate the extent to which misalignment between biological rhythms and occupational demands contribute to cognitive decline over time. These findings could guide the design of workplace interventions to improve sleep health and mitigate long-term dementia risk in vulnerable occupational cohorts.
The Semmelweis-EUniWell Workplace Health Promotion Program, which targets the same population, is well-positioned to translate these findings into actionable interventions. Programs that focus on improving sleep hygiene—such as reducing exposure to blue light from screens, implementing stress management strategies, and promoting regular sleep schedules—could be tailored to address the specific needs of shift workers and high-stress professionals. By incorporating workplace-based interventions, such as rotating shift schedules aligned with natural circadian rhythms and protected rest periods, the Semmelweis-EUniWell Workplace Health Promotion Program could provide effective tools to reduce the adverse effects of occupational sleep disturbances on brain health.
This meta-analysis has several strengths, including its comprehensive synthesis of data from diverse cohorts, robust methodology, and focus on a range of sleep disorders. By differentiating between various sleep disturbances, this study provides a nuanced understanding of the unique contributions of insomnia, obstructive sleep apnea, and other disorders to dementia risk. However, several limitations must be acknowledged. A notable limitation of this meta-analysis is the variability in study populations, diagnostic criteria, and sleep disorder assessment methods. Many of the included studies relied on self-reported sleep measures, which are subject to recall bias and misclassification. Participants may underreport or overestimate their sleep problems due to individual differences in perception or recall limitations. Self-reported measures may also fail to capture the full extent of sleep disturbances. Objective assessments, such as actigraphy or polysomnography, would provide more accurate evaluations of sleep patterns and disturbances. While self-report remains a widely used method in epidemiological studies due to its practicality, future investigations should prioritize objective sleep assessments to strengthen the reliability of findings and minimize potential bias. Additionally, significant heterogeneity was observed across studies, reflecting variations in study design, sleep disorder definitions, and follow-up duration.
Our analysis of publication bias using funnel plots revealed asymmetry, particularly in studies examining the relationship between other sleep disorders and dementia risk. This suggests a potential underrepresentation of studies with non-significant findings, which may skew the pooled effect estimates. Egger’s test confirmed a small-study effect in certain subgroup analyses, indicating that studies with smaller sample sizes and non-significant results might be less likely to be published. While we attempted to mitigate this by including all available cohort studies, future research should focus on addressing publication bias through pre-registered studies and the inclusion of null findings to provide a more balanced perspective on the association between sleep disturbances and dementia.
Building on the insights from this meta-analysis, future research should explore the causal pathways linking specific sleep disturbances to dementia using advanced imaging and biomarker-based approaches. Longitudinal studies are needed to assess the cumulative impact of sleep disorders on cognitive decline while accounting for occupational stress and lifestyle factors.
The question of whether treating sleep disorders can mitigate dementia risk remains an important avenue for future research. Evidence suggests that continuous positive airway pressure (CPAP) therapy for obstructive sleep apnea can improve cognitive performance, particularly in executive function and memory [158–164]. However, long-term data on whether CPAP prevents or delays dementia onset remain limited. Similarly, cognitive behavioral therapy for insomnia (CBT-I) has been shown to enhance sleep quality and reduce systemic inflammation, both of which may help preserve cognitive function. Future studies should examine the extent to which early interventions, such as CPAP, oral appliances, or pharmacological approaches, reduce dementia risk in high-risk populations. Given the increasing recognition of sleep disturbances as modifiable risk factors, integrating sleep health into dementia prevention strategies could have significant public health implications.
In conclusion, this meta-analysis highlights a significant association between sleep disorders and dementia risk, emphasizing the critical role of sleep health in preventing neurodegeneration. The findings underscore the importance of early identification and management of sleep disturbances, particularly in high-risk populations such as healthcare professionals. The Semmelweis Study, with its focus on occupational cohorts and sleep health, is uniquely positioned to address these knowledge gaps, offering valuable insights into the interplay between sleep disorders, occupational stress, and cognitive decline. By informing targeted interventions [165] and public health strategies, this work provides a foundation for reducing dementia risk and promoting healthy brain aging in vulnerable populations.
Acknowledgements
The support of ELIXIR Hungary is acknowledged.
Author contribution
ZU, MF, PV, AU, and BG contributed to the study conception and design. MF, AL, PV, and AU performed the systematic literature search and data extraction. Statistical analyses were conducted by GM, JTF, AU, and BG. ZU, MF, BG, AL, and AU drafted the manuscript. GP, GM, JTF, VZ, and PV provided critical revisions for intellectual content. BG, AL, GM, and JTF prepared the figures. All authors reviewed, edited, and approved the final manuscript.
Funding
Open access funding provided by Semmelweis University. This work was supported by grants from the National Institute on Aging (RF1 AG072295, R01 AG055395, R01 AG068295, R01 AG070915); the National Institute of Neurological Disorders and Stroke (R01 NS100782); and the National Cancer Institute (R01 CA255840). This work was also supported by TKP2021-NKTA- 47, implemented with the support provided by the Ministry of Innovation and Technology of Hungary from the National Research, Development and Innovation Fund, financed under the TKP2021-NKTA funding scheme; by funding through the National Cardiovascular Laboratory Program (RRF- 2.3.1–21 - 2022–00003) and by the National Laboratory for Drug Research and Development (PharmaLab, RRF- 2.3.1–21 - 2022–00015) provided by the Ministry of Innovation and Technology of Hungary from the National Research, Development and Innovation Fund; by the Semmelweis Momentum Programme; Project no. 135784 implemented with the support provided from the National Research, Development and Innovation Fund of Hungary, financed under the K20 funding scheme and the European University for Well-Being (EUniWell) program (grant agreement number 101004093/EUniWell/EAC-A02 - 2019/EAC-A02 - 2019–1). The computational infrastructure of A5 Genetics Ltd (Kutaso, Hungary) was used for the study. This work was also supported by the EKÖP- 2024–2 and EKÖP- 2024–9 New National Excellence Program of the Ministry for Culture and Innovation from the source of the National Research, Development, and Innovation Fund.
Declarations
Ethics approval and consent to participate
NA.
Consent for publication
NA.
Competing interests
Dr. Balázs Győrffy serves as Associate Editor for GeroScience. Dr. Zoltan Ungvari serves as Editor-in-Chief for GeroScience and has personal relationships with individuals involved in the submission of this paper.
Disclaimer
The funding sources had no role in the study design; in the collection, analysis and interpretation of data; in the writing of the report; and in the decision to submit the article for publication. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. The 4o version of ChatGPT, developed by OpenAI, and Claude 3.5 Sonnet, developed by Anthropic were used as a language tool to refine our writing and enhance the clarity of our work.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Zoltan Ungvari, Mónika Fekete, and Andrea Lehoczki contributed equally to this work.
References
- 1.Kroeger D, Vetrivelan R. To sleep or not to sleep - Effects on memory in normal aging and disease. Aging Brain. 2023;3:100068. 10.1016/j.nbas.2023.100068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Lewis LD. The interconnected causes and consequences of sleep in the brain. Science. 2021;374:564–8. 10.1126/science.abi8375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Stephens ER, Sarangi A, Gude J. Short sleep duration and dementia: a narrative review. Proc (Bayl Univ Med Cent). 2022;35:328–31. 10.1080/08998280.2022.2026123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Chen J, Peng G, Sun B. Alzheimer’s disease and sleep disorders: a bidirectional relationship. Neuroscience. 2024;557:12–23. 10.1016/j.neuroscience.2024.08.008. [DOI] [PubMed] [Google Scholar]
- 5.Shi L, Chen S-J, Ma M-Y, Bao Y-P, Han Y, Wang Y-M, Shi J, Vitiello MV, Lu L. Sleep disturbances increase the risk of dementia: a systematic review and meta-analysis. Sleep Med Rev. 2018;40:4–16. [DOI] [PubMed] [Google Scholar]
- 6.Guay-Gagnon M, Vat S, Forget MF, Tremblay-Gravel M, Ducharme S, Nguyen QD, Desmarais P. Sleep apnea and the risk of dementia: a systematic review and meta-analysis. J Sleep Res. 2022;31:e13589. [DOI] [PubMed] [Google Scholar]
- 7.Bubu OM, Brannick M, Mortimer J, Umasabor-Bubu O, Sebastião YV, Wen Y, Schwartz S, Borenstein AR, Wu Y, Morgan D, Anderson WM. Sleep, cognitive impairment, and Alzheimer’s disease: a systematic review and meta-analysis. Sleep. 2017;40(1):zsw032. 10.1093/sleep/zsw032. [DOI] [PubMed] [Google Scholar]
- 8.Wang S, Zheng X, Huang J, Liu J, Li C, Shang H. Sleep characteristics and risk of Alzheimer’s disease: a systematic review and meta-analysis of longitudinal studies. J Neurol. 2024;271:3782–93. 10.1007/s00415-024-12380-7. [DOI] [PubMed] [Google Scholar]
- 9.Tian Q, Sun J, Li X, Liu J, Zhou H, Deng J, Li J. Association between sleep apnoea and risk of cognitive impairment and Alzheimer’s disease: a meta-analysis of cohort-based studies. Sleep Breath. 2024;28:585–95. 10.1007/s11325-023-02934-w. [DOI] [PubMed] [Google Scholar]
- 10.Kocevska D, Lysen TS, Dotinga A, Koopman-Verhoeff ME, Luijk M, Antypa N, Biermasz NR, Blokstra A, Brug J, Burk WJ, Comijs HC, Corpeleijn E, Dashti HS, de Bruin EJ, de Graaf R, Derks IPM, Dewald-Kaufmann JF, Elders PJM, Gemke R, Grievink L, Hale L, Hartman CA, Heijnen CJ, Huisman M, Huss A, Ikram MA, Jones SE, Velderman MK, Koning M, Meijer AM, et al. Sleep characteristics across the lifespan in 1.1 million people from the Netherlands, United Kingdom and United States: a systematic review and meta-analysis. Nat Hum Behav. 2021;5:113–22. 10.1038/s41562-020-00965-x. [DOI] [PubMed] [Google Scholar]
- 11.Hilmisson H, Sveinsdottir E, Lange N, Magnusdottir S. Insomnia symptoms in primary care: a prospective study focusing on prevalence of undiagnosed co-morbid sleep disordered breathing. Eur J Intern Med. 2019;63:19–26. 10.1016/j.ejim.2019.01.011. [DOI] [PubMed] [Google Scholar]
- 12.Morin CM, Jarrin DC. Epidemiology of insomnia: prevalence, course, risk factors, and public health burden. Sleep Med Clin. 2022;17:173–91. 10.1016/j.jsmc.2022.03.003. [DOI] [PubMed] [Google Scholar]
- 13.Appleton SL, Reynolds AC, Gill TK, Melaku YA, Adams RJ. Insomnia prevalence varies with symptom criteria used with implications for epidemiological studies: role of anthropometrics, sleep habit, and comorbidities. Nat Sci Sleep. 2022;14:775–90. 10.2147/NSS.S359437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Fung CH, Martin JL, Dzierzewski JM, Jouldjian S, Josephson K, Park M, Alessi C. Prevalence and symptoms of occult sleep disordered breathing among older veterans with insomnia. J Clin Sleep Med. 2013;9:1173–8. 10.5664/jcsm.3162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Heinzer R, Vat S, Marques-Vidal P, Marti-Soler H, Andries D, Tobback N, Mooser V, Preisig M, Malhotra A, Waeber G, Vollenweider P, Tafti M, Haba-Rubio J. Prevalence of sleep-disordered breathing in the general population: the HypnoLaus Study. Lancet Respir Med. 2015;3:310–8. 10.1016/S2213-2600(15)00043-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kent BD, Grote L, Ryan S, Pepin JL, Bonsignore MR, Tkacova R, Saaresranta T, Verbraecken J, Levy P, Hedner J, McNicholas WT. Diabetes mellitus prevalence and control in sleep-disordered breathing: the European Sleep Apnea Cohort (ESADA) study. Chest. 2014;146:982–90. 10.1378/chest.13-2403. [DOI] [PubMed] [Google Scholar]
- 17.Peppard PE, Young T, Barnet JH, Palta M, Hagen EW, Hla KM. Increased prevalence of sleep-disordered breathing in adults. Am J Epidemiol. 2013;177:1006–14. 10.1093/aje/kws342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Dominguez-Mayoral A, Sanchez-Gomez J, Guerrero P, Ferrer M, Gutierrez C, Aguilar M, Fouz-Roson N, Benitez JM, Perez-Sanchez S, Gamero-Garcia MA, De Torres-Chacon R, Barragan-Prieto A, Algaba P, Ruiz-Bayo L, Montaner J. High prevalence of obstructive sleep apnea syndrome in Spain’s Stroke Belt. J Int Med Res. 2021;49:3000605211053090. 10.1177/03000605211053090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kinoshita R, Quint JK, Kallis C, Polkey MI. Estimated prevalence of obstructive sleep apnea by occupation and industry in England: a descriptive study. Sleep Adv. 2024;5:zpae069. 10.1093/sleepadvances/zpae069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Chen L, Luo C, Liu S, Chen W, Liu Y, Li Y, Du Y, Zou H, Pan J. Excessive daytime sleepiness in general hospital nurses: prevalence, correlates, and its association with adverse events. Sleep Breath. 2019;23:209–16. 10.1007/s11325-018-1684-9. [DOI] [PubMed] [Google Scholar]
- 21.Hayley AC, Williams LJ, Kennedy GA, Berk M, Brennan SL, Pasco JA. Prevalence of excessive daytime sleepiness in a sample of the Australian adult population. Sleep Med. 2014;15:348–54. 10.1016/j.sleep.2013.11.783. [DOI] [PubMed] [Google Scholar]
- 22.Karunanayake C, Dosman J, Fenton M, Rennie D, Kirychuk S, Ramsden V, Seeseequasis J, Abonyi S, Pahwa P, First Nations Lung Health Project Research T. Association between co-morbidities and the prevalence of excessive daytime sleepiness over a four-year period. Clocks Sleep. 2019;1:459–70. 10.3390/clockssleep1040035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ozder A, Eker HH. The prevalence of excessive daytime sleepiness among academic physicians and its impact on the quality of life and occupational performance. Int J Occup Med Environ Health. 2015;28:721–30. 10.13075/ijomeh.1896.00367. [DOI] [PubMed] [Google Scholar]
- 24.Ren J, Liu R, Zhao T, Lu J, Liu C, Hou T, Wang Y, Cong L, Du Y, Tang S, Qiu C. Prevalence and associated factors of excessive daytime sleepiness in rural older adults: a population-based study. Sleep Breath. 2024;28:1459–64. 10.1007/s11325-024-03004-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Seneviratne U, Puvanendran K. Excessive daytime sleepiness in obstructive sleep apnea: prevalence, severity, and predictors. Sleep Med. 2004;5:339–43. 10.1016/j.sleep.2004.01.021. [DOI] [PubMed] [Google Scholar]
- 26.Kim JH, Elkhadem AR, Duffy JF. Circadian rhythm sleep-wake disorders in older adults. Sleep Med Clin. 2022;17:241–52. 10.1016/j.jsmc.2022.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Sack RL, Auckley D, Auger RR, Carskadon MA, Wright KP Jr, Vitiello MV, Zhdanova IV, American Academy of Sleep M. Circadian rhythm sleep disorders: part II, advanced sleep phase disorder, delayed sleep phase disorder, free-running disorder, and irregular sleep-wake rhythm. An American Academy of Sleep Medicine review. Sleep. 2007;30:1484–501. 10.1093/sleep/30.11.1484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Sack RL, Auckley D, Auger RR, Carskadon MA, Wright KP Jr, Vitiello MV, Zhdanova IV, American Academy of Sleep M. Circadian rhythm sleep disorders: part I, basic principles, shift work and jet lag disorders. An American Academy of Sleep Medicine review. Sleep. 2007;30:1460–83. 10.1093/sleep/30.11.1460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Iranzo A. Parasomnias and sleep-related movement disorders in older adults. Sleep Med Clin. 2018;13:51–61. 10.1016/j.jsmc.2017.09.005. [DOI] [PubMed] [Google Scholar]
- 30.Trotti LM. Restless legs syndrome and sleep-related movement disorders. Continuum (Minneap Minn). 2017;23:1005–16. 10.1212/CON.0000000000000488. [DOI] [PubMed] [Google Scholar]
- 31.Belloir J, Makarem N, Shechter A. Sleep and circadian disturbance in cardiovascular risk. Curr Cardiol Rep. 2022;24:2097–107. 10.1007/s11886-022-01816-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Cappuccio FP, Cooper D, D’Elia L, Strazzullo P, Miller MA. Sleep duration predicts cardiovascular outcomes: a systematic review and meta-analysis of prospective studies. Eur Heart J. 2011;32:1484–92. 10.1093/eurheartj/ehr007. [DOI] [PubMed] [Google Scholar]
- 33.Peracaula M, Torres D, Poyatos P, Luque N, Rojas E, Obrador A, Orriols R, Tura-Ceide O. Endothelial dysfunction and cardiovascular risk in obstructive sleep apnea: a review article. Life (Basel). 2022;12(4):537. 10.3390/life12040537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Sun H, Gao Y, Li M, Zhang S, Shen T, Yuan X, Shang X, Li Z, Zhang J. Altered amyloid-beta and tau proteins in neural-derived plasma exosomes in obstructive sleep apnea. Sleep Med. 2022;94:76–83. 10.1016/j.sleep.2022.03.021. [DOI] [PubMed] [Google Scholar]
- 35.Elias A, Cummins T, Tyrrell R, Lamb F, Dore V, Williams R, Rosenfeld JV, Hopwood M, Villemagne VL, Rowe CC. Risk of Alzheimer’s disease in obstructive sleep apnea syndrome: amyloid-beta and Tau imaging. J Alzheimers Dis. 2018;66:733–41. 10.3233/JAD-180640. [DOI] [PubMed] [Google Scholar]
- 36.Ju YS, Zangrilli MA, Finn MB, Fagan AM, Holtzman DM. Obstructive sleep apnea treatment, slow wave activity, and amyloid-beta. Ann Neurol. 2019;85:291–5. 10.1002/ana.25408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Calvin AD, Covassin N, Kremers WK, Adachi T, Macedo P, Albuquerque FN, Bukartyk J, Davison DE, Levine JA, Singh P, Wang S, Somers VK. Experimental sleep restriction causes endothelial dysfunction in healthy humans. J Am Heart Assoc. 2014;3:e001143. 10.1161/JAHA.114.001143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Carreras A, Zhang SX, Peris E, Qiao Z, Gileles-Hillel A, Li RC, Wang Y, Gozal D. Chronic sleep fragmentation induces endothelial dysfunction and structural vascular changes in mice. Sleep. 2014;37:1817–24. 10.5665/sleep.4178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Piber D. The role of sleep disturbance and inflammation for spatial memory. Brain Behav Immun Health. 2021;17:100333. 10.1016/j.bbih.2021.100333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Li Y, Zhang B, Zhou Y, Wang D, Liu X, Li L, Wang T, Zhang Y, Jiang M, Tang H, Amsel LV, Fan F, Hoven CW. Gut microbiota changes and their relationship with inflammation in patients with acute and chronic insomnia. Nat Sci Sleep. 2020;12:895–905. 10.2147/NSS.S271927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Walker JL, Slavish DC, Dolan M, Dietch JR, Wardle-Pinkston S, Messman B, Ruggero CJ, Kohut M, Borwick J, Kelly K, Taylor DJ. Age-dependent associations among insomnia, depression, and inflammation in nurses. Psychol Health. 2021;36:967–84. 10.1080/08870446.2020.1805450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zagaria A, Ballesio A. Insomnia symptoms as long-term predictors of anxiety symptoms in middle-aged and older adults from the English Longitudinal Study of Ageing (ELSA), and the role of systemic inflammation. Sleep Med. 2024;124:120–6. 10.1016/j.sleep.2024.09.020. [DOI] [PubMed] [Google Scholar]
- 43.Hachul de Campos H, Brandao LC, D’Almeida V, Grego BH, Bittencourt LR, Tufik S, Baracat EC. Sleep disturbances, oxidative stress and cardiovascular risk parameters in postmenopausal women complaining of insomnia. Climacteric. 2006;9:312–9. 10.1080/13697130600871947. [DOI] [PubMed] [Google Scholar]
- 44.Chen D, Wang X, Huang T, Jia J. Sleep and late-onset Alzheimer’s disease: shared genetic risk factors, drug targets, molecular mechanisms, and causal effects. Front Genet. 2022;13:794202. 10.3389/fgene.2022.794202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Larsson SC, Wolk A. The role of lifestyle factors and sleep duration for late-onset dementia: a cohort study. J Alzheimers Dis. 2018;66:579–86. 10.3233/JAD-180529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Burke SL, Cadet T, Alcide A, O’Driscoll J, Maramaldi P. Psychosocial risk factors and Alzheimer’s disease: the associative effect of depression, sleep disturbance, and anxiety. Aging Ment Health. 2018;22:1577–84. 10.1080/13607863.2017.1387760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Yaffe K, Laffan AM, Harrison SL, Redline S, Spira AP, Ensrud KE, Ancoli-Israel S, Stone KL. Sleep-disordered breathing, hypoxia, and risk of mild cognitive impairment and dementia in older women. JAMA. 2011;306:613–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Chang W-P, Liu M-E, Chang W-C, Yang AC, Ku Y-C, Pai J-T, Huang H-L, Tsai S-J. Sleep apnea and the risk of dementia: a population-based 5-year follow-up study in Taiwan. PLoS One. 2013;8:e78655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Tsapanou A, Gu Y, Manly J, Schupf N, Tang M-X, Zimmerman M, Scarmeas N, Stern Y. Daytime sleepiness and sleep inadequacy as risk factors for dementia. Dement Geriatr Cogn Disord Extra. 2015;5:286–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Yaffe K, Nettiksimmons J, Yesavage J, Byers A. Sleep quality and risk of dementia among older male veterans. Am J Geriatr Psychiatry. 2015;23:651–4. [DOI] [PubMed] [Google Scholar]
- 51.Lutsey PL, Misialek JR, Mosley TH, Gottesman RF, Punjabi NM, Shahar E, MacLehose R, Ogilvie RP, Knopman D, Alonso A. Sleep characteristics and risk of dementia and Alzheimer’s disease: the atherosclerosis risk in communities study. Alzheimer’s Dement. 2018;14:157–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Yeh N-C, Tien K-J, Yang C-M, Wang J-J, Weng S-F. Increased risk of Parkinson’s disease in patients with obstructive sleep apnea: a population-based, propensity score-matched, longitudinal follow-up study. Medicine. 2016;95:e2293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kummer BR, Diaz I, Wu X, Aaroe AE, Chen ML, Iadecola C, Kamel H, Navi BB. Associations between cerebrovascular risk factors and Parkinson disease. Ann Neurol. 2019;86:572–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Morgan K, Lilley JM. Risk factors among incident cases of dementia in a representative british sample. Int J Geriatr Psychiatry. 1994;9:11–5. [Google Scholar]
- 55.Elwood PC, Bayer AJ, Fish M, Pickering J, Mitchell C, Gallacher JE. Sleep disturbance and daytime sleepiness predict vascular dementia. J Epidemiol Community Health. 2011;65:820–4. [DOI] [PubMed] [Google Scholar]
- 56.Chen P-L, Lee W-J, Sun W-Z, Oyang Y-J, Fuh J-L. Risk of dementia in patients with insomnia and long-term use of hypnotics: a population-based retrospective cohort study. PLoS One. 2012;7:e49113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Benedict C, Byberg L, Cedernaes J, Hogenkamp PS, Giedratis V, Kilander L, Lind L, Lannfelt L, Schiöth HB. Self-reported sleep disturbance is associated with Alzheimer’s disease risk in men. Alzheimer’s Dement. 2015;11:1090–7. [DOI] [PubMed] [Google Scholar]
- 58.Selbaek-Tungevåg S, Selbaek G, Strand BH, Myrstad C, Livingston G, Lydersen S, Bergh S, Ernstsen L. Insomnia and risk of dementia in a large population-based study with 11-year follow-up: the HUNT study. J Sleep Res. 2023;32:e13820. 10.1111/jsr.13820. [DOI] [PubMed] [Google Scholar]
- 59.Xiong Y, Tvedt J, Åkerstedt T, Cadar D, Wang HX. Impact of sleep duration and sleep disturbances on the incidence of dementia and Alzheimer’s disease: a 10-year follow-up study. Psychiatry Res. 2024;333:115760. 10.1016/j.psychres.2024.115760. [DOI] [PubMed] [Google Scholar]
- 60.Shieu MM, Dunietz GL, Paulson HL, Chervin RD, Braley TJ. The association between obstructive sleep apnea risk and cognitive disorders: a population-based study. J Clin Sleep Med. 2022;18:1177–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Damsgaard L, Janbek J, Laursen TM, Erlangsen A, Spira AP, Waldemar G. Hospital-diagnosed sleep disorders and incident dementia: a nationwide observational cohort study. Eur J Neurol. 2022;29:3528–36. 10.1111/ene.15517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Foley D, Monjan A, Masaki K, Ross W, Havlik R, White L, Launer L. Daytime sleepiness is associated with 3-year incident dementia and cognitive decline in older Japanese-American men. J Am Geriatr Soc. 2001;49:1628–32. [DOI] [PubMed] [Google Scholar]
- 63.Jaussent I, Bouyer J, Ancelin M-L, Berr C, Foubert-Samier A, Ritchie K, Ohayon MM, Besset A, Dauvilliers Y. Excessive sleepiness is predictive of cognitive decline in the elderly. Sleep. 2012;35:1201–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Lin C-C, Chou C-H, Fan Y-M, Yin J-H, Chung C-H, Chien W-C, Sung Y-F, Tsai C-K, Lin G-Y, Lin Y-K. Increased risk of dementia among sleep-related movement disorders: a population-based longitudinal study in Taiwan. Medicine. 2015;94:e2331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lim AS, Kowgier M, Yu L, Buchman AS, Bennett DA. Sleep fragmentation and the risk of incident Alzheimer’s disease and cognitive decline in older persons. Sleep. 2013;36:1027–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Tranah GJ, Blackwell T, Stone KL, Ancoli-Israel S, Paudel ML, Ensrud KE, Cauley JA, Redline S, Hillier TA, Cummings SR. Circadian activity rhythms and risk of incident dementia and mild cognitive impairment in older women. Ann Neurol. 2011;70:722–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Bokenberger K, Ström P, Dahl Aslan AK, Johansson AL, Gatz M, Pedersen NL, Åkerstedt T. Association between sleep characteristics and incident dementia accounting for baseline cognitive status: a prospective population-based study. J Gerontol Ser A Biomed Sci Med Sci. 2017;72:134–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Lobo A, LóPez-Antón R, De-La-CÁmara C, Quintanilla MÁ, Campayo A, Saz P, Workgroup Z. Non-cognitive psychopathological symptoms associated with incident mild cognitive impairment and dementia, Alzheimer’s type. Neurotoxicity Res. 2008;14:263–72. [DOI] [PubMed] [Google Scholar]
- 69.Hahn EA, Wang H-X, Andel R, Fratiglioni L. A change in sleep pattern may predict Alzheimer disease. Am J Geriatr Psychiatry. 2014;22:1262–71. [DOI] [PubMed] [Google Scholar]
- 70.Cavaillès C, Berr C, Helmer C, Gabelle A, Jaussent I, Dauvilliers Y. Complaints of daytime sleepiness, insomnia, hypnotic use, and risk of dementia: a prospective cohort study in the elderly. Alzheimers Res Ther. 2022;14:12. 10.1186/s13195-021-00952-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Lee W, Gray SL, Barthold D, Maust DT, Marcum ZA. Association between informant-reported sleep disturbance and incident dementia: an analysis of the National Alzheimer’s Coordinating Center Uniform Data Set. J Appl Gerontol. 2022;41:285–94. 10.1177/0733464820967202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Guo C, Harshfield EL, Markus HS. Sleep characteristics and risk of stroke and dementia: an observational and Mendelian randomization study. Neurology. 2024;102:e209141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kim KY, Kim EH, Lee M, Ha J, Jung I, Kim E. Restless leg syndrome and risk of all-cause dementia: a nationwide retrospective cohort study. Alzheimer’s Res Ther. 2023;15:46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Cavaillès C, Letellier N, Berr C, Samieri C, Empana JP, Tzourio C, Dartigues JF, Benmarhnia T, Dauvilliers Y, Jaussent I. The role of cardiovascular health and vascular events in the relationship between excessive daytime sleepiness and dementia risk. J Sleep Res. 2024;33:e14053. [DOI] [PubMed] [Google Scholar]
- 75.Liu R, Tang S, Wang Y, Dong Y, Hou T, Ren Y, Cong L, Liu K, Qin Y, Sindi S. Self-reported sleep characteristics associated with dementia among rural-dwelling Chinese older adults: a population-based study. BMC Neurol. 2022;22:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Baek MS, Han K, Kwon HS, Lee YH, Cho H, Lyoo CH. Risks and prognoses of Alzheimer’s disease and vascular dementia in patients with insomnia: a nationwide population-based study. Front Neurol. 2021;12:611446. 10.3389/fneur.2021.611446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Virta JJ, Heikkilä K, Perola M, Koskenvuo M, Räihä I, Rinne JO, Kaprio J. Midlife sleep characteristics associated with late life cognitive function. Sleep. 2013;36:1533–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Lee JE, Yang SW, Ju YJ, Ki SK, Chun KH. Sleep-disordered breathing and Alzheimer’s disease: a nationwide cohort study. Psychiatry Res. 2019;273:624–30. [DOI] [PubMed] [Google Scholar]
- 79.Tsai MS, Li HY, Huang CG, Wang RY, Chuang LP, Chen NH, Liu CH, Yang YH, Liu CY, Hsu CM. Risk of Alzheimer’s disease in obstructive sleep apnea patients with or without treatment: real-world evidence. Laryngoscope. 2020;130:2292–8. [DOI] [PubMed] [Google Scholar]
- 80.Osorio RS, Pirraglia E, Agüera-Ortiz LF, During EH, Sacks H, Ayappa I, Walsleben J, Mooney A, Hussain A, Glodzik L. Greater risk of Alzheimer’s disease in older adults with insomnia. J Am Geriatr Soc. 2011;59:559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Luojus MK, Lehto SM, Tolmunen T, Brem AK, Lönnroos E, Kauhanen J. Self-reported sleep disturbance and incidence of dementia in ageing men. J Epidemiol Community Health. 2017;71:329–35. 10.1136/jech-2016-207764. [DOI] [PubMed] [Google Scholar]
- 82.Wong R, Lovier MA. Sleep disturbances and dementia risk in older adults: findings from 10 years of national US prospective data. Am J Prev Med. 2023;64:781–7. [DOI] [PubMed] [Google Scholar]
- 83.Sterniczuk R, Theou O, Rusak B, Rockwood K. Sleep disturbance is associated with incident dementia and mortality. Curr Alzheimer Res. 2013;10:767–75. 10.2174/15672050113109990134. [DOI] [PubMed] [Google Scholar]
- 84.Asada T, Motonaga T, Yamagata Z, Uno M, Takahashi K. Associations between retrospectively recalled napping behavior and later development of Alzheimer’s disease: association with APOE genotypes. Sleep. 2000;23:629–34. [PubMed] [Google Scholar]
- 85.Li P, Gao L, Yu L, Zheng X, Ulsa MC, Yang HW, Gaba A, Yaffe K, Bennett DA, Buchman AS, Hu K, Leng Y. Daytime napping and Alzheimer’s dementia: a potential bidirectional relationship. Alzheimers Dement. 2023;19:158–68. 10.1002/alz.12636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Lysen TS, Wolters FJ, Luik AI, Ikram MK, Tiemeier H, Ikram MA. Subjective sleep quality is not associated with incident dementia: the Rotterdam study. J Alzheimers Dis. 2018;64:239–47. 10.3233/jad-180055. [DOI] [PubMed] [Google Scholar]
- 87.Sung PS, Yeh CC, Wang LC, Hung PH, Muo CH, Sung FC, Chen CH, Tsai KJ. Increased risk of dementia in patients with non-apnea sleep disorder. Curr Alzheimer Res. 2017;14:309–16. 10.2174/1567205013666161108104703. [DOI] [PubMed] [Google Scholar]
- 88.Dun C, Walsh CM, Chu NM, McAdams-DeMarco M, Hashim F, Makary MA. Sleep disorders and the development of Alzheimer’s disease among U.S. Medicare beneficiaries. J Am Geriatr Soc. 2022;70:299–301. 10.1111/jgs.17498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Nedelec T, Couvy-Duchesne B, Darves-Bornoz A, Couronne R, Monnet F, Gantzer L, Lekens B, Wu Y, Villain N, Schrag A, Durrleman S, Corvol JC. A comparison between early presentation of dementia with lewy bodies, Alzheimer’s disease, and Parkinson’s disease: evidence from routine primary care and UK Biobank data. Ann Neurol. 2023;94:259–70. 10.1002/ana.26670. [DOI] [PubMed] [Google Scholar]
- 90.Blackwell T, Yaffe K, Laffan A, Redline S, Ancoli-Israel S, Ensrud KE, Song Y, Stone KL, Group OFiMS. Associations between sleep-disordered breathing, nocturnal hypoxemia, and subsequent cognitive decline in older community-dwelling men: the osteoporotic fractures in men sleep study. J Am Geriatr Soc. 2015;63:453–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Lutsey PL, Bengtson LG, Punjabi NM, Shahar E, Mosley TH, Gottesman RF, Wruck LM, MacLehose RF, Alonso A. Obstructive sleep apnea and 15-year cognitive decline: the atherosclerosis risk in communities (ARIC) study. Sleep. 2016;39:309–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Martin MS, Sforza E, Roche F, Barthelemy JC, Thomas-Anterion C, group Ps. Sleep breathing disorders and cognitive function in the elderly: an 8-year follow-up study the proof-synapse cohort. Sleep. 2015;38:179–87. 10.5665/sleep.4392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Sharafkhaneh A, Giray N, Richardson P, Young T, Hirshkowitz M. Association of psychiatric disorders and sleep apnea in a large cohort. Sleep. 2005;28:1405–11. [DOI] [PubMed] [Google Scholar]
- 94.Ding X, Kryscio RJ, Turner J, Jicha GA, Cooper G, Caban-Holt A, Schmitt FA, Abner EL. Self-reported sleep apnea and dementia risk: findings from the prevention of Alzheimer’s disease with vitamin E and selenium trial. J Am Geriatr Soc. 2016;64:2472–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Choe YM, Suh G-H, Kim JW, Initiative AsDN. Association of a history of sleep disorder with risk of mild cognitive impairment and Alzheimer’s disease dementia. Psychiatry Investig. 2022;19:840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Agudelo C, Ramos AR, Sun X, Kaur S, Del Papa DF, Kather JM, Wallace DM, Initiative AsDN. Sleep-disordered breathing risk with comorbid insomnia is associated with mild cognitive impairment. Appl Sci. 2022;12:2414. [Google Scholar]
- 97.Bubu OM, Pirraglia E, Andrade AG, Sharma RA, Gimenez-Badia S, Umasabor-Bubu OQ, Hogan MM, Shim AM, Mukhtar F, Sharma N, Mbah AK, Seixas AA, Kam K, Zizi F, Borenstein AR, Mortimer JA, Kip KE, Morgan D, Rosenzweig I, Ayappa I, Rapoport DM, Jean-Louis G, Varga AW, Osorio RS. Obstructive sleep apnea and longitudinal Alzheimer’s disease biomarker changes. Sleep. 2019;42(6):zsz048. 10.1093/sleep/zsz048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Cricco M, Simonsick EM, Foley DJ. The impact of insomnia on cognitive functioning in older adults. J Am Geriatr Soc. 2001;49:1185–9. 10.1046/j.1532-5415.2001.49235.x. [DOI] [PubMed] [Google Scholar]
- 99.Lin W, Lin YK, Yang FC, Chung CH, Hu JM, Tsao CH, Weng ZX, Ko CA, Chien WC. Risk of neurodegenerative diseases in patients with sleep disorders: a nationwide population-based case-control study. Sleep Med. 2023;107:289–99. 10.1016/j.sleep.2023.05.014. [DOI] [PubMed] [Google Scholar]
- 100.Tworoger SS, Lee S, Schernhammer ES, Grodstein F. The association of self-reported sleep duration, difficulty sleeping, and snoring with cognitive function in older women. Alzheimer Dis Assoc Disord. 2006;20:41–8. 10.1097/01.wad.0000201850.52707.80. [DOI] [PubMed] [Google Scholar]
- 101.Keage HA, Banks S, Yang KL, Morgan K, Brayne C, Matthews FE. What sleep characteristics predict cognitive decline in the elderly? Sleep Med. 2012;13:886–92. 10.1016/j.sleep.2012.02.003. [DOI] [PubMed] [Google Scholar]
- 102.Zhang L, Li T, Lei Y, Cheng G, Liu B, Yu Y, Yin H, Song L, La Q, Li B. Association between sleep structure and amnesic mild cognitive impairment in patients with insomnia disorder: a case-control study. J Clin Sleep Med. 2021;17:37–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Mecca AP, Michalak HR, McDonald JW, Kemp EC, Pugh EA, Becker ML, Mecca MC, van Dyck CH. Sleep disturbance and the risk of cognitive decline or clinical conversion in the ADNI cohort. Dement Geriatr Cogn Disord. 2018;45:232–42. 10.1159/000488671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Blackwell T, Yaffe K, Laffan A, Ancoli-Israel S, Redline S, Ensrud KE, Song Y, Stone KL. Associations of objectively and subjectively measured sleep quality with subsequent cognitive decline in older community-dwelling men: the MrOS sleep study. Sleep. 2014;37:655–63. 10.5665/sleep.3562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Geda YE, Roberts RO, Mielke MM, Knopman DS, Christianson TJ, Pankratz VS, Boeve BF, Sochor O, Tangalos EG, Petersen RC, Rocca WA. Baseline neuropsychiatric symptoms and the risk of incident mild cognitive impairment: a population-based study. Am J Psychiatry. 2014;171:572–81. 10.1176/appi.ajp.2014.13060821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Potvin O, Lorrain D, Forget H, Dubé M, Grenier S, Préville M, Hudon C. Sleep quality and 1-year incident cognitive impairment in community-dwelling older adults. Sleep. 2012;35:491–9. 10.5665/sleep.1732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Zawar I, Mattos MK, Manning C, Patrie J, Quigg M. Sleep disturbances predict cognitive decline in cognitively healthy adults. J Alzheimers Dis. 2023;92:1427–38. 10.3233/jad-221244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Vgontzas AN, Bixler EO, Lin HM, Prolo P, Mastorakos G, Vela-Bueno A, Kales A, Chrousos GP. Chronic insomnia is associated with nyctohemeral activation of the hypothalamic-pituitary-adrenal axis: clinical implications. J Clin Endocrinol Metab. 2001;86:3787–94. 10.1210/jcem.86.8.7778. [DOI] [PubMed] [Google Scholar]
- 109.Astara K, Tsimpolis A, Kalafatakis K, Vavougios GD, Xiromerisiou G, Dardiotis E, Christodoulou NG, Samara MT, Lappas AS. Sleep disorders and Alzheimer’s disease pathophysiology: the role of the Glymphatic system. A scoping review. Mech Ageing Dev. 2024;217:111899. 10.1016/j.mad.2023.111899. [DOI] [PubMed] [Google Scholar]
- 110.Eide PK, Pripp AH, Berge B, Hrubos-Strom H, Ringstad G, Valnes LM. Altered glymphatic enhancement of cerebrospinal fluid tracer in individuals with chronic poor sleep quality. J Cereb Blood Flow Metab. 2022;42:1676–92. 10.1177/0271678X221090747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Kroesbergen E, Riesselmann LV, Gomolka RS, Pla V, Esmail T, Stenmo VH, Kovacs ER, Nielsen ES, Goldman SA, Nedergaard M, Weikop P, Mori Y. Glymphatic clearance is enhanced during sleep. bioRxiv. 2024. 10.1101/2024.08.24.609514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Gavrilin MA, Porter K, Samouilov A, Khayat RN. Pathways of microcirculatory endothelial dysfunction in obstructive sleep apnea: a comprehensive ex vivo evaluation in human tissue. Am J Hypertens. 2022;35:347–55. 10.1093/ajh/hpab169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Sawatari H, Chishaki A, Nishizaka M, Tokunou T, Adachi S, Yoshimura C, Ohkusa T, Ando S. Cumulative hypoxemia during sleep predicts vascular endothelial dysfunction in patients with sleep-disordered breathing. Am J Hypertens. 2016;29:458–63. 10.1093/ajh/hpv135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Stockelman KA, Bain AR, Dow CA, Diehl KJ, Greiner JJ, Stauffer BL, DeSouza CA. Regular aerobic exercise counteracts endothelial vasomotor dysfunction associated with insufficient sleep. Am J Physiol Heart Circ Physiol. 2021;320:H1080–8. 10.1152/ajpheart.00615.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.He J, Hsuchou H, He Y, Kastin AJ, Wang Y, Pan W. Sleep restriction impairs blood-brain barrier function. J Neurosci. 2014;34:14697–706. 10.1523/JNEUROSCI.2111-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Hurtado-Alvarado G, Dominguez-Salazar E, Pavon L, Velazquez-Moctezuma J, Gomez-Gonzalez B. Blood-brain barrier disruption induced by chronic sleep loss: low-grade inflammation may be the link. J Immunol Res. 2016;2016:4576012. 10.1155/2016/4576012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Puech C, Badran M, Runion AR, Barrow MB, Cataldo K, Gozal D. Cognitive impairments, neuroinflammation and blood-brain barrier permeability in mice exposed to chronic sleep fragmentation during the daylight period. Int J Mol Sci. 2023;24(12):9880. 10.3390/ijms24129880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Sun J, Wu J, Hua F, Chen Y, Zhan F, Xu G. Sleep deprivation induces cognitive impairment by increasing blood-brain barrier permeability via CD44. Front Neurol. 2020;11:563916. 10.3389/fneur.2020.563916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Csipo T, Lipecz A, Owens C, Mukli P, Perry JW, Tarantini S, Balasubramanian P, Nyul-Toth A, Yabluchanska V, Sorond FA, Kellawan JM, Purebl G, Sonntag WE, Csiszar A, Ungvari Z, Yabluchanskiy A. Sleep deprivation impairs cognitive performance, alters task-associated cerebral blood flow and decreases cortical neurovascular coupling-related hemodynamic responses. Sci Rep. 2021;11:20994. 10.1038/s41598-021-00188-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Mukli P, Csipo T, Lipecz A, Stylianou O, Racz FS, Owens CD, Perry JW, Tarantini S, Sorond FA, Kellawan JM, Purebl G, Yang Y, Sonntag WE, Csiszar A, Ungvari ZI, Yabluchanskiy A. Sleep deprivation alters task-related changes in functional connectivity of the frontal cortex: a near-infrared spectroscopy study. Brain Behav. 2021;11:e02135. 10.1002/brb3.2135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Gills JL, Bubu OM. Obstructive sleep apnea and Alzheimer’s disease pathology: is sleep architecture the missing key? J Alzheimers Dis. 2024;98:69–73. 10.3233/JAD-231385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Irwin MR. Sleep and inflammation: partners in sickness and in health. Nat Rev Immunol. 2019;19:702–15. 10.1038/s41577-019-0190-z. [DOI] [PubMed] [Google Scholar]
- 123.Miller MA, Kandala NB, Kivimaki M, Kumari M, Brunner EJ, Lowe GD, Marmot MG, Cappuccio FP. Gender differences in the cross-sectional relationships between sleep duration and markers of inflammation: Whitehall II study. Sleep. 2009;32:857–64. [PMC free article] [PubMed] [Google Scholar]
- 124.Korsiak J, Tranmer J, Day A, Aronson KJ. Sleep duration as a mediator between an alternating day and night shift work schedule and metabolic syndrome among female hospital employees. Occup Environ Med. 2018;75:132–8. 10.1136/oemed-2017-104371. [DOI] [PubMed] [Google Scholar]
- 125.Wehrens SM, Hampton SM, Finn RE, Skene DJ. Effect of total sleep deprivation on postprandial metabolic and insulin responses in shift workers and non-shift workers. J Endocrinol. 2010;206:205–15. 10.1677/JOE-10-0077. [DOI] [PubMed] [Google Scholar]
- 126.Yoon SJ, Long NP, Jung KH, Kim HM, Hong YJ, Fang Z, Kim SJ, Kim TJ, Anh NH, Hong SS, Kwon SW. Systemic and local metabolic alterations in sleep-deprivation-induced stress: a multiplatform mass-spectrometry-based lipidomics and metabolomics approach. J Proteome Res. 2019;18:3295–304. 10.1021/acs.jproteome.9b00234. [DOI] [PubMed] [Google Scholar]
- 127.Kecklund G, Axelsson J. Health consequences of shift work and insufficient sleep. BMJ. 2016;355:i5210. 10.1136/bmj.i5210. [DOI] [PubMed] [Google Scholar]
- 128.Fekete M, Major D, Feher A, Fazekas-Pongor V, Lehoczki A. Geroscience and pathology: a new frontier in understanding age-related diseases. Pathol Oncol Res. 2024. 10.3389/pore.2024.1611623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Tessema B, Sack U, Konig B, Serebrovska Z, Egorov E. Effects of intermittent hypoxia in training regimes and in obstructive sleep apnea on aging biomarkers and age-related diseases: a systematic review. Front Aging Neurosci. 2022;14:878278. 10.3389/fnagi.2022.878278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Sosnowski DW, Smail EJ, Maher BS, Moore AZ, Kuo PL, Wu MN, Low DV, Stone KL, Simonsick EM, Ferrucci L, Spira AP. Sleep duration polygenic risk and phenotype: associations with biomarkers of accelerated aging in the Baltimore longitudinal study of aging. Int J Aging Hum Dev. 2024;100(2):135–64. 10.1177/00914150241231192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Noroozi R, Rudnicka J, Pisarek A, Wysocka B, Masny A, Boron M, Migacz-Gruszka K, Pruszkowska-Przybylska P, Kobus M, Lisman D, Zielinska G, Iljin A, Wiktorska JA, Michalczyk M, Kaczka P, Krzysztofik M, Sitek A, Ossowski A, Spolnicka M, Branicki W, Pospiech E. Analysis of epigenetic clocks links yoga, sleep, education, reduced meat intake, coffee, and a SOCS2 gene variant to slower epigenetic aging. Geroscience. 2024;46:2583–604. 10.1007/s11357-023-01029-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Baldelli L, Pirazzini C, Sambati L, Ravaioli F, Gentilini D, Calandra-Buonaura G, Guaraldi P, Franceschi C, Cortelli P, Garagnani P, Bacalini MG, Provini F. Epigenetic clocks suggest accelerated aging in patients with isolated REM sleep behavior disorder. NPJ Parkinsons Dis. 2023;9:48. 10.1038/s41531-023-00492-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Carroll JE, Ross KM, Horvath S, Okun M, Hobel C, Rentscher KE, Coussons-Read M, Schetter CD. Postpartum sleep loss and accelerated epigenetic aging. Sleep Health. 2021;7:362–7. 10.1016/j.sleh.2021.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Villafuerte G, Miguel-Puga A, Rodriguez EM, Machado S, Manjarrez E, Arias-Carrion O. Sleep deprivation and oxidative stress in animal models: a systematic review. Oxid Med Cell Longev. 2015;2015:234952. 10.1155/2015/234952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Zhang YM, Wang YT, Wei RM, Li XY, Luo BL, Zhang JY, Zhang KX, Fang SK, Liu XC, Chen GH. Mitochondrial antioxidant elamipretide improves learning and memory impairment induced by chronic sleep deprivation in mice. Brain Behav. 2024;14:e3508. 10.1002/brb3.3508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Zhao H, Wu H, He J, Zhuang J, Liu Z, Yang Y, Huang L, Zhao Z. Frontal cortical mitochondrial dysfunction and mitochondria-related beta-amyloid accumulation by chronic sleep restriction in mice. NeuroReport. 2016;27:916–22. 10.1097/WNR.0000000000000631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Mc Carthy CE, Yusuf S, Judge C, Alvarez-Iglesias A, Hankey GJ, Oveisgharan S, Damasceno A, Iversen HK, Rosengren A, Avezum A, Lopez-Jaramillo P, Xavier D, Wang X, Rangarajan S, O’Donnell M. Sleep patterns and the risk of acute stroke: results from the INTERSTROKE international case-control study. Neurology. 2023;100:e2191–203. 10.1212/WNL.0000000000207249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.McDermott M, Brown DL, Chervin RD. Sleep disorders and the risk of stroke. Expert Rev Neurother. 2018;18:523–31. 10.1080/14737175.2018.1489239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Ferrie JE, Shipley MJ, Cappuccio FP, Brunner E, Miller MA, Kumari M, Marmot MG. A prospective study of change in sleep duration: associations with mortality in the Whitehall II cohort. Sleep. 2007;30:1659–66. 10.1093/sleep/30.12.1659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Cappuccio FP, D’Elia L, Strazzullo P, Miller MA. Sleep duration and all-cause mortality: a systematic review and meta-analysis of prospective studies. Sleep. 2010;33:585–92. 10.1093/sleep/33.5.585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Chen Y, Tan F, Wei L, Li X, Lyu Z, Feng X, Wen Y, Guo L, He J, Dai M, Li N. Sleep duration and the risk of cancer: a systematic review and meta-analysis including dose-response relationship. BMC Cancer. 2018;18:1149. 10.1186/s12885-018-5025-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Papantoniou K, Konrad P, Haghayegh S, Strohmaier S, Eliassen AH, Schernhammer E. Rotating night shift work, sleep, and thyroid cancer risk in the Nurses’ Health Study 2. Cancers (Basel). 2023;15(23):5673. 10.3390/cancers15235673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Dickerman BA, Markt SC, Koskenvuo M, Hublin C, Pukkala E, Mucci LA, Kaprio J. Sleep disruption, chronotype, shift work, and prostate cancer risk and mortality: a 30-year prospective cohort study of Finnish twins. Cancer Causes Control. 2016;27:1361–70. 10.1007/s10552-016-0815-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Ungvari Z, Tabak AG, Adany R, Purebl G, Kaposvari C, Fazekas-Pongor V, Csipo T, Szarvas Z, Horvath K, Mukli P, Balog P, Bodizs R, Ujma P, Stauder A, Belsky DW, Kovacs I, Yabluchanskiy A, Maier AB, Moizs M, Ostlin P, Yon Y, Varga P, Voko Z, Papp M, Takacs I, Vasarhelyi B, Torzsa P, Ferdinandy P, Csiszar A, Benyo Z, et al. The Semmelweis Study: a longitudinal occupational cohort study within the framework of the Semmelweis Caring University Model Program for supporting healthy aging. Geroscience. 2024;46:191–218. 10.1007/s11357-023-01018-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Fekete M, Szarvas Z, Fazekas-Pongor V, Feher A, Csipo T, Forrai J, Dosa N, Peterfi A, Lehoczki A, Tarantini S, Varga JT. Nutrition strategies promoting healthy aging: from improvement of cardiovascular and brain health to prevention of age-associated diseases. Nutrients. 2022;15(1):47. 10.3390/nu15010047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Fekete M, Lehoczki A, Tarantini S, Fazekas-Pongor V, Csipo T, Csizmadia Z, Varga JT. Improving cognitive function with nutritional supplements in aging: a comprehensive narrative review of clinical studies investigating the effects of vitamins, minerals, antioxidants, and other dietary supplements. Nutrients. 2023;15(24):5116. 10.3390/nu15245116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Cao X, Wang M, Zhou M, Mi Y, Fazekas-Pongor V, Major D, Lehoczki A, Guo Y. Trends in prevalence, mortality, and risk factors of dementia among the oldest-old adults in the United States: the role of the obesity epidemic. Geroscience. 2024. 10.1007/s11357-024-01180-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Iannuzzi V, Narboux-Neme N, Lehoczki A, Levi G, Giuliani C. Stay social, stay young: a bioanthropological outlook on the processes linking sociality and ageing. Geroscience. 2024. 10.1007/s11357-024-01416-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Cao X, Peng H, Hu Z, Xu C, Ning M, Zhou M, Mi Y, Yu P, Fazekas-Pongor V, Major D, Ungvari Z, Fekete M, Lehoczki A, Guo Y. Exploring the global impact of obesity and diet on dementia burden: the role of national policies and sex differences. Geroscience. 2024. 10.1007/s11357-024-01457-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Pandics T, Major D, Fazekas-Pongor V, Szarvas Z, Peterfi A, Mukli P, Gulej R, Ungvari A, Fekete M, Tompa A, Tarantini S, Yabluchanskiy A, Conley S, Csiszar A, Tabak AG, Benyo Z, Adany R, Ungvari Z. Exposome and unhealthy aging: environmental drivers from air pollution to occupational exposures. Geroscience. 2023;45:3381–408. 10.1007/s11357-023-00913-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Ungvari Z, Adany R, Szabo AJ, Dornyei G, Moizs M, Purebl G, Kalabay L, Varga P, Torzsa P, Kellermayer M, Merkely B. Semmelweis Caring University Model Program based on the development of a center of preventive services: health for all employees at a university occupational setting. Front Public Health. 2021;9:727668. 10.3389/fpubh.2021.727668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Vitale E, Lupo R, Artioli G, Mea R, Lezzi P, Conte L, De Nunzio G. How shift work influences anxiety, depression, stress and insomnia conditions in Italian nurses: an exploratory study. Acta Biomed. 2023;94:e2023102. 10.23750/abm.v94i2.14230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Rosa D, Terzoni S, Dellafiore F, Destrebecq A. Systematic review of shift work and nurses’ health. Occup Med (Lond). 2019;69:237–43. 10.1093/occmed/kqz063. [DOI] [PubMed] [Google Scholar]
- 154.Ki J, Choi-Kwon S. Health problems, turnover intention, and actual turnover among shift work female nurses: analyzing data from a prospective longitudinal study. PLoS One. 2022;17:e0270958. 10.1371/journal.pone.0270958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.de Bruijn L, Berentzen NE, Vermeulen RCH, Vlaanderen JJ, Kromhout H, van Leeuwen FE, Schaapveld M. Chronotype in relation to shift work: a cohort study among 37,731 female nurses. J Sleep Res. 2024;e14308. 10.1111/jsr.14308 [DOI] [PMC free article] [PubMed]
- 156.Carroll CM, Macauley SL. The interaction between sleep and metabolism in Alzheimer’s disease: cause or consequence of disease? Front Aging Neurosci. 2019;11:258. 10.3389/fnagi.2019.00258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Lacerda RAV, Desio JAF, Kammers CM, Henkes S, Freitas de Sa M, de Souza EF, da Silva DM, Teixeira Pinheiro Gusmao C, Santos J. Sleep disorders and risk of Alzheimer’s disease: a two-way road. Ageing Res Rev. 2024;101:102514. 10.1016/j.arr.2024.102514. [DOI] [PubMed] [Google Scholar]
- 158.Liu X, Wei Z, Chen L, Duan W, Li H, Kong L, Shu Y, Li P, Li K, Xie W, Zeng Y, Huang L, Long T, Peng D. Effects of 3-month CPAP therapy on brain structure in obstructive sleep apnea: a diffusion tensor imaging study. Front Neurol. 2022;13:913193. 10.3389/fneur.2022.913193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Li J, Yan W, Yi M, Lin R, Huang Z, Zhang Y. Efficacy of CPAP duration and adherence for cognitive improvement in patients with obstructive sleep apnea: a meta-analysis of randomized controlled trials. Sleep Breath. 2023;27:973–82. 10.1007/s11325-022-02687-y. [DOI] [PubMed] [Google Scholar]
- 160.Costa YS, Lim ASP, Thorpe KE, Colelli DR, Mitchell S, Masellis M, Lam B, Black SE, Boulos MI. Investigating changes in cognition associated with the use of CPAP in cognitive impairment and dementia: a retrospective study. Sleep Med. 2023;101:437–44. 10.1016/j.sleep.2022.11.037. [DOI] [PubMed] [Google Scholar]
- 161.Velescu DR, Marc MS, Pescaru CC, Traila D, Vastag E, Papava I, Motofelea AC, Ciuca IM, Manolescu D, Oancea C. Impact of CPAP therapy adherence on global cognition in patients with moderate to severe obstructive sleep apnea: a one-year follow-up. Medicina (Kaunas). 2023;59(5):846. 10.3390/medicina59050846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Costa YS, Boulos MI. CPAP adherence and cognitive function in OSA: addressing discussion points and controversies. Sleep Med. 2023;109:303. 10.1016/j.sleep.2023.07.025. [DOI] [PubMed] [Google Scholar]
- 163.Xu H, Liu Y, Li C, Li X, Shen L, Wang H, Liu F, Zou J, Xia Y, Huang W, Liu Y, Gao Z, Fu Y, Wang F, Huang S, Song Z, Song F, Gao Y, Peng Y, Zou J, Zhu H, Liu S, Li L, Zhu X, Xiong Y, Hu Y, Yang J, Li Y, Gao F, Guo Q, et al. Effects of CPAP on neuroimaging biomarkers and cognition in adult OSA: a randomized controlled trial. Am J Respir Crit Care Med. 2025. 10.1164/rccm.202406-1170OC. [DOI] [PubMed] [Google Scholar]
- 164.Benkirane O, Mairesse O, Peigneux P. Impact of CPAP therapy on cognition and fatigue in patients with moderate to severe sleep apnea: a longitudinal observational study. Clocks Sleep. 2024;6:789–816. 10.3390/clockssleep6040051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Franks KH, Rowsthorn E, Nicolazzo J, Boland A, Lavale A, Baker J, Rajaratnam SMW, Cavuoto MG, Yiallourou SR, Naughton MT, Hamilton GS, Churilov L, Lim YY, Pase MP. The treatment of sleep dysfunction to improve cognitive function: a meta-analysis of randomized controlled trials. Sleep Med. 2023;101:118–26. 10.1016/j.sleep.2022.10.021. [DOI] [PubMed] [Google Scholar]






