281
Sleep and Alzheimer’s disease: A critical examination of the risk that Sleep Problems or Disorders particularly Obstructive Sleep Apnea pose towards developing Alzheimer’s disease by Omonigho A. Michael Bubu A dissertation submitted in partial fulfillment of the requirements of the degree of Doctor of Philosophy Public Health with a concentration in Epidemiology Department of Epidemiology and Biostatistics College of Public Health University of South Florida Major Professor: Kevin Kip, Ph.D., FAHA, AAAS Fellow Co-Major Professor: Dave Morgan, Ph.D. Alfred Mbah, Ph.D. Ricardo Osorio, M.D. MA Date of Approval: November 16, 2017 Keywords: Mild Cognitive Impairment, Beta Amyloid, Cerebrospinal Fluid, Hippocampal Atrophy Copyright © 2017, Omonigho A. Michael Bubu

Omonigho A. Michael Bubu

  • Upload
    others

  • View
    7

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Omonigho A. Michael Bubu

Sleep and Alzheimer’s disease:

A critical examination of the risk that Sleep Problems or Disorders particularly Obstructive Sleep Apnea

pose towards developing Alzheimer’s disease

by

Omonigho A. Michael Bubu

A dissertation submitted in partial fulfillment of the requirements of the degree of Doctor of Philosophy Public Health

with a concentration in Epidemiology Department of Epidemiology and Biostatistics

College of Public Health University of South Florida

Major Professor: Kevin Kip, Ph.D., FAHA, AAAS Fellow Co-Major Professor: Dave Morgan, Ph.D.

Alfred Mbah, Ph.D. Ricardo Osorio, M.D. MA

Date of Approval: November 16, 2017

Keywords: Mild Cognitive Impairment, Beta Amyloid, Cerebrospinal Fluid, Hippocampal Atrophy

Copyright © 2017, Omonigho A. Michael Bubu

Page 2: Omonigho A. Michael Bubu

DEDICATION

First, I humbly dedicate this body of work to God Almighty, who graced and enabled me to be

able to do this. Without YOU Lord, I would not have had this opportunity. I am truly thankful for YOUR

infinite mercies and grace.

Second, I dedicate this work to my wife, Queen Ogie Umasabor-Bubu, M.D., M.P.H, CPH, CIC.

Your unwavering love, support and resilience through thick and thin, I sincerely appreciate. Without you,

this could not have materialized.

Third, I dedicate this to my kids, Oruno Darah Bubu, and Oreva Charis Bubu. Thank you for

being the best kids in the world and coping with daddy and mummy, as we joggled being international

students and being parents at the same time. Your smiles and the joy you bring to our lives, make the

challenges we faced worthwhile. You both, as well as mummy, are truly God’s expression of HIS divine

love towards me.

Lastly, I dedicate this work to the memory of parents, Chief Samuel Onogharigho Bubu, and Mrs.

Elizabeth Itete-Bubu. Your labor was not in vain, and even though it saddens my heart that both of you

are not around to see us your children achieve our dreams and goals in life, be rest assured that we would

do you proud. Keep on resting in the bosom of our Lord.

Page 3: Omonigho A. Michael Bubu

ACKNOWLEDGMENTS

I thank my committee members for their support, encouragement and belief in me. Dr. Kevin

Kip, thank you for accepting to be my major advisor and providing me the opportunity to see this work

through. To me, you are God sent, and I am truly grateful for your guidance, and counsel. Dr. Dave

Morgan, thank you for standing by me in my most trying period. Your belief in me is palpable and I am

extremely fortunate to have such an authority in the field in my committee. Dr. Ricardo Osorio, thank you

for welcoming me with open arms and having confidence in me. Your belief in me emboldens me to aim

higher in the field as a researcher. Your friendship is of inestimable value. Dr. Alfred Mbah, thank you for

being a friend and a brother. Your help and guidance in helping me consolidate my understanding of SAS

and helping me whenever I was stuck, is sincerely appreciated. I must thank Dr. Jim Mortimer and Dr.

Amy Borenstein for your mentorship, counsel, guidance and support in my most trying time. Thank you

for your constant belief in me, and pushing me to know no limitations. Dr. Karen Liller, I am eternally

thankful for your mentorship and grace. You stood by me in my most trying times, patient, insightful and

empathetic. You all have contributed in no small way to any success I have had.

I am thankful for all my critics, for you made me a better person. I am grateful for librarians

Allison Howard and John Orriola’s assistance in developing search strategies as I embarked on my

research. I am also thankful for Pharmacists Kayode and Salewa Ogundipe; Pastor Zak Moussa, Pastor

Mathew Owotoki, Pastors (Mr. & Mrs. Faith), Pastor Isaac Olabisi and Rev. Adetunji for their spiritual

and moral guidance. You all were my helpers of the war and I remain eternally grateful. Thank you to all

my other professors and academic staff for making me the academic that I am today. God bless you all.

Page 4: Omonigho A. Michael Bubu

i

TABLE OF CONTENTS

List of Tables v

List of Figures vii

Abstract ix

Section 1: Systematic Review: Sleep and Alzheimer’s disease 1 Note to the Reader 1 Abstract 1 Introduction 3 Sleep and Normal Aging 3 Sleep and Demented Older Adults 4 Previous narrative reviews 4

Objective of this systematic review 5 Methods 5

Identification of Eligible studies 6 Electronic search 6 Searching other sources 7 Selection criteria 7

Type of study 7 Sleep abnormalities and/or sleep disorders 8 Cognitive decline and/or Alzheimer’s disease 9 Data extraction 9 Assessment of study quality 10 Risk of bias in individual studies 11 Observational data from studies 11

Results 11 Study selection 11

Study characteristics 12 Participant and setting 13

Sleep abnormalities and/or disorders 13 Cognitive decline and/or Alzheimer’s dementia 14 Study findings 14

A. Sleep depth, cognitive decline and Alzheimer’s disease 14 A.1: Cross-sectional studies 15

A.2: Case-control studies 17 A.3: Cohort studies 19 A.4: Experimental studies 22 A.5: Randomized Controlled Trials 25

B. Sleep wake-cycle/circadian rhythm, Cognitive decline and Alzheimer’s disease 25

Page 5: Omonigho A. Michael Bubu

ii

B.1: Cross-sectional studies 25 B.2: Case-control studies 27

B.3: Cohort studies 29 B.4: Experimental studies 30 B.5: Randomized Controlled Trials 33

C. Hypoxia, Cognitive decline and Alzheimer’s disease 33 C.1: Cross-sectional studies 33 C.2: Case-control studies 35 C.3: Cohort studies 35 C.4: Experimental studies 36 C.5: Randomized Control Trials 39 D. Insomnia, Cognitive decline and Alzheimer’s disease 39 D.1: Cross-sectional studies 39 D.2: Cohort studies 40 E. Restless Leg Syndrome, Cognitive decline and Alzheimer’s disease 42 E.1: Cross-sectional studies 42 E.2: Case-control studies 42

Discussion 43 Sleep depth, Cognitive decline and Alzheimer’s disease 43

Circadian Rhythm abnormalities, Cognitive decline and Alzheimer’s disease 46 Hypoxia, Cognitive decline and Alzheimer’s disease 48 Insomnia, Cognitive decline and Alzheimer’s disease 50 Restless Leg Syndrome, Cognitive decline and Alzheimer’s disease 50 Summary of evidence 50

Sleep is associated with cognitive decline and/or AD/AD pathology 50 Sleep problems precede the onset of cognitive impairment 52

Sleep problems may lead to increased AD pathology or may have their effects on dementia through other pathways 53

Limitations 54 Conclusion and Future Directions 54 Research agenda and gaps in literature I intend to address 55 References 56 Section 2: Sleep, Cognitive Impairment and Alzheimer’s disease: A Systematic Review and Meta- Analysis 110

Note to the Reader 110 List of Authors 110 Author contributions 111

Abstract 111 Introduction 113 Methods 113

Identification of eligible studies 114 Electronic search 114 Searching other sources 114 Selection criteria 115

Types of studies 115 Sleep problems and/or disorders 115 Alzheimer’s disease/cognitive impairment 116 Participant and setting 116 Data extraction 116 Assessment of study quality 116

Page 6: Omonigho A. Michael Bubu

iii

Statistical analysis 117 Results 118

Study selection 118 Study characteristics 119

Design and quality 119 Participant and setting 119

Sleep problems and/or disorders 120 Cognitive impairment and/or Alzheimer’s disease 120 Meta-analysis 121

Overall meta-analysis based on risk estimates for the effect of sleep on cognitive impairment and Alzheimer’s disease 121 Sub-group meta-analysis based on risk estimates for the effect of sleep on cognitive impairment, Pre-clinical and ICD9/DSMIV Diagnoses of Alzheimer’s disease 121 Funnel Plot Asymmetry Test and Trim and Fill Analysis Assessment of Publication Bias 122 Sub-group meta-analysis based on risk estimates for the effect of different sleep problems and disorders on cognitive impairment and Alzheimer’s disease 122 Sub-group meta-analysis based on risk estimates for the effect of self-report and actigraphic data of poor sleep on cognitive impairment and/or Alzheimer’s disease 123 Exploring Potential Causes of Heterogeneity 124 Sensitivity Analyses 124

Meta-regression analysis 126 Population Attributable Risk percent 126

Discussion 126 Summary of main results 126 Effect of sleep problems on cognitive impairment or Alzheimer’s disease (meta-analysis) 127 Potentially influencing factors (meta-regression analysis) 128 Study results in comparison with other reviews on the topic 128 Strengths and Limitations of the study 129 Future directions 131 Conclusion 131

References 132 Appendix A Supplemental Tables and Figures: Sleep and AD Meta-analysis 153

Section 3: Obstructive Sleep Apnea is associated with longitudinal increases in brain Aβ42, CSF-TAU, PTAU, and decrease in CSF Aβ42 burden, in elderly Cognitive Normal and MCI Individuals 165

Key Points 165 Abstract 165 Introduction 167 Methods 168

Study Participants 168 OSA diagnosis 169 NL, MCI and AD diagnosis 169 Florbetapir-PET Imaging Acquisition and Interpretation 169 Cerebrospinal Fluid Methods 169

Page 7: Omonigho A. Michael Bubu

iv

Covariates/Potential Confounders 170 Data Analysis 170

Results 171 Demographic and Clinical Characteristics 171 Baseline AD Biomarker levels by clinical group and OSA status 172 Rate of change in AD Biomarker by OSA status 172

Brain Aβ-42 levels 172 CSF Aβ-42, TAU and PTAU levels 173

Discussions 173 Baseline AD Biomarker levels by clinical group and OSA status 174 Rate of change in AD Biomarker by OSA status 174 Strengths and Limitations 176

Conclusions 176 References 176

Section 4: Obstructive Sleep Apnea: A Distinct Physiological Phenotypic Risk Factor in older adults with Cognitive decline and Alzheimer’s disease 189

Abstract 189 Introduction 190

Age-related and Age-dependent OSA co-morbidities 190 Review Objective 193

Methods 193 Search strategy 193 Selection criteria 194 Reviewing procedure and Data extraction 195 Assessment of study quality 195

Results 196 OSA and Cognition (Cross-sectional studies) 196

Main findings 196 OSA and Cognitive Decline (Longitudinal studies) 199

Main findings 199 Summary, critique and future research directions 200

OSA and Alzheimer’s disease (Cross-sectional studies) 202 Main findings 202

OSA and AD Pathology 203 Main findings 203

Cross-sectional studies 203 Prospective/Longitudinal studies 204 Randomized Controlled Trials 205

Summary, critique and future research directions 206 Discussion 208 Conclusion 211 References 211

Appendix B Oxford University Press License Terms and Conditions 249 Appendix C IRB Letter 252 Appendix D Sleep, Cognitive Decline, and Alzheimer’s disease: A Systematic Review and Meta- Analysis 254

Page 8: Omonigho A. Michael Bubu

v

LIST OF TABLES

Table 1.1 Literature Search Terms for the Systematic Review of Sleep and Alzheimer’s disease 78

Table 1.2 Scoring System for Quality of Paper of the Systematic Review of Sleep and Alzheimer’s disease 79 Table 1. 3 Quality Assessment Scores for cross-sectional studies 82 Table 1.4 Quality Assessment Scores for case-control studies 83 Table 1. 5 Quality Assessment Scores for cohort studies/Randomized Clinical Trials 84 Table 1.6 Quality Assessment Scores for Experimental Studies 85 Table 1.7 Sleep Depth and Cognitive/Alzheimer’s disease measures: Descriptive Study Characteristics and Main Findings 86 Table 1.8 Sleep-wake cycle abnormalities and Cognitive/Alzheimer’s disease measures: Descriptive Study Characteristics and Main Findings 95 Table 1.9 Sleep Disordered Breathing/Sleep Apnea and Cognitive/Alzheimer’s disease measures: Descriptive Study Characteristics, and Main Findings 101 Table 1.10 Insomnia and Cognitive/Alzheimer’s disease measures: Descriptive Study Characteristics and Main Findings 105 Table 1.11 Restless Leg Syndrome (RLS) and Cognitive/Alzheimer’s disease measures: Descriptive Study Characteristics and Main Findings 106 Table 2.1 Characteristics of included studies. Meta-analysis of sleep, cognitive decline and Alzheimer's disease 137 Table 2. 2 Pooled relative risks from sub-group meta-analysis for the effect of sleep on Cognitive decline and/or Alzheimer's disease 142 Table 2.3 Results of meta-regression analysis examining potential effects of different factors on the natural logarithm of the odds ratio between sleep and cognitive decline and/or Alzheimer's disease 144 Table 1A Main findings from included studies and comments regarding computed conversions of the different indices to a common index: odds ratios and regarding computing a summary effect size of the impact of sleep problems on AD 151 Table 2A Formula used for converting among effect sizes and for calculating the variance of

Page 9: Omonigho A. Michael Bubu

vi

the effect sizes 157 Table 3A Formula used for computing the variance of a composite or a difference 158

Table 3.1 Descriptive Characteristics of Participants by Obstructive Sleep Apnea Status at Baseline 181 Table 3.2 Covariance Parameter estimates and Between Subject variation in AD Biomarker Deposition 182 Table 4.1 Literature Search Terms for the Systematic Review of Obstructive Sleep Apnea, Cognition and Alzheimer’s disease 233 Table 4.2 Descriptive Study Characteristics and Main Findings: Obstructive Sleep Apnea and Cognition (Cross-sectional studies) 234 Table 4.3 Descriptive Study Characteristics and Main Findings: Obstructive Sleep Apnea and Cognitive Decline (Longitudinal studies) 241 Table 4.4 Descriptive Study Characteristics and Main Findings: Obstructive Sleep Apnea, and Alzheimer’s disease/AD Pathology 243

Page 10: Omonigho A. Michael Bubu

vii

LIST OF FIGURES Figure 1.1 Flow Diagram of Systematic Review of Sleep and Alzheimer’s disease 107

Figure 2.1 Study retrieval and selection for effects of sleep on cognitive decline and/or Alzheimer’s disease meta-analysis 145 Figure 2.2 Forest plot presenting overall meta-analysis based on risk estimates for the effect of sleep on cognitive decline and/or Alzheimer’s disease 146 Figure 2.3 Forest plot presenting sub-group meta-analysis based on risk estimates for the effect of sleep on Cognitive decline, Pre-clinical and Symptomatic Alzheimer’s disease 147 Figure 2.4 Trim and fill funnel plot for effects of sleep on cognitive decline and/or Alzheimer’s Disease meta-analysis 148 Figure 2.5 Forest plot presenting sub-group meta-analysis based on risk estimates for the effect of different sleep problems and disorders on cognitive decline and/or Alzheimer’s Disease 149 Figure 2.6 Forest plot presenting sub-group meta-analysis based on risk estimates for the effect of self-report versus actigraphic data of poor sleep on cognitive decline and/or Alzheimer’s disease 150 Figure 1A Forest plot presenting sub-group meta-analysis based on continent in which study was published for the effect of sleep problems and disorders on cognitive decline and/or Alzheimer’s disease 159 Figure 2A: Forest plot presenting sub-group meta-analysis based on day versus night time sleepiness for the effect of sleep problems and disorders on cognitive decline and/or Alzheimer’s disease 160 Figure 3A Forest plot presenting sub-group meta-analysis based on study temporality for the effect of sleep problems and disorders on cognitive decline and/or Alzheimer’s Disease 161 Figure 4A Forest plot presenting meta-analysis risk estimates based on omitting one study for the effect of sleep problems and disorders on cognitive decline and/or Alzheimer’s Disease 162 Figure 3.1 Baseline Alzheimer Disease CSF Biomarker burden and Brain Aβ-42 levels in Cognitive Normal subjects by OSA status 184 Figure 3.2 Baseline Alzheimer Disease CSF Biomarker burden and Brain Aβ-42 levels in Mild Cognitive Impairment subjects by OSA status 185

Page 11: Omonigho A. Michael Bubu

viii

Figure 3.3 Baseline Alzheimer Disease CSF Biomarker burden and Brain Aβ-42 levels in Alzheimer Disease subjects by OSA status 186 Figure 4.1 Study retrieval and selection for Obstructive Sleep Apnea, Cognitive Decline and/or Alzheimer’s disease Review 246

Page 12: Omonigho A. Michael Bubu

ix

ABSTRACT

This dissertation is a critical examination of the relationship between sleep problems and/or

disorders, particularly Obstructive Sleep Apnea (OSA) and Alzheimer Disease (AD). First, I conducted an

exhaustive systematic review of existing literature, and identified gaps in research that led to specific

research aims. For the first aim, I conducted the first ever-published meta-analysis examining sleep,

cognitive decline and AD, providing an aggregate effect of sleep on AD. Second, focusing on OSA, I

conducted a study examining OSA’s effect on longitudinal changes on AD biomarkers in cognitive

normal, MCI and AD subjects, using data from the Alzheimer Disease Neuroimaging Initiative (ADNI).

Lastly, I conducted a review, integrating over 3 decades of research examining OSA and cognition; OSA

and subsequent cognitive decline; and OSA and AD; with particular focus in appreciating the

heterogeneity of OSA and its outcomes in distinct age groups.

Results and implications from my research indicate that ample evidence exists linking sleep

impairments and circadian regulating mechanisms directly to clinical symptoms in AD. Sleep problems

and/or disorders increases your risk of cognitive decline and AD. OSA is associated with increased AD

biomarker burden over time, and effects longitudinal changes in these biomarkers, such that OSA subjects

progress faster than non-OSA subjects do. OSA may be age-dependent in older adults (60 – 70 years old)

and the elderly (70 years and above) and is associated with neurodegenerative diseases particularly,

cognitive decline and AD. Intermittent hypoxia and sleep fragmentation are two main processes by which

OSA induces neurodegenerative changes. Therefore, clinical interventions aimed at OSA, such as

treatment with CPAP or dental appliances, in cognitive normal and MCI patients, could possibly slow the

progression of cognitive impairment to AD.

Page 13: Omonigho A. Michael Bubu

1

SECTION 1

SYSTEMATIC REVIEW: SLEEP AND ALZHEIMER’S DISEASE

Note to the Reader:

Portions of this section have been previously published in SLEEP journal, 2017, Jan 1; 40(1). doi:

10.1093/sleep/zsw032 and have been reproduced with permission from Oxford University Press.

ABSTRACT

Background/Objective: Alzheimer’s disease (AD) is a neurodegenerative disorder with expected

incidence of approximately a million people each year, and a total estimated prevalence of 11 to 16

million people by 2050 in the United States. Mounting evidence implicates sleep as one of the risk factors

for AD with an association between sleep disruption, cognitive ability, and neurodegenerative disease.

We systematically reviewed all available studies examining any association and/or relationship and/or

biologic plausibility between sleep and its related problems/disorders and Alzheimer’s disease, cognitive

decline or dementia and evaluated the evidence for a causal association.

Methods: Original published literature assessing any association of sleep and its related

problems/disorders with AD, cognitive decline or dementia was identified by searching four bibliographic

databases, namely PubMed, Embase, Web of Science, and Cochrane library (i.e., the Cochrane Central

Register of Controlled Trials). Searches on all databases were from the onset of the archives until and

including November, 2014. The terms Alzheimer, Mild Cognitive Impairment, Sleep, Sleep Disorders

and Circadian Rhythm were identified as MeSH terms. Other MeSH search headings related to sleep and

various study types including cross-sectional, case control, cohort (retrospective and prospective),

Randomized Clinical Trials (RCTs) were also identified. Articles identified from the search were first

Page 14: Omonigho A. Michael Bubu

2

screened using titles and abstracts of the publications and eligible articles for this review had to meet

certain inclusion and exclusion criteria.

Results: Seventy-two publications that met the selection and final inclusion criteria were identified by the

literature search: 18 studies and 12 studies used a cross-sectional analysis and case-control design

respectively, to examine the association of sleep alterations/disturbances, circadian rhythm abnormalities,

and/or sleep disorders with AD, cognitive decline and/or dementia; 22 studies were prospective studies

examining the relationship between sleep and cognitive outcomes; 4 RCTs examined cognitive effects of

treating sleep and its related problems/disorders in AD; and 16 experimental studies examined the

associations between sleep and AD pathology and/or AD biomarkers. Altogether, there is substantial

evidence providing support that sleep is associated with cognitive decline and/or AD/AD pathology.

Prospective studies provide support that sleep problems precede the onset of cognitive impairment,

including AD and dementia, and experimental studies provide the most compelling evidence of the

plausibility of causal associations between sleep deprivation and AD pathology and/or AD biomarkers.

Conclusion: There is growing experimental and epidemiological evidence for a reciprocal interaction

between cognitive decline and sleep alteration. Ample evidence exists linking sleep impairments and

circadian regulating mechanisms directly to clinical symptoms in AD. However, long-term longitudinal

studies are needed to determine the relationship of chronic sleep deprivation, circadian rhythm/sleep-

wake abnormalities, sleep-disordered breathing and sleep disorders with cognitive decline.

Page 15: Omonigho A. Michael Bubu

3

INTRODUCTION

Alzheimer’s disease (AD) is a neurodegenerative disorder with an incidence in the United States

expected by 2050 to be of a million new cases yearly, and a corresponding prevalence of 11 to 16 million

people.1 In 2013, approximately 5.2 million Americans had Alzheimer’s Disease (AD), including an

estimated 5 million age 65 and older.2 AD and other dementias cause significant morbidity and mortality

to those afflicted, with an enormous socio-economic impact: an estimated US$604 billion worldwide,

constituting about 1% of the aggregated global gross domestic product.3 Preventive and/or therapeutic

measures targeted at delaying or curing AD are therefore imperative.4 Advancing age is the most

significant risk factor for AD with most individuals with AD diagnosed at age 65 or older. Various risk

factors for AD including family history,5-7 Apolipoprotein E epsilon4 allele (APOE-ε4 gene),8,9

smoking,10-12 diabetes,13-15 obesity in midlife,16-20 high cholesterol in midlife,17,21 hypertension in

midlife,22-24 physical activity,25-28 education,29,30 head circumference,31,32 head trauma,33,34 brain reserve35

and social and cognitive engagement,36-38 have been identified in epidemiological studies and additional

risk factors are being investigated.39 Mounting evidence implicates sleep as one of these factors.

Sleep and Normal Aging

Structural alterations of sleep, including sleep maintenance difficulty, sleep latency increase,

frequent nighttime and early morning awakenings, occur as part of the normal aging process.40 Various

sleep stages also show some level of alteration with increasing age including a reduction in slow wave

sleep (SWS) and a compensatory increase in sleep stages 1 and 2.40 Rapid-eye movement (REM) sleep

changes are less distinct and seem to appear later with increasing age.41,42 Microstructural alterations such

as decrease in K complexes and sleep spindles are also observed.43 Apart from these structural alterations

of sleep, sleep-wake rhythm disturbances have also been described in the elderly. Greater tendency for

daytime sleep and increased propensity to phase advancing are observed frequently.41 Circadian sleep-

wake rhythm disorders do not result from behavioral factors alone but also from neuro-hormonal

modifications, especially melatonin whose plasma concentration declines with age.44 Psychiatric and

Page 16: Omonigho A. Michael Bubu

4

somatic pathologies, pharmacological treatments, decrease in physical activity and exposure to light also

play a role in the origin of sleep disturbances in normal aging.40 Sleep disorders such as sleep-related

breathing disorders (SRBD), restless legs syndrome (RLS) and periodic limb movements in sleep (PLMS)

also increase significantly with age.41

Sleep and Demented older adults

Demented older adults exhibit significant sleep disturbance and degenerate sleep patterns,

including shorter sleep duration and fragmented sleep,43,45-47 altered circadian rest/activity patterns,45 and

elevated rates of sleep disordered breathing (SDB).48,49 It is estimated that about 45% of AD patients have

sleep disturbances.50,51 When symptoms appear at an early stage of the disease, they are frequently

associated with a more severe cognitive decline.50,52 Both circadian rhythm disruptions and sleep

disturbances have a major impact on the quality of life of patients and their caregivers.53-56 They have

been conventionally thought of as complications of various psychiatric co-morbidities50,57 secondary to

disease pathology. However, researchers have begun to examine whether sleep and its related problems

contribute to the risk of AD.58 Sources of sleep disturbance in AD are thought to include degeneration of

neural pathways that regulate sleep-wake patterns and sleep architecture.50,57 Sleep disorders may also

exacerbate cognitive symptoms through impairment of sleep-dependent memory consolidation

processes.59,60 Interestingly, circadian and sleep disorders may have a causal role in the pathophysiology

of AD, and could be critical to the goal of preventing AD, because effective interventions exist to

improve sleep.61

Previous narrative reviews

There are few narrative review articles,40,62-64 that have examined this association with focus on

different aspects of sleep disturbances and/or neurologic diseases. Palma et al.,62 focusing on sleep

deprivation, reviewed how sleep loss constitutes a risk factor for various neurologic diseases including

AD. In discussing sleep loss and AD, they concluded that it was necessary to distinguish between the

Page 17: Omonigho A. Michael Bubu

5

possibility that the risk of AD is influenced by changes in rest activity patterns and sleep-wake cycles,

through their effects on underlying neurodegenerative processes or the alternative explanation that rest

activity and sleep-wake patterns are just markers of a subclinical neurodegenerative process. Slats et al.,63

focusing on the role of hypocretin and melatonin, described the impairments of both sleep regulating

systems and circadian rhythms in AD and their link to clinical symptoms. They further discussed how

sleep regulating systems, especially neurotransmitters such as melatonin and hypocretin, could affect AD

pathophysiology. They concluded that there is substantial evidence of impairments in both sleep and

circadian regulating mechanisms in AD pathophysiology, which may be linked directly to clinical

symptoms in AD patients. Peter-Derex et al.,40 did a thorough clinical review of sleep and Alzheimer’s

disease, observing that micro-architectural sleep alterations, nocturnal sleep fragmentation, decrease in

nocturnal sleep duration, diurnal napping and inversion of the sleep-wake cycle are the main disorders

observed in patients with AD. Further, they noted that there is growing experimental and epidemiological

evidence for a close reciprocal interaction between cognitive decline and sleep alterations.

Objective of this systematic review

To the best of our knowledge, there are no systematic review articles on sleep and AD or

Dementia. In this context, we conducted a broad systematic review of all available studies examining any

association of sleep and its related problems/disorders with Alzheimer’s disease, cognitive decline or

dementia, and evaluated the evidence for a causal association. Recommendations for future directions of

research are also proposed.

METHODS

We conducted this systematic review in accordance with the exemplified step-by-step guide for

systematic reviews and meta-analysis by Pai et al.,65 and for the purpose of reporting, we adhered to the

Preferred Reporting Items for Systematic Reviews and Meta-Analyses: The PRISMA Statement by

Moher et al.66 We also consulted the Cochrane handbook for systematic reviews of interventions.67

Page 18: Omonigho A. Michael Bubu

6

Identification of Eligible Studies

Electronic search

An extremely sensitive search strategy was developed and assisted with identification of all

eligible published articles in scientific (medical, psychological and epidemiological) journals for all years

up to November 2014. The electronic search strategy combined terms characterizing AD/AD

pathogenesis, as the outcome variable, Sleep, as the exposure variable, and a third set specified various

study types. All terms within each set were combined with the Boolean operator OR, and then all three

sets were then combined using AND.

Original published literature assessing associations of sleep and its related problems/disorders

with Alzheimer’s disease, cognitive decline or dementia were identified by searching four bibliographic

databases on November 21, 2014, namely:

• Medline (through PubMed; all years 1946 - present)

• Embase (through www.embase.com by Elsevier B.V. 2011; all years from 1947 - present)

• Web of ScienceTM (Thomson Reuters 2014; all years from 1900 – present)

• Cochrane library (i.e., the Cochrane Central Register of Controlled Trials; all years from 1900 -

present).

The terms Alzheimer, Mild Cognitive Impairment, Sleep, Sleep Disorders and Circadian Rhythm were

identified as MeSH terms. Other MeSH search headings related to sleep included sleep loss, sleep

deprivation, sleep apnea, sleep disruption, excessive daytime sleepiness, Obstructive Sleep Apnea (OSA),

Sleep-Disordered Breathing, Rapid Eye Movement Behavioral Disorder, melatonin, and hypocretin.

Search terms related to Alzheimer’s disease and AD pathogenesis included amyloid beta, amyloid

cascade hypothesis, Amyloid Precursor Protein (APP) and APOE-ε4. MeSH terms associated with

various study types included cross-sectional, case control, cohort (retrospective and prospective),

Randomized Clinical Trials (RCTs), literature reviews, systematic reviews, and ecological studies.

Page 19: Omonigho A. Michael Bubu

7

Articles written in other languages other than English were also considered and screened with the help of

Google translator.

In PubMed, a search using Alzheimer’s disease and AD pathogenesis MeSH terms yielded

136,119 articles; and a search using sleep and its related problems/disorders MeSH terms yielded 185,925

articles. The combination of the Alzheimer’s disease and AD pathogenesis MeSH terms and sleep and its

related problems/disorders MeSH with MeSH terms associated with various study types, yielded a total of

904 publications. The PubMed search was adapted to searching the other databases. Table 1.1 provides a

list of the search terms used. The complete search strategy including the saved search histories in the

various databases is available on request.

Searching other sources

To locate additional relevant studies, the reference lists of articles identified through database

searches and the Bibliographies of narrative review articles were examined. We also hand-searched the

previous year’s issues (November 2013 to November 2014) of high ranking journals that published more

than one relevant identified article during the initial search, including Sleep Medicine Reviews,

Neurology, JAMA (Journal of American Medical Association), Sleep Medicine, American Journal of

Alzheimer’s Disease and other Dementias, Sleep, Journal of Sleep Research, Journal of clinical sleep

medicine, Journal of Alzheimer’s Disease, Brain, and Neurobiology of Aging.

Selection criteria

Type of study

Original articles from observational studies (cross-sectional, case-control and

retrospective/prospective cohort), Randomized Controlled Trials (RCT), and Animal Experimental

studies were included in this systematic review. Articles identified in the search were first screened using

titles and abstracts of the publications. Eligible studies for this review had to meet the following final

study inclusion criteria: (1) They reported a primary or secondary data analysis using an appropriate

Page 20: Omonigho A. Michael Bubu

8

epidemiologic study design of any association of sleep and its related problems/disorders with

Alzheimer’s disease, cognitive decline or dementia; (2) The diagnosis of Alzheimer's

disease/dementia/cognitive decline was based on a self-reported diagnosis, on meeting standardized

criteria (e.g. International Classification of Disease-9 (ICD -9), Diagnostic Statistical Manual-IV (DSM-

IV)) or objective measures of Alzheimer pathology (e.g., Positron Emission Tomography (PET) scan); (3)

The finding of sleep and its related abnormalities/disorders was based on self-reported diagnosis or

standardized criteria/questionnaires (e.g., PQSI Pittsburg Sleep Quality Index (PQSI),68 Epworth

Sleepiness Scale (ESS)) or objective measures (e.g., wrist actigraphy69 or polysomnography (PSG)), (4)

Inclusion of a comparison group with or without varying levels of sleep disturbances/disorders or

cognitive decline/dementia/AD, whichever is applicable, and (5) The study provided a valid measure of

association between sleep and its related problems/disorders and Alzheimer’s disease, cognitive decline

or dementia with 95% confidence intervals and/or p-values. Exclusion criteria included the following: (1)

case series/reports, systematic and/or literature reviews, abstracts or editorials, (2) articles that did not

examine an association between sleep and its related problems/disorders and Alzheimer’s disease,

cognitive decline or dementia, and (2) any RCT that did not examine the cognitive effects of treating

sleep and its related problems/disorders in AD.

Sleep abnormalities and/or disorders

Sleep abnormality and/or disorder was considered as the risk factor of interest in this systematic

review. The international Classification of Sleep Disorders (ICSD-2)70 aptly describes and classifies sleep

disorders, however, previous studies71,72 examining the association between sleep and workplace injuries

made use of various concepts to define sleep problems in order to fit their study design. Accordingly, in

this systematic review, we classified sleep based on the measured sleep parameter and eligible studies in

this review were grouped on this basis. Sleep parameters including duration, quality, efficiency and/or

disturbance were classified as sleep depth problems; circadian rhythm abnormalities, and excessive

daytime sleepiness (EDS) were classified as sleep-wake cycle abnormalities; the apnea hypopnea index

Page 21: Omonigho A. Michael Bubu

9

(AHI) that measures hypoxia levels in sleep was used to refer to Sleep Disordered Breathing (SDB),

Sleep Apnea (SA) and/or Obstructive Sleep Apnea Syndrome (OSAS); and lastly other sleep disorders

referred to insomnia and Restless Leg Syndrome (RLS). Studies that examined multiple sleep parameters

e.g., circadian rhythm abnormalities and Insomnia, were reviewed under the various sleep categories,

however, were still counted as one study.

Cognitive decline and/or Alzheimer’s disease Dementia

The outcome of interest in this review was Cognitive Decline and/or Alzheimer’s disease.

Eligible studies were expected to have diagnosed cognitive decline and/or Alzheimer's disease based on a

self-reported diagnosis that met standardized criteria (e.g. International Classification of Disease-9 (ICD -

9), Diagnostic Statistical Manual-IV (DSM-IV)) or on objective measures of Alzheimer pathology (e.g.,

Positron Emission Tomography (PET) scan). AD is the most common form of Dementia accounting for

up to 50-80% of dementia cases,1 as such studies that had dementia as an outcome were also considered.

Studies that solely looked at specific forms of dementia other than AD e.g., vascular, Lewy body,

frontotemporal dementia were excluded.

Data Extraction

An independent assessment of all the titles and abstracts of all eligible study identified by the

search strategy was performed by two authors (OB, SS) using EndNote X6. Potential articles for the study

were retrieved and examined for possible inclusion by the same two authors. Discordances that were

unresolved in unison were referred to a third author (AB). Excel spreadsheet and extraction sheets created

in Access (Microsoft Office version 2010) was then used for the data extraction. This was independently

done by two reviewers (OB, JM), and extracted categories included the study design, study population,

exposure assessment, outcome assessment, statistical methods and tests used, covariates as well as the

main results of the study. Differences in the information extracted were resolved by a third author (MD)

Reviewers were not blinded to the funding, authors, or institutions.

Page 22: Omonigho A. Michael Bubu

10

Assessment of study quality

Study quality was assessed using an adaptation from previous systematic reviews73,74 and the

modified version of the Newcastle-Ottawa scale (NOS) for quality assessment of the observational

studies,75,76 with addition of new items relevant to this review (Table 1.2). Two review authors (OB, JM)

assessed each included study separately. Any disagreement between the two review authors was settled

by consensus, or where necessary, by a third party (YW). Parameters used for the quality assessment

included well specified hypothesis, study design type, appropriately described sample population, sample

size, definition of sleep and its related abnormalities, definition of Alzheimer's disease/dementia/cognitive

decline, statistical analytical methods, and inclusion or approach used to adjust for potential confounders.

The quality rating scale was adapted to specific study design, and each category had specific maximum

number of awarded stars (Tables 1.3 – 1.6). Quality scores were awarded such that the minimum number

of stars a study can get was 5. For cross-sectional studies, 16 stars was the maximum possible; for case-

control, cohort studies and RCTs 20 stars was the maximum possible, and for animal experimental studies

15 stars was the maximum possible. Quality indicators of most significance for this review (given the

highest weights) included appropriate definition of sleep and its related abnormalities, study population

and adjustment for potential confounders (such as age, years of education, marital status, forced

expiratory volume (FEV) in one second based on spirometry, self-reported history of physician’s

diagnosis of asthma, number of hours of sleep and napping, and use of benzodiazepines, Chronic

Obstructive Pulmonary Disease (COPD), nonfatal coronary heart disease including myocardial infarction,

stroke, the ankle-brachial index, Apolipoprotein E-epsilon4 (APOE-ε4) genotype, Body Mass Index

(BMI) and Depression). Studies were divided into three categories: low quality (< 50% of the maximum

number of stars), medium quality (≈ 55-70% of the maximum number of stars), and high quality (75% or

more of the maximum number of stars) (Tables 1.3 – 1.6).

Page 23: Omonigho A. Michael Bubu

11

Risk of Bias in Individual Studies

Studies were thoroughly reviewed and the risk of bias within individual studies was assessed by

examining the appropriateness of the study design as it relates to the study hypothesis, how each study

defined and/or ascertained their exposure and/or outcome, and whether appropriate comparison was done

with the unexposed participants. Appropriate identification and/or selection of study participants to

minimize selection bias risk was also important. It was essential that diagnoses were properly assessed

and/or validated, and that studies reported which diagnosis or symptomatology was utilized, and if using

various databases, how overlapping data was handled. Study data were also assessed to determine if

completeness and potential publication/reporting bias could be evaluated.

Observational data from studies

Evidence from the reviewed studies was harmonized and integrated using a narrative synthesis77

and reported data is presented making use of accepted and recognized conventional guidelines78

RESULTS

Study Selection

A total of 2341 publications were identified doing a literature search in PubMed, Embase, Web of

Science, and Cochrane library (i.e., the Cochrane Central Register of Controlled Trials), with each

yielding 904, 1026, 305 and 106 publications respectively (Figure 1.1). One thousand, one hundred and

twenty duplicates were identified and removed of which 90% were from PubMed, and Embase. After

duplicates were removed, 1221 publications were screened for titles resulting in the exclusion of 180

publications because titles did not reflect an examination of an association between sleep and Alzheimer’s

disease. The remaining 1041 publications were screened using the published abstracts resulting in the

exclusion of 840 publications that were removed because they were either case series/reports or were

systematic and/or literature reviews, abstracts or editorials. Two hundred and one full text articles were

assessed for eligibility and 130 publications were further excluded, of which 105 publications identified

Page 24: Omonigho A. Michael Bubu

12

were excluded because they did not address an association between sleep and its related

problems/disorders and Alzheimer’s disease, cognitive decline or dementia or did not estimated a measure

of association that included 95% confidence intervals or p-values. Twenty-five RCTs were excluded

because they did not examine cognitive effects of treating sleep and its related problems/disorders in AD.

One study was included from a search of the reference lists of articles identified through database

searches. Seventy-two articles met all eligibility criteria.

Study Characteristics (Design and Quality)

Among the 72 studies identified, 18 studies51,79-95 and 12 studies96-107 used a cross-sectional

analysis and case-control design respectively, to examine the association of sleep alterations/disturbances,

circadian rhythm abnormalities, and/or sleep disorders with AD, cognitive decline and/or dementia; 22

studies108-129 were prospective studies examining the relationship between sleep and cognitive outcomes;

4 RCTs130-133 examined cognitive effects of treating sleep and its related problems/disorders in AD; and

16 experimental studies134-149 examined the associations between sleep and AD pathology and/or AD

biomarkers (Table 3). One110 of the prospective studies examining the relationship between sleep and

cognitive outcomes was in Polish; the remaining studies were in English. In total 36 studies were assessed

to be of high quality (16 experimental studies,134-149 3 RCTs,130,132,133 14 prospective studies,109,111,112,114-

116,118,119,121,122,124,126-128 and 3 cross-sectional studies79,83,86); 22 studies were assessed to be of medium

quality (1 RCT,131 6 prospective studies,108,117,120,123,125,129 7 case-control studies,97-99,101,105-107 and 8 cross-

sectional studies51,84,85,88-90,93,94); and 14 studies were assessed to be of low quality (2 prospective

studies,110,150 5 case-control96,100,102-104 and 7 cross-sectional studies80-82,87,91,92,95) (Table 1.3 – 1.6). Type of

study, selection bias related to sampling, measurements of sleep and/or AD solely based on self-reporting,

and insufficient adjustment for core confounders, were the main shortfalls.

Page 25: Omonigho A. Michael Bubu

13

Participants and setting

The 72 studies included in the systematic review were published between 1998 and 2014. Human

participants of the included studies were middle aged and elderly adults of both sexes with age ranging

from 40 – 91 years cutting across North America (38 studies), Europe (22 studies), Australia (1 study),

Asia (7 studies) and collaborations between continents (5 studies). Participants were drawn from settings

that included communities (32 studies), hospital/patient population (20 studies), institutions e.g., nursing

home residents (4 studies), and brain autopsies (1 study). In total, 50 studies included both sexes, 6

studies were based on females, and three studies were based on males only. Fifteen experimental studies

that made use of AD transgenic mice were also included.

Sleep abnormalities and/or disorders

There was a considerable amount of variation in the methods used by the studies to assess for

sleep problems. Most studies (n=35) utilized objective measures (e.g., wrist actigraphy69 or

polysomnography (PSG), while 11 studies utilized standardized questionnaires including the Pittsburg

Sleep Quality Index (PQSI),68 Epworth Sleepiness Scale (ESS), Berlin questionnaire,151 Behavioral

pathology in AD rating scale, the BEHAVE-AD,152 Women’s Health Initiative Insomnia Rating Scale

(WHIIRS),153 and the International RLS Study Group questionnaire.154,155 Only five studies utilized self-

constructed questionnaires while 4 studies made use of Fluorescence Immuno Assays (FIA) to determine

melatonin/hypocretin concentrations. The experimental studies (n=16) conducted cell culture, transfection

and hypoxic treatment tests or utilized immuno-assays, Enzyme Linked Immuno-sorbent Assay (ELISA),

Western Blot tests to determine melatonin concentrations and/or forced experimental mice into states of

wakefulness. In total, 30 studies (i.e., 10 cross-sectional,51,79,80,82,84,85,88,92-94 5 case-control,96,98-100,105 9

prospective cohort,109-111,116,118,120,122,125,126 1 retrospective cohort,123 4 experimental studies136,138,142 and 1

RCT133) examined sleep depth parameters, 23 studies examined sleep-wake /circadian rhythm

abnormalities (i.e., 5 cross-sectional,87,89,91,95,102 4 case-control,97,101,106,107 6 prospective

cohort,108,118,119,124,127,129 7 experimental studies134,135,137,140,143-145 and 1 RCT131), 16 studies examined

Page 26: Omonigho A. Michael Bubu

14

hypoxia states (AHI) in sleep apnea/sleep disordered breathing (i.e., 4 cross-sectional,81,83,86,90 1 case-

control,103 3 prospective cohort,125,128,150 1 retrospective cohort,112 2 RCTs,130,132 and 5 experimental

studies139,141,147-149), 6 studies (i.e., 2 cross-sectional87,92 and 4 prospective cohort studies114,115,117,121)

examined sleep as insomnia and 3 studies (i.e., 2 cross-sectional84,92 and 1 case-control104) examined

sleep as RLS.

Cognitive decline and/or Alzheimer’s disease Dementia

There was also great variability in the methods used to diagnose cognitive decline and/or

Alzheimer’s disease Dementia. Most studies made use of self-reported diagnosis that met standardized

criteria (e.g. International Classification of Disease-9 (ICD -9),156 Diagnostic Statistical Manual-IV

Revised Text (DSM-IV TR),157 National Institute of Neurological and Communicative Disorders and

Stroke/Alzheimer’s Disease and Related Disorders Association (NINCS–ARDRA).158 Mild Cognitive

Impairment (MCI) was mostly diagnosed using the Petersen criteria. 159 Cognitive function was assessed

by a battery of neuropsychological tests (mostly with the Mini Mental State Examination (MMSE), 160

California Verbal Learning Test (short form),161 Rey-Osterrieth complex figure,162 Verbal Fluency,

Wechsler Adult Intelligence Score (digit symbol, digit span).163 Emotional state was mostly assessed with

the Geriatric Depression Scale164 and functioning in daily living with Functional Activities

Questionnaire,165 and dementia severity was mostly assessed using the Clinical Dementia Rating Scale

(CDR). Objective measures of Alzheimer pathology (e.g., Positron Emission Tomography (PET) scan for

amyloid load (Aβ), Polymerase Chain reaction (PCR) for APOE genotyping), were also done.

Study findings

A. Sleep depth and cognitive decline and/or Alzheimer’s disease dementia

In total, 30 studies (i.e., 10 cross-sectional,51,79,80,82,84,85,88,92-94 5 case-control,96,98-100,105 9

prospective cohort,109-111,116,118,120,122,125,126 1 retrospective cohort,123 4 experimental studies136,138,142 and 1

RCT133) examined the association between sleep depth (i.e., sleep quality, duration, sleep latency, sleep

Page 27: Omonigho A. Michael Bubu

15

efficiency and/or disturbance), and cognitive decline and/or Alzheimer’s disease Dementia. Table 1.7

presents the results of the descriptive study characteristics, statistical methods and main findings of the

studies.

A.1 Cross-sectional studies

Nebes et al88 examined the association between sleep quality (i.e., sleep duration, sleep latency

and sleep efficiency) and cognitive performance while controlling for the effects of some common

confounding conditions including anticholinergic drug burden and found that sleep quality was associated

with cognition. The association was not explained by confounding factors such as cerebrovascular

disease, depression, or medication usage. Blackwell et al79 (2006) examined the association between

objectively measured fragmented sleep and decreased cognitive function in a cohort of 2,242 community-

dwelling women 65 years old or older with a baseline Cognitive Abilities Screening Instrument (CASI)

score of >=74 points. The CASI is a composite of the Hasegawa Dementia Screening Scale, the Folstein

Mini-Mental State Examination,160 and the Modified Mini-Mental State Test.166 They found that women

with <70% sleep efficiency had a 61% higher risk of cognitive impairment when compared with women

with sleep efficiency >=70%. Higher sleep latency was also associated with higher risk of cognitive

impairment as was higher Wake after Sleep Onset (WASO). There was no significant relationship for

total sleep time. Saint Martin et al93 examined the association between sleep quality, subjective

cognitive complaints, and neuropsychological performance in a cohort of 272 healthy elderly subjects

(mean age 74.8 ± 1.1 years) recruited from a population-based cross-sectional study on aging and

cardiovascular morbidity, and found that in healthy elderly subjects subjective sleep quality and duration

was not associated with subjective and objective cognitive performances, except for attention. However,

in this study, sleep duration and quality were self-reported and the authors had no information on sleep

structure and sleep fragmentation, two factors implicated in self-reported cognitive deficits.

Guarnieri et al84 described the frequency and characteristics of sleep disturbances in a large

cohort of persons with mild cognitive impairment or dementia and found that over 60% of the participants

Page 28: Omonigho A. Michael Bubu

16

had one or more sleep disturbances, almost invariably associated with each other but without any evident

and specific pattern of co-occurrence. Persons with AD and those with MCI had the same frequency of

any sleep disorder. This study suffered from considerable selection bias issues as patients were enrolled

consecutively and selected for specific characteristics including the presence and type of sleep disorders.

Moran et al51 using 215 attendees at a memory clinic who were diagnosed with AD found the prevalence

of sleep disturbance in the sample to be 24.5%. Sleep disturbance in AD patients was also associated with

other behavioral symptoms, notably aggressiveness. Pistacchi et al92 enrolled 236 patients (78 men and

158 women) with different subtypes of dementia and investigated the frequency and characteristics of

sleep disorders in the sample. Patients with MCI had a frequency of sleep disturbances of any type equal

to that of patients with AD. Fetveit et al82 aimed to describe the characteristics of 24-hour sleep patterns

in nursing home patients with dementia and to determine whether various degrees of dementia have any

influence on total sleep duration. They found that sleep duration was positively correlated with the

severity of dementia in nursing home patients.

Studies that examine sleep associations with Alzheimer’s disease pathology or Alzheimer’s

disease biomarkers are extremely important to help clarify and consolidate efforts at establishing that AD

pathology might link poor sleep to cognitive outcomes. Since AD biomarkers are evident early in the

Alzheimer’s disease process, such findings might help foster directed preventive efforts at a time when

prevention may still be feasible.167 Spira et al94 examined the association between self-reported sleep

variables and Aβ deposition in 70 community-dwelling older adults who were part of the Baltimore

Longitudinal Study of Aging cohort. Participants underwent [11C]-Pittsburgh compound B PET (PiB

PET) amyloid imaging and completed self-report sleep measures. The investigators found that self-

reported shorter sleep duration and poorer sleep quality both were associated with greater Aβ burden

according to a cortical (i.e. global) measure of PiB uptake, and a measure of uptake in the precuneus – a

region in which Aβ aggregates early in the Alzheimer’s disease course.168 Ju et al85 determined whether

Aβ deposition in preclinical AD, prior to the appearance of cognitive impairment, is associated with

Page 29: Omonigho A. Michael Bubu

17

changes in quality or quantity of sleep in 142 cognitively normal middle-aged and older adults. They

found that amyloid deposition was associated with poorer sleep quality, specifically poorer sleep

efficiency (% time in bed that was spent asleep), compared to those without amyloid deposition. On the

contrary, sleep quantity (i.e., total sleep time) did not vary between groups. Napping also frequently was

associated with amyloid deposition.

Craig et al80 investigated the monoamine-A-oxidase promoter polymorphism that may impact

sleep/wake control by balancing the availability of serotonin, a precursor of melatonin. The authors found

that the high-activity 4-repeat allele of the monoamine-A-oxidase variable number tandem repeat

promoter polymorphism conferred increased susceptibility to sleep disturbance (p = .008). Nevertheless,

it is worth noting that allelic variations in this gene have been associated with behavioral and personality

traits in other contexts, which may explain this association.169 A quantitative sleep disturbance score was

significantly higher in the patients possessing MAO-A 4-repeat allele genotypes. APOE had no influence

on the development of an altered sleep

A.2 Case-control studies

Hita-Yañez et al98 determined whether changes in polysomnographic (PSG) sleep patterns

accompany subjective sleep complaints in patients with mild cognitive impairment (MCI), whether

meaningful changes in objective sleep physiology are predicted by self-reported sleep measures in MCI

patients, and whether incipient neurodegeneration contributes to exacerbate sleep misperception. Both

PSG and self-reported sleep measures confirmed that sleep is altered in patients with MCI. Whereas

subjective sleep responses predicted fragmentation of slow wave sleep (SWS) in healthy elderly (HE)

individuals, this relationship was not evident in MCI patients. Furthermore, patients with MCI showed

significant discrepancies in the estimation of sleep onset latency when compared with HE subjects. This

study compared PSG sleep data recorded in 1 night with subjective sleep quality over the past few months

to establish relationships between objective and subjective sleep in HE subjects and patients with MCI.

This approach implicitly assumes that PSG data obtained from one single night is representative of a

Page 30: Omonigho A. Michael Bubu

18

typical night in the past few months. PSG sleep studies were performed without previous adaptation of

participants to the sleep laboratory. As such, authors note that results might have been affected by the

first-night effect (i.e., differences observed on the first PSG sleep recording in comparison with

consecutive ones), which has previously been demonstrated to affect older patients more than younger

ones.170-172

The ApoE4 allele is a known susceptibility gene for AD,9,173-177 especially in homozygous

cases.178 In another study, Hita-Yañez et al99 determined if impaired sleep patterns appear years before

AD diagnosis and whether changes in sleep patterns parallel memory decline as well as its relationship

with the Apolipoprotein E (APOE) e4 genotype in 25 Mild Cognitive Impairment (MCI) patients (7

females, mean age: 70.5 ± 6.8 yr) and 25 healthy old (HO) volunteers (13 females, mean age: 67.1 ± 5.3

yr). The study results showed a significant shortening of rapid eye movement (REM) sleep together with

increased fragmentations of slow-wave sleep in MCI patients relative to healthy elders. Reduction of

REM sleep in MCI patients with APOE e4 was more noticeable than in e4 non-carriers. Changes in sleep

patterns were not correlated with memory performance in MCI patients. Instead, increased REM sleep

was associated with enhanced immediate recall in MCI e4 non-carriers.

Chen et al96 investigated changes in muscular activity, which may indicate a deficiency of

Acetylcholine (ACh), among patients with cognitive impairment, and recruited 9 controls without

dementia, 6 patients with mild cognitive impairment (MCI), and 6 patients with mild Alzheimer's disease

(AD). None of the participants had sleep complaints, and all AD patients were receiving cholinesterase

inhibitors. All participants underwent PSG examination using surface electromyographic (sEMG)

recording of both legs. A one-way analysis of variance (ANOVA) test was used to analyze the differences

in root mean square (rms) amplitude, peak frequency, and mean frequency among the three groups and

the results showed that all rms, mean frequencies, and peak frequencies increased significantly (p<0.05)

in MCI patients. The authors concluded that a deficiency of Ach may result in an increase sEMG activity

in MCI patients, and that because cholinesterase inhibitors are capable of suppressing sEMG activity in

Page 31: Omonigho A. Michael Bubu

19

AD patients, an increase in sEMG activity is associated with a deficiency of Ach, which could be an early

indicator of dementia. Hot et al100 examined relationships between episodic memory deficits and

electroencephalography [EEG] abnormalities occurring during sleep in patients with early AD.

Polysomnographic sleep acquisition was performed and included continuous recordings of EEG. Results

showed that relative to age-matched controls, AD patients presented faster mean theta frequency in both

REM sleep and slow wave sleep [SWS]. In AD patients, a correlative analysis revealed that faster theta

frequency during SWS was associated with better delayed episodic recall. Authors concluded that the

results suggest that changes in theta rhythm during REM and SWS are a relevant index of brain

impairments in the early stage of AD whereas sleep architecture is not, and that changes in theta activity

during SWS are related to episodic memory performance.

Sanchez-Espinosa et al105 examined associations between changes in physiological sleep and

plasma Aβ concentrations in 21 healthy old (HO) adults and 21 amnestic Mild Cognitive Impairment

(aMCI) subjects. They further assessed whether these two factors were associated with cortical volume

loss in each group. Sleep parameters were assessed with PSG. The investigators found significant

relationships between disrupted slow-wave sleep (SWS) and increased plasma levels of Aβ42 among

aMCI, but not HO subjects. They also found correlations between shortened rapid-eye movement (REM)

sleep in aMCI and thinning of the posterior cingulate, precuneus, and post-central gyrus; whereas higher

levels of Aβ40 and Aβ42 accounted for grey matter (GM) loss of posterior cingulate and entorhinal

cortex, respectively.

A.3 Cohort studies

Keage et al118 investigated the effect of self- reported sleeping behaviors and incidence of

cognitive impairment over two and 10 years in two healthy aged English populations. Napping behavior

was found to be negatively associated with incident cognitive impairment, with those who napped more

being less likely to be cognitively impaired at two years (OR = 0.38, 95% CI 0.22–0.67) and 10 years

(OR = 0.48, 95% CI 0.29–0.78). Short night time sleep duration (<= 6.5 h) increased the risk of incident

Page 32: Omonigho A. Michael Bubu

20

cognitive impairment over 10 years (OR = 2.02, 95% CI 1.17–3.48). The effect of long sleep duration (>=

9 h) failed to reach significance at both two and 10 years. The study did not account for relationships

between sleep and cognition with medication, cardiovascular factors, and metabolic diseases (other than

BMI). In addition, only cognitively healthy individuals were included at baseline so that the prognostic

effect of these sleep measures in those cognitively impaired or demented at baseline cannot be

extrapolated. Tworoger et al125 examined the association of sleep duration, snoring, and difficulty

sleeping with cognitive function in a cohort of community-dwelling women. Results showed that women

sleeping ≤5 hours/night scored worse than women sleeping 7 hours/night (mean difference on global

score combining all cognitive tests = 0.15 standard units, 95% CI: -0.28, -0.02). Women who regularly

had difficulty falling or staying asleep scored 0.11 units lower on the global score (95% CI: -0.22, 0.01)

compared with those who rarely had difficulty sleeping. These differences were equivalent to the mean

differences in score observed between participants who were 4 to 5 years apart in age. Authors found no

associations with snoring or with any of the sleep variables and cognitive decline over 2 years. In a

unique study, Virta et al126 investigated the association between midlife sleep characteristics (mean age

52 years) and late life cognitive function and dementia 18–26 years later in 2,336 members of the Finnish

Twin cohort who were at least 65 years of age, and found that reports of short (<7 h) and long (>8 h)

sleep duration, poor sleep quality, and use of hypnotic medications for more than 60 days per year were

associated with lower cognitive composite scores in old age; with long sleep being associated with

roughly 1.8 times the odds of developing AD. In this study, questionnaire data from 1981 were used in

the assessment of sleep characteristics, use of hypnotics, and covariates at baseline and between 1999 and

2007, and participants were assigned a linear cognitive score with a maximum score of 51 based on a

telephone interview (mean score 38.3, SD 6.1). Some of the associations differed by participant

characteristics, including Apolipoprotein E (APOE) e4 allele status, sex, and age, suggesting that poor

sleep may differentially affect cognition in different populations. The study revealed an association

between midlife sleep length and cognitive performance and AD based on medical records. With a mean

Page 33: Omonigho A. Michael Bubu

21

follow-up time of above 20 years, temporality can be assessed between sleep disturbance and cognitive

outcomes, thereby providing evidence of a potential causal link from poor sleep to cognitive decline.

Song et al122 examined the associations between sleep stage distributions and subsequent decline

in cognitive function in community-dwelling men aged 67 or older (n = 2,601) who were free of probable

dementia at the time of sleep evaluation. Follow-up averaged 3.4 years. Sleep stages were identified by

in-home polysomnography at the initial sleep visit. The average percent of time spent in stages of sleep

were as follows: N1, 6.7% ± 4.1%; N2, 62.6% ± 9.5%; N3, 11.3% ± 8.9%; and R, 19.4% ± 6.6%. The

authors found that increased time in Stage N1 and less time in Stage R were associated with worsening

cognitive performance in older men over time. Blackwell et al111 (2014) examined associations of

objectively- and subjectively-measured sleep with subsequent cognitive decline over 3.4 ± 0.5 y in 2,822

cognitively intact community-dwelling older men (mean baseline age 76.0 ± 5.3 y). Diminished

actigraphic sleep efficiency (i.e. spending a lower proportion of time in bed asleep), greater nighttime

wakefulness, greater number of long wake episodes, and poor self-reported sleep quality measured by a

score greater than 5 on the Pittsburgh Sleep Quality Index (PSQI) were all associated with subsequent

cognitive decline. However, there was no association between sleep duration and cognition.

Sterniczuk et al123 examined whether self-reported sleep problems in individuals who were not

demented at baseline, are early independent markers of risk for developing AD or dementia over an

approximately 4-year period. Data on four sleep variables (sleeping problems and fatigue in past 6

months, sleep medication use, and ‘recent trouble sleeping or a change in pattern’) were collected, and the

authors created a composite ‘sleep disturbance index’ from those items. The authors found a 23% greater

odds of dementia or Alzheimer’s disease among participants with higher scores on the sleep disturbance

index compared to those with lower scores after adjusting for age, sex, education, body mass index

(BMI), and cognition. Also, individual sleep items independently predicted AD or dementia and mortality

within ~4 years. Benito-León et al109 using a population-based study of 3,857 people without dementia

aged 65 years and older (NEDICES [Neurological Disorders in Central Spain]) examined whether long

sleep duration was associated with increased risk of dementia mortality. In a Cox model that adjusted for

Page 34: Omonigho A. Michael Bubu

22

numerous demographic factors and comorbidities, the hazard ratio for dementia mortality in long sleepers

was 1.63 (p = 0.03). Hahn et al116 examined a subjective report of change (reduced duration and/or

depth) in sleep pattern in relation to subsequent risk of incident all-cause dementia and Alzheimer disease

(AD) over a 9-year follow-up. Participants included 214 Swedish adults aged 75 and over who were

dementia-free both at baseline and at first follow-up (3 years later). Results showed that reduced sleep

was associated with a 75% increased all-cause dementia risk (hazard ratio: 1.75; 95% confidence interval:

1.04-2.93; Wald = 4.55, df = 1, p = 0.035) and double the risk of AD (hazard ratio: 2.01; 95% confidence

interval: 1.12-3.61; Wald = 5.47, df = 1, p = 0.019) after adjusting for age, gender, education, lifestyle and

vascular factors. However, results did not hold after adjusting for depressive symptoms, which was

measured with only one self-reported item. Findings are limited by the fact that the study did not include

objective longitudinal data on sleep. Bidzan et al110 found that patients in the preclinical period of

dementia were more likely to experience sleep disturbances and with greater intensity. The study was

conducted in Poland using 150 care centers residents age 55 and older. The presence of dementia was

excluded in the participants. After a follow-up period of 7 years, diagnosis of AD was established in 25

participants, while 111 people formed the control group.

Lim et al120 examined whether better sleep consolidation attenuates the relationship of the APOE

genotype to the risk of incident AD and the burden of AD pathology, in 698 community-dwelling older

adults without dementia (mean age, 81.7 years; 77% women) of the Rush Memory and Aging Project.

Participants were followed for up to 6 years. They found that better sleep consolidation attenuated the

effect of APOE genotype on incident AD and development of neurofibrillary tangle pathology, and

concluded that assessment of sleep consolidation may identify APOE+ individuals at high risk for

incident AD, and interventions to enhance sleep consolidation should be studied as potentially useful

means to reduce the risk of AD and development of neurofibrillary tangles in APOE ε4+ individuals.

A.4 Experimental studies

Mander et al142 determined whether regional brain atrophy, reduced slow wave activity (SWA)

during non–rapid eye movement (NREM) sleep and impaired long-term retention of episodic memories,

Page 35: Omonigho A. Michael Bubu

23

independently recognized features of aging, are inter-related. The authors found that “age-related medial

prefrontal cortex (mPFC) gray-matter atrophy was associated with reduced NREM SWA in older adults

and statistically mediated the impairment of overnight sleep–dependent memory retention. Moreover, this

memory impairment was further associated with persistent hippocampal activation and reduced task-

related hippocampal-prefrontal cortex functional connectivity, potentially representing impoverished

hippocampal-neocortical memory transformation”. Together, these data support a model in which age-

related mPFC atrophy diminishes SWA, the functional consequence of which is impaired long-term

memory. Such findings suggest that sleep disruption in the elderly, mediated by structural brain changes,

represents a contributing factor to age-related cognitive decline in later life. At a translational level, data

from this study endorses the possibility that improvements of SWA in older adults, through physiological,

behavioral or pharmacological means may represent a treatment target for minimizing the cognitive

decline associated with deficient long-term memory retention in later life.

Studies in AD mouse models support an association between sleep deprivation and AD

pathology. Kang et al138 in their study used in-vivo micro-dialysis in amyloid precursor protein (APP)

transgenic (Tg2576) mice (i.e. mice that overexpress mutant familial AD APP genes) and demonstrated

that both acute and chronic sleep deprivation alters brain interstitial fluid (ISF) Aβ diurnal rhythm. The

study also provided evidence of effects of hypocretin signaling on Aβ metabolism in studies of humans.

Circadian rhythm mean peak levels between 7 p.m. and 9 p.m. in human CSF Aβ levels were 28% higher

than the mean trough levels, which occurred between 9 a.m. and 11 a.m. A similar rhythm was found in

mice, with increased Aβ levels in brain interstitial fluid (ISF) while awake, compared to the sleep, both in

wild type C57BL6 mice and human APP transgenic mice, Tg2576. After being sleep deprived, mice spent

more time sleeping resulting in reductions in ISF Aβ levels. This suggests an association between

wakefulness, and increased ISF Aβ. Moreover, infusing the ventricle with a dual hypocretin receptor

antagonist resulted in suppressed ISF Aβ levels and the natural diurnal variation of Aβ was abolished. In

addition, hypocretin infusion led to significant decreases in ISF Aβ levels. The amount of ISF Aβ also

Page 36: Omonigho A. Michael Bubu

24

significantly increased during acute sleep deprivation and during orexin infusion in multiple sub-regions

of the cortex. Remarkably, systemic treatment with infusion of a dual orexin receptor antagonist once

daily for 8 weeks decreased the Aβ plaque formation in aged, sleep deprived, APP transgenic mice. This

finding suggests that Aβ diurnal rhythm is related to sleep/wake regulation and that sleep deprivation

increases Aβ levels and promotes subsequent Aβ plaque formation in APP transgenic mice. Rothman et

al146 compared male AD transgenic mice that were subjected to 6-hours/day sleep for 6-weeks to controls

(>7-hour/day sleep). Biochemical (immunoblot) outcomes were compared to mice undergoing daily cage

transfers (large cage control; LCC) as well as control mice that remained in their home cage (control;

CTL). Mice subjected to 6-hours/day sleep displayed a non-significant increase of ∼80% and a two- fold

increase of both Aβ and P-Tau levels respectively in the cortex (P <0.22). Significant positive correlations

between cortical Aβ and p -Tau levels and circulating corticosterone were seen and possibly indicate a

potential role for glucocorticoids in mediating behavioral and biochemical changes observed after sleep

restriction in a mouse model of AD. Di Meco et al136 evaluated the functional and biological

consequences on 3xTg mice that underwent 4 hours sleep restraint per day for 8 weeks. Starting at the age

of 8 months, mice were randomized to 2 groups, one underwent a 20/ 4-hour light and/or dark cycle for 2

months (4 males and 5 females), the other was kept on a normal light and/or dark cycle, as controls (5

males and 4 females). Compared to controls, behavioral assessment showed that sleep deprived mice had

a significant decline in their learning and memory. Differences in the levels of soluble amyloid-b peptides

between the groups were not observed, however reductions in tau phosphorylation, with a significant

increase in its insoluble fraction was observed. Sleep deprivation also resulted in lower levels of

postsynaptic density protein 95 and increased glial fibrillary acidic protein levels. Finally, although total

levels of the transcription factor cellular response element binding protein were unchanged, its

phosphorylated form was significantly diminished in brains of sleep-deprived mice when compared with

controls. The authors noted that their mice were relatively young in age and that the observed absence of

effect on the Aβ levels provides support for their hypothesis that sleep deprivation is not a simple

biomarker of the underlying neurodegenerative disease, but a direct contributor to its pathogenesis.

Page 37: Omonigho A. Michael Bubu

25

A.5 Randomized Controlled Trials

Ooms et al133 conducted an RCT to determine the effect of 1 night of total sleep deprivation on

cerebrospinal fluid Aβ42 protein levels in healthy middle-aged men. Participants were cognitively normal

middle-aged men (40-60 years of age) with normal sleep (n = 26) recruited from the local population.

Participants were randomized to 1 night with unrestricted sleep (n = 13) or 1 night of total sleep

deprivation (24 hours of wakefulness) (n = 13). Sleep was monitored using continuous polysomnographic

recording from 3 PM until 10 AM. Cerebrospinal fluid samples were collected using an intrathecal

catheter at defined times to compare cerebral Aβ42 concentrations between evening and morning. A night

of unrestricted sleep led to a 6% decrease in Aβ42 levels of 25.3 pg/mL (95% CI [0.94, 49.6], P = .04),

whereas sleep deprivation counteracted this decrease. A difference of 75.8 pg/mL of Aβ42 was shown

between the unrestricted sleep and sleep deprivation group (95% CI [3.4, 148.4], P = .04) when

accounting for the individual trajectories of Aβ42 over time. Individual evening and morning trajectories

of Aβ42 concentrations varied between the sleep deprivation and unrestricted sleep groups (P = .04) in

contrast to stable Aβ40, tau, and total protein levels.

B. Sleep wake-cycle/circadian rhythm and Cognitive decline and/or Alzheimer’s disease Dementia

Table 1.8 presents the results of the descriptive study characteristics, statistical methods and main

findings of studies that examined the association between sleep-wake /circadian rhythm abnormalities and

cognitive decline and/or AD. In total, there were 23 studies (i.e., 5 cross-sectional,87,89,91,95,102 4 case-

control,97,101,106,107 6 prospective cohort,108,118,119,124,127,129 7 experimental studies134,135,137,140,143-145 and 1

RCT131).

B.1 Cross-sectional studies

Oosterman et al89 examined whether nocturnal sleep and the circadian organization of the sleep-

wake rhythm had predictive value for cognitive functioning in home-dwelling elderly people and found

Page 38: Omonigho A. Michael Bubu

26

an association between the rest-activity rhythm and cognitive performance independent of age. Rest-

activity was assessed using actigraphy, and mental speed, memory, and executive function were evaluated

using a battery of standardized cognitive tests. The authors noted that majority of the subjects suffered

from at least one cardiovascular risk factor. Merlino et al87 examined the relationship between disturbed

sleep and cognitive impairment in the elderly and found that among sleep disturbances, excessive daytime

sleepiness was independently associated with the presence of dementia. In addition, the frequency of

excessive daytime sleepiness increased progressively across the different categories of cognitive decline,

as measured by means of MMSE and Global Deterioration Scale (GDS)179 scores.

Pat-Horenczyk et al91 examined differences in hour-by-hour sleep/wakefulness profiles between

severely and mild to moderately demented patients and assessed how many elderly patients remained

almost fully asleep or nearly fully awake in each hour of a 24-hour period. They found that patients with

mild to moderate dementia showed a disproportionate amount of wakefulness during the night, in contrast

to patients with severe dementia showing a disproportionate amount of sleepiness during the day. They

also found that with the progression of dementia both the capacity to maintain sleep and the capacity to

maintain wakefulness were impaired, resulting in almost complete fragmentation of sleep/wakefulness

during the night and day. Sleep/wakefulness patterns were recorded using actigraphy and levels of

dementia were assessed by the Mini-Mental State Examination (MMSE). This study suggests that sleep

disturbances parallel the severity of dementia\. However, patients were not evaluated for type of

dementia nor were records available to check whether patients had Alzheimer’s disease or dementia of

other types. Yesavage et al95 examined indices of sleep/wake function collected from individuals with

AD in relation to relevant polymorphisms in circadian clock-related genes in two cohorts of AD

participants. [Cohort 1 (n=124): individuals with probable AD recruited from the Stanford/Veterans

Affairs NIA Alzheimer’s Disease Core Center (n=81) and the Memory Disorders Clinic at the University

of Nice School of Medicine (n=43). Cohort 2 (n=176): individuals with probable AD derived from the

Alzheimer’s Disease Neuroimaging Initiative (ADNI) data set]. Adjusting for multiple tests, none of the

Page 39: Omonigho A. Michael Bubu

27

candidate gene SNPs were significantly associated with the amount of wake after sleep onset, a marker of

sleep consolidation, and no relationships likely to be of clinical relevance were detected. The authors

therefore concluded that it was unlikely that a relationship with a clinically meaningful correlation exists

between the circadian rhythm-associated SNPs and WASO in individuals with AD.

A pacemaker located in the suprachiasmatic nucleus (SCN) which serves as a master clock180,181

organizes circadian regulation of sleep and wakefulness mechanisms. Melatonin, N-acetyl-5-

methoxytryptamine, is one of the most important timing signals generated by the SCN and is mostly

known for its relation to circadian rhythms and sleep. Naismith et al102 examined whether patients with

mild cognitive impairment (MCI) had significant alterations in the timing of melatonin secretion onset

and amount as well as changes in sleep architecture. They found that patients with MCI had advanced

timing of their melatonin secretion onset relative to controls, but the levels of melatonin secreted did not

differ between groups. The MCI group also had greater wake after sleep onset (WASO) and increased

rapid eye movement sleep latency. There were differential associations between dim light melatonin onset

and cognition between the two groups, with earlier dim light melatonin onset being associated with poorer

memory performance in MCI patients. Participants’ sleep-wake behavior for the 14-nights prior to

commencing the in-laboratory portion of the protocol was assessed using actigraphy and sleep diaries.

Assessment of circadian function (melatonin rhythm timing and levels) and sleep architecture were

examined using melatonin and polysomnographic (PSG) assessments on separate nights.

B.2 Case-control studies

Two studies conducted in the Netherlands investigated the circadian rhythm of hypocretin-1

(Hypocretin-1 and -2 are hypothalamic neuropeptides, also known as orexins, that form an important

regulating system acting on the sleep–wake cycle182,183) in Alzheimer’s disease. Fronczek et al97

assessed whether the neurodegenerative process of AD affects hypothalamic hypocretin/orexin neurons.

Investigators quantified the total number of hypocretin-1 immuno-reactive neurons in postmortem

hypothalami of AD patients (n = 10) and matched controls (n = 10) and measured hypocretin-1

Page 40: Omonigho A. Michael Bubu

28

concentrations in postmortem ventricular cerebrospinal fluid (CSF) of 24 AD patients and 25 controls.

The numbers and levels of hypocretin-1 immunoreactive neurons and cerebrospinal fluid (CSF)

hypocretin-1 respectively, were significantly lower (i.e., 40% decreased cell number, and 14% lower CSF

hypocretin-1 levels respectively) in AD patients compared to controls. (p = 0.038). These findings

indicate that the hypocretin system is affected in advanced AD. Slats et al107 examined the association

between Hypocretin-1 and Amyloid-β42 CSF levels in 6 patients with AD (59-85 yrs., MMSE 16-26) and

6 healthy volunteers (64-77 yrs.). The authors could show no difference in CSF hypocretin-1 circadian

rhythm between AD and controls. However, lower mean CSF Aβ42 levels were significantly associated

with lower hypocretin-1 levels. The authors postulated that this finding might suggest a relationship

between AD pathology and hypocretin disturbance, which could hold possibilities for treatment of AD

related sleep disorders.107

Schmidt et al106 investigated cerebrospinal fluid (CSF) levels of melanin-concentrating hormone

(MCH) and hypocretin-1 (HCRT-1, orexin-A) in patients with Alzheimer’s disease (AD) and Healthy

subjects (HS) and the potentially causal relationship between these polypeptides and CSF levels of the

AD marker total Tau (T-tau), hyper phosphorylated tau (P-tau) and Ab42, cognitive performance and

behavioral symptoms in AD. The study was carried out in 33 patients with mild to severe AD and 33

subjects without any psychiatric or neurological disorder (HS). A significant main effect of diagnosis

(p=0.01) on MCH levels was found between AD and HS. MCH correlated with T-tau (p=0.01) and P-tau

(p=0.05) in the AD but not in the HS. CSF-MCH correlated negatively with MMSE scores in the AD

(p=0.05) and was increased in more severely affected patients (MMSE≤20) compared to HS (p=0.001)

and BPSD-positive patients compared to HS (p=0.05). For CSF-HCRT- 1, a significant main effect of sex

(p=0.05) with elevated levels in females was found whereas diagnosis and the sex*diagnosis interaction

were not significant. These results should stimulate additional research on the role of the hypothalamus in

AD pathogenesis and development of behavioral disturbances in AD. Future studies should include

repeated MCH/HCRT-1 level measurements in combination with sleep-wake-actigraphy and assessment

of sleep by standardized questionnaires, histochemical determinations of MCH in animal and post-

Page 41: Omonigho A. Michael Bubu

29

mortem tissues. Liu et al101 analyzed data from 85 patients with AD (mean age, 75 ± 1.1 yr) and 82 age-

matched controls (mean age, 76 ± 1.4 yr), to determine melatonin levels in postmortem cerebrospinal

fluid in relation to aging, AD, and APOE ε4 genotype. They found that melatonin levels in AD patients

expressing APOE e3/4 was significantly higher than that in patients expressing APOE e4/4 (p = 0.02).

CSF melatonin levels were significantly lower and about half of those in control subjects (p=.00126).

B.3 Cohort studies

Walsh et al127 using a study cohort of 1,287 community-dwelling older women (82.8 ± 3.1 y)

who were free of dementia at the baseline visit and were followed for 5 years, found that weaker circadian

activity rhythms (CAR) patterns were associated with lower cognitive function, especially poorer

executive function. Keage et al118 found that daytime sleepiness was significantly associated with

incident cognitive impairment over 10 years (OR = 2.43, 95% CI 1.24–4.75). Anderson et al108 also

examined the association between subjective and objective measures of sleep and wakefulness and other

health parameters in a 421 men and women, aged 87–89. The study was nested in the Newcastle 85+

Study, a population-based longitudinal study of health and ageing in the very old. Sleep-wake parameters

were assessed subjectively with the Pittsburgh Sleep Questionnaire Inventory (PSQI) and the Epworth

Sleepiness Score (ESS), while wrist actigraphy was used as an objective measure. The authors found that

impaired sleep–wake cycles were significantly associated with cognitive impairment, disability,

depression, increased number of falls, body mass index and arthritis but not with any other specific

disease markers or decreased survival. The subjective sleep questionnaires did not differentiate well

between objectively determined disturbance of sleep–wake cycles and those with normal cycles. Tranah

et al124 studied whether measured circadian activity rhythms are prospectively associated with incident

dementia or mild cognitive impairment (MCI) in 1,282 community-dwelling older women. They found

that older women with decreased circadian activity rhythm amplitude and robustness and delayed rhythms

had a higher likelihood of developing dementia or MCI when comparing those in the lowest quartile of

amplitude (OR =1.57, 95% CI, 1.09–2.25) or rhythm robustness (OR=1.57, 95%CI, 1.10–2.26) compared

Page 42: Omonigho A. Michael Bubu

30

to women in the highest quartile. An increased risk of dementia or MCI (OR=1.83, 95% CI, 1.29–2.61)

also was found for women whose timing of peak activity occurred later in the day (after 3:51PM) when

compared to those with average timing (1:34PM–3:51PM). Lim et al119 tested the hypothesis that sleep

fragmentation is associated with incident Alzheimer’s disease (AD) and the rate of cognitive decline

among 737 older adults without dementia at baseline, and reported that those with significant actigraphic

sleep fragmentation (in the 90th percentile) had a 1.5-fold increased risk of developing AD as compared

with those with low sleep fragmentation (10th percentile). A Cox regression model controlling for age,

sex, and education, also revealed that a higher level of sleep fragmentation was associated with an

increased risk of incident AD (HR = 1.22, 95%CI 1.03-1.44, P = 0.02 per 1 SD increase in sleep

fragmentation). In an earlier prospective study Yesavage et al129 determined if sleep parameters in AD

patients change over time as a function of APOE carrier status in 44 subjects, of which 25 were APOE ε4

carriers and 19 were non-ε4 carriers. Participants had been assessed at least during 2 stages of cognitive

impairment, defined as a decline from one stage of AD to another. They found that among APOE ε4

carriers, wake after sleep onset was associated with lower cognitive function; for non-ε4 subjects,

increases in WASO and declines in total sleep time, sleep efficiency, and the amplitude of the rest/activity

circadian rhythm over time were associated with lower cognitive function.

B.4 Experimental studies

Craig et al134 explored the effects of acute and chronic disruption of circadian rhythms on

memory using a phase shifting schedule that can continually challenge the rats’ circadian system by using

repeated phase shifts and recovery sessions. Twenty-four male Long Evans hooded rats (300–400 g)

obtained from Charles River (Saint-Constant, PQ) were used for all studies. They were divided into three

groups: acutely phase shifted rats (acute; n = 8), chronically phase shifted rats (chronic; n = 8), control

rats for water maze testing (control; n=7) and control rats for fear conditioning (control; n = 14). The

authors demonstrated a significant learning and memory deficit on a spatial version of the water maze

task (used for behavioral testing in the animals) in the chronically phase shifted, but not in the acutely

Page 43: Omonigho A. Michael Bubu

31

phase shifted animals. Moreover, they did not find any impairment in fear conditioning suggesting that

chronic phase shifting predominantly affects hippocampal memory. They proposed that chronic circadian

disruption may play a role in the development of age-related cognitive deficits and dementia in the

elderly. Jyoti et al137 characterized sleep, electroencephalogram (EEG) disturbances and cognitive

dysfunction in transgenic mice carrying transgenes for amyloid-protein precursor (APPswe) and

presenilin 1 (PSEN1A246E) at 5 (pre-plaque) and 20 months, relative to PSEN1 and wild-type (WT)

mice, using a novel wireless microchip device. The study findings suggested that APP/PSEN1 mice

exhibit abnormalities in activity and sleep architecture preceding amyloid plaque deposition as well as

age-related changes in cortical EEG power. Though not fully recapitulating the profile of AD patients,

this suggests activity and EEG recordings are sensitive and translational biomarkers in murine models.

Several anti-amyloidogenic effects of melatonin have been demonstrated in vitro and in animal

studies. Pappolla et al144 examined the interactive ability of melatonin with Aβ1–40 and Aβ1–42 to

determine whether melatonin inhibits the progressive formation of β-sheets and amyloid fibrils. These

investigators found that melatonin crosses the blood-brain barrier, is relatively devoid of toxicity, and

concluded that it may constitute a potential new therapeutic agent in Alzheimer’s disease. Olcese et al143

examined the potential for long-term melatonin treatment to protect Alzheimer’s transgenic mice against

cognitive impairment and development of β-amyloid (Aβ) neuropathology. Authors administered

melatonin (100 mg/L drinking water) to AD transgenic (Tg) mice from 2–2.5 months of age to their

killing at age 7.5 months. Findings revealed that melatonin suppresses Aβ aggregation in brain

homogenates. Inflammatory cytokines such as tumor necrosis factor (TNF)-α were decreased in

hippocampus (but not plasma) of Tg+ melatonin mice. Finally, the cortical mRNA expression of three

antioxidant enzymes (SOD-1, glutathione peroxidase, and catalase) was significantly reduced to non-Tg

levels by long-term melatonin treatment in Tg mice. Lahiri et al140 examined the effects of dietary

melatonin on brain levels of nitric oxide synthase, synaptic proteins and amyloid beta-peptides (Aβ) in

mice. They found that dietary melatonin supplementation led to a significant reduction in levels of toxic

Page 44: Omonigho A. Michael Bubu

32

cortical Aβ of both short and long forms that are involved in amyloid deposition and plaque formation in

Alzheimer's diseases. However, melatonin supplements did not significantly change cerebral cortical

levels of nitric oxide synthase or synaptic proteins such as synaptophysin and SNAP-25.

Deregulation of protein kinases/protein phosphatases is responsible for tau hyper

phosphorylation. Among them, glycogen synthase kinase-3 (GSK-3) is a known tau kinase that plays a

crucial role in AD pathology.184 In-vivo and in-vitro tau phosphorylation can be undertaken by GSK-3

and over-expression of GSK-3 leads to tau hyper phosphorylation.185,186 In an Alzheimer’s-affected brain,

GSK-3 is activated in pretangle neurons and accumulates in paired helical filaments.187 Under the above

premise, Deng et al135 carried out an in-vitro study on N2a (Neuro-2a: a fast growing neuroblastoma cell

line) cells that were grown and differentiated in 96-well culture plates at density of 1.5×105 cells in 100

μL. Cells were exposed to various concentrations of wortmannin (an indirect GSK-3 activator) for 2 h at

37 ºC in the presence or absence of a 12-h-preincubation with melatonin 25, 50, and 100 μmol/L or

Vitamin E (VE). The authors explored the underlying mechanism of tau hyper phosphorylation in an

Alzheimer's affected brain and the possible arresting strategies. One of the findings of the study was that

preincubation of the cells with melatonin or vitamin E attenuated differentially wortmannin induced

oxidative stress as well as GSK-3 over-activation and tau hyper phosphorylation. The authors concluded

that wortmannin might be an effective tool for reproducing AD-like tau hyper phosphorylation cell model

and that melatonin/vitamin E can effectively protect the cells from wortmannin-induced impairments.

Contrary to the above findings on the possible therapeutic value of melatonin on cortical Aβ level

reduction, Quinn et al145 examined the effect of chronic melatonin therapy initiated after the appearance

of plaques in Tg2576 mice and failed to demonstrate any effect, as chronic melatonin therapy did not alter

amyloid burden or oxidative damage after 4-months of treatment. Variations in melatonin dose, age of

treatment initiation, and species of Aβ measured could possibly have accounted for the different results.

Quinn et al. noted that one of the prior studies used a lower dose and still found an effect at this dose

could not have accounted for the different result. The age at treatment initiation varied across the studies

and might possibly explain the different results. Another study188 using similar Tg2576 mice initiated

Page 45: Omonigho A. Michael Bubu

33

treatment at 4 months of age (prior to the appearance of hippocampal and cortical plaques) also found

diminished Aβ and nitrotyrosine at 7–8 months, but no significant difference in histologically detectable

Aβ or brain nitrotyrosine in animals at 15 months of age. The Quinn et al study initiated treatment at 14

months.

B.5 Randomized Controlled Trial

Asayama et al131 conducted a double blind study to examine the effects of melatonin on the

sleep-wake rhythm, cognitive and non-cognitive functions in Alzheimer type of dementia. The subjects

were 9 persons given a placebo (PLA), and 11 given melatonin (3 mg) (MLT). The drugs were given at

8:30pm each day for 4 weeks. Melatonin administration improved sleep time and night activity

significantly, but had no significant effect with regard to improving daytime naps and activity. Melatonin

administration improved cognitive and non-cognitive functions significantly. Melatonin seemed to be

useful for care of the Alzheimer type of dementia patients

C. Hypoxia (Apnea Hypopnea Index) and Cognitive decline and/or Alzheimer’s disease

Sixteen studies (i.e., 4 cross-sectional,81,83,86,90 1 case-control,103 3 prospective cohort,125,128,150 1

retrospective cohort,112 2 RCTs,130,132 and 5 experimental studies139,141,147-149) examined hypoxia states

(AHI) in sleep apnea/sleep disordered breathing and its association with cognitive decline and/or AD.

Table 1.9 presents the results of the descriptive study characteristics, statistical methods and main

findings of the studies.

C.1 Cross-sectional studies

Kadotani et al86 conducted a cross-sectional study to determine whether genetic variation at the

level of APOE is associated with Sleep Disordered-Breathing (SDB) in a probability based sampling of

791 middle-aged adults, mean age of 49 and age range 32-68. They found that independent of age, sex,

body mass index, and ethnicity, participants with APOE epsilon4 had a significantly higher risk of

moderate-to-severe Sleep Disordered-Breathing (apnea-hypopnea index (AHI) ≥15%) (P =.003).

Page 46: Omonigho A. Michael Bubu

34

Participants with APOE epsilon4 also had a significantly higher mean (SEM) apnea-hypopnea index

(p=.01). These effects increased with the number of APOE epsilon4 alleles carried. In a similar study,

Gottlieb et al83 examined the association of the APOE e4 allele with Obstructive Sleep Apnea/Hypopnea

(OSAH) in a community-dwelling cohort, exploring age dependency of the association. The investigators

conducted a genetic association study, nested within a prospective cohort study of the cardiovascular

consequences of OSAH. APOE e4 allele was associated with increased odds of OSAH (OR 1.41, 95% CI

1.06 to 1.87, p = 0.02) after controlling for age, sex and body mass index (BMI). The effect was greater in

subjects < 75 (OR 1.61, CI 1.02 to 2.54) as in those ≥75 years old (OR 1.32, CI 0.91 to 1.90). Exploratory

analyses revealed that the strongest effect of APOE e4 was in subjects age <65 (OR 3.08, CI 1.43 to

6.64), and was stronger in those with hypertension or cardiovascular disease than in those without. The

authors therefore concluded that the APOE e4 allele is associated with increased risk of OSAH,

particularly in individuals under age 65.83 Osorio et al90 examined the interaction between sleep-

disordered breathing and Apolipoprotein E genotype on cerebrospinal fluid biomarkers for Alzheimer’s

disease in a total of 95 cognitively normal elderly participants who were analyzed for SDB severity.

Among the 95 participants, 25 were considered free of SDB (apnea and/or hypopnea with 4% O2-

desaturation index (AHI4%) < 5) and were included as normal controls, 51 had mild SDB (AHI4% 5-

14.99), and 19 had moderate to severe SDB (AHI4% > 15). Results showed that in ApoE3+ subjects,

significant differences were found between sleep groups for p-tau and t-tau (p=0.017, p = 0.043

respectively), and the AHI4% was significantly correlated with p-tau (p = 0.023), t-tau (p = 0.021), and

Aβ-42 (p = 0.021). In ApoE2+ subjects, the AHI4% was significantly correlated with lower levels of CSF

Aβ-42 (p = 0.004), identical to ApoE4+ subjects where there was also a trend toward lower CSF Aβ-42

levels.

Lastly, Djonlagic et al81 examined the effect of aging on sleep-dependent motor memory

consolidation in patients with and without OSA. Participants included 44 patients (19–68 years) who had

been referred by a physician for a baseline polysomnography (PSG) evaluation and were assigned either

Page 47: Omonigho A. Michael Bubu

35

to the OSA group (AHI>5/h), or control (Non-OSA) group (AHI<5/h). All subjects performed the

Psychomotor Vigilance Task (PVT) and the Motor Sequence Learning Task (MST) in the evening and

again in the morning after their PSG. Results showed that healthy sleepers displayed a greater degree of

sleep-dependent overnight improvement on the MST, an effect not diminished by increasing age. The

existence of untreated obstructive sleep apnea was associated with an aging-related cognitive deficit,

otherwise not present in individuals without OSA. Other research has linked OSA to a greater risk of

developing dementia. Future studies are therefore necessary to determine if the restriction of memory

consolidation is tied to or has a role to play in the onset of neurodegenerative disease.

C.2 Case-control studies

O’Hara et al103 investigated the relationship between obstructive sleep apnea/hypopnea (OSAH)

and cognition in 36 older adults, i.e., 18 APOE epsilon4 carriers, and 18 non-carriers. These authors

found that higher levels of AHI were associated with lower memory scores in the APOE epsilon4 carriers

only. Minimum oxygen saturation (MinSaO2) did not differ significantly between the groups, but was

lower than 90% in both, indicative of the occurrence of hypoxia. MinSaO2 was not associated with

cognition, although there was a non-significant trend for minSaO2 to negatively impact memory in the

APOE epsilon4 carriers only.

C.3 Cohort studies

Cohen-Zion et al150 examined changes in cognitive function associated with Sleep Disordered

Breathing (SDB) in 46 participants aged 65 and older who completed visits 3 and 4. The original cohort

included community-dwelling elders age 65 and older with high risk for SDB studied from 1981 through

1985 and then followed every 2 years. The study results suggest that declining cognitive function is

associated primarily with increases in daytime sleepiness. Although cognitive decline was also associated

with increases in respiratory disturbance index (RDI: defined as number of apneas plus hypopneas per

hour of sleep), this association did not hold in a more inclusive model which also included variable of

Page 48: Omonigho A. Michael Bubu

36

SDB, oximetry, sleep and subjective report. The findings could suggest that any relationship between

SDB and cognitive function may be mediated by the effect of SDB on daytime sleepiness. Tworoger et

al125 found no associations with snoring, sleep duration, and difficulty sleeping and cognitive decline over

2 years. Chang et al112 in a retrospective matched-control cohort study estimated and compared the risk

of dementia in Sleep Apnea (SA) and non-SA patients among persons aged 40 and above over a 5-year

follow-up period. Overall findings suggest that SA patients had a 70% greater risk of developing

dementia within 5 years of diagnosis compared to non-SA age- and sex-matched patients, after adjusting

for other risk factors (95% confidence interval (CI) = 1.26-2.31; P < .01).

Yaffe et al128 examined the prospective relationship between sleep disordered breathing (SDB),

and cognitive impairment and investigated potential mechanisms of this association. Using data from 298

women without dementia (mean [SD] age: 82.3 [3.2] years), the authors found that women with SDB had

an 85% increased risk of developing MCI or dementia compared to women without SDB after adjusting

for age, race, body mass index, education, smoking status, presence of diabetes, presence of hypertension,

medication use (antidepressants, benzodiazepines, or non- benzodiazepine anxiolytics) and baseline

cognitive scores (adjusted OR=1.85, 95% CI 1.11-3.08). Measures of disordered breathing i.e., elevated

oxygen desaturation index (≥15 events/hour) and high percentage (>7%) of sleep time in apnea or

hypopnea, were both associated with risk of developing MCI or dementia (adjusted OR=1.71, 95% CI

1.04 − 2.83 and adjusted OR=2.04, 95% CI 1.10 − 3.78, respectively). Sleep fragmentation measures

(arousal index and wake after sleep onset) or sleep duration (total sleep time) were not associated with

risk of cognitive impairment.

C.4 Experimental studies

Shiota et al147 examined the direct effect of intermittent hypoxia (IH) in pathophysiology of AD

in-vivo and in-vitro. In-vivo, 15 male triple transgenic AD mice were exposed to either chronic

intermittent hypoxia (CIH) or normoxia (5% O2 and 21% O2 every 10 min, 8 h/day for 4 weeks). In-

vitro, human neuroblastoma SH-SY5Y cells stably expressing wild-type amyloid-β protein precursor

were exposed to either IH (8 cycles of 1% O2 for 10 min followed by 21% O2 for 20 min) or normoxia.

Page 49: Omonigho A. Michael Bubu

37

Results showed that CIH significantly increased levels of Aβ42 but not Aβ40 in the brains of mice

without the increase in hypoxia-inducible factor 1, alpha subunit (HIF-1α) expression. Furthermore, CIH

significantly increased intracellular Aβ-42 in the brain cortex. There were no significant changes in

cognitive function. IH significantly increased levels of Aβ42 in the medium of SH-SY5Y cells without

the increase in the HIF-1α expression. CIH directly and selectively increased levels of Aβ42 in the AD

model. Kaushal et al139 hypothesized that acute short-term Intermittent hypoxia (IH) and sleep

fragmentation (SF) (major manifestations of sleep apnea), as well as their combination (IH+SF) may

reveal unique susceptibility in sleep integrity in a murine model of AD. They found that slow wave sleep

(SWS) was significantly decreased, and rapid eye movement (REM) sleep was almost suppressed during

acute exposure to IH alone and IH+SF for 6 h in hApoE4 animals, with milder effects in WT controls.

Reduced delta power during SWS did not show post exposure rebound in hApoE4 unlike WT controls. IH

and IH+SF induced hypothermia was more prominent in hApoE4 than wild type (WT) controls. Mice

subjected to SF also showed sleep deficits but without hypothermia. “hApoE4 mice, unlike WT controls,

exhibited increased sleep propensity, especially following IH and IH+SF, suggesting limited ability for

sleep recovery in hApoE4 mice.” These findings affirm the probable impact of IH and SF in adjusting

sleep architecture and sleep homeostasis including sustenance of body temperature. In addition, the

increased susceptibility and defined recovery capability of hApoE4 mice to sleep apnea suggests that

early identification and treatment of sleep apnea in AD patients may permit interventions to slow AD

progression.

Zhang et al149 investigated the possibility of a direct molecular link between hypoxic insults and

Amyloid Precursor Protein (APP) processing and found that acute hypoxia increased the expression and

the enzymatic activity of β-secretase (BACE1) (Aβ is derived from the β-amyloid precursor protein

(APP) by sequential proteolytic cleavages from β-secretase (BACE1) and presenilin-1 (PS1)/γ-secretase)

by up-regulating the level of BACE1 mRNA, resulting in increases in the APP C-terminal fragment-β

(βCTF) and Aβ. Hypoxia had no effect on the level of PS1, APP, and tumor necrosis factor--converting

Page 50: Omonigho A. Michael Bubu

38

enzyme (TACE, an enzyme known to cleave APP at the α-secretase cleavage site). Sequence analysis,

mutagenesis, and gel shift studies revealed binding of Hypoxia Inducible Factor-1 (HIF-1) to the BACE1

promoter. Overexpression of HIF-1 α increased BACE1 mRNA and protein level, whereas down-

regulation of HIF-1 α reduced the level of BACE1. Hypoxic treatment failed to further potentiate the

stimulatory effect of HIF-1α overexpression on BACE1 expression, suggesting that hypoxic induction of

BACE1 expression is primarily mediated by HIF-1α. Lastly, the authors observed significant reduction in

BACE1 protein levels in the hippocampus and the cortex of HIF-1α conditional knock-out mice. These

results provide evidence that hypoxia/HIF-1α plays a vital role in balancing the amyloidogenic processing

of APP. It also provides a molecular mechanism to explain increased incidence of AD following cerebral

ischemic and stroke injuries. Following in the same line of thought, Li et al141 investigated molecular

mechanisms underlying hypoxia mediated AD using AD transgenic mice. The authors demonstrated that

repeated hypoxia increased β-amyloid (Aβ) generation and neuritic plaque formation by elevating β-

cleavage of APP in APPswe + PS1A246E transgenic mice. They also found that hypoxia enhanced the

expression of APH-1a, a component of ɣ-secretase complex, which in turn may lead to increase in ɣ-

cleavage activity. Furthermore, these authors demonstrated that repeated hypoxia treatment can activate

macroautophagy, which may contribute to the increases in Aβ production. Taken together, the results

suggest an important role of hypoxia in modulating the APP processing by facilitating both β- and ɣ-

cleavage which may result in a significant increase of Aβ generation. Sun et al148 constructed a series of

deletion luciferase reporter plasmids containing human BACE1 gene promoter regions to investigate

whether BACE1 is one of the downstream target genes of the hypoxia signaling pathway. In the study,

authors found that hypoxia significantly increased BACE1 gene expression, resulting in increased β-

secretase activity and Aβ production. Furthermore, hypoxia treatment markedly increased Aβ deposition

and potentiated the memory deficit in AD transgenic mice. The results demonstrate that hypoxia can

facilitate AD pathogenesis, and they provide a molecular mechanism to link vascular factors to AD.

Page 51: Omonigho A. Michael Bubu

39

C.5 Randomized Controlled Trials

Ancoli-Israel et al130 examined whether treatment of obstructive sleep apnea (OSA) with

continuous positive airway pressure (CPAP) in patients with Alzheimer's disease (AD) would result in

improved cognitive function. Participants were randomized to either therapeutic CPAP for six weeks or

placebo CPAP for three weeks followed by therapeutic CPAP for three weeks. A comparison of subjects

randomized to 3 weeks of therapeutic versus placebo CPAP suggested no significant improvements in

cognition. A comparison of pre- versus post-treatment neuropsychological test scores after 3 weeks of

therapeutic CPAP in both groups showed a significant improvement in cognition. Similarly, Kushida et

al132 determined the neurocognitive effects of continuous positive airway pressure (CPAP) therapy on

patients with obstructive sleep apnea (OSA) in a 6-month, randomized, double-blind, 2-arm, sham-

controlled, multicenter trial. The results showed that CPAP treatment improved both subjectively and

objectively measured sleepiness, especially in individuals with severe OSA (AHI >30). CPAP use also

resulted in mild, transient improvement in the most sensitive measures of executive and frontal-lobe

function for those with severe disease, which suggests the existence of a complex OSA-neurocognitive

relationship.

D. Insomnia and cognitive decline and/or Alzheimer’s disease

Six studies (i.e., 2 cross-sectional87,92 and 4 prospective cohort studies114,115,117,121) examined the

association between insomnia and cognitive decline and/or AD. Table 1.10 presents the results of the

descriptive study characteristics, statistical methods and main findings of the studies.

D.1 Cross-sectional studies

Pistacchi et al92 in their study found that patients with MCI had a frequency of sleep disturbances

of any type equal to that of patients with AD and presented mostly with insomnia. In the study insomnia

occurred more frequently in AD. In their cross-sectional study examining the association between

multiple sleep parameters including insomnia and cognitive impairment in the elderly, Merlino et al87 did

not find an association between insomnia, the most common sleep complaint in their sample, and the

Page 52: Omonigho A. Michael Bubu

40

presence of cognitive decline. These studies assessed the presence of sleep disorders by means of

subjective symptoms and not using objective instruments, such as wrist actigraphy or polysomnography.

Polysomnography is essential for a correct diagnosis of some sleep disturbances. In particular, sleep

disordered breathing can be diagnosed only by means of polysomnographic recordings. Because

assessment of sleep parameters was based on a structured interview, obtaining self-reported description of

sleep characteristics from subjects with significant cognitive impairment becomes challenging and

questionable.

D.2 Cohort studies

Examining whether self-reported symptoms of insomnia independently increase risk of cognitive

decline in older adults, Osorio et al121 and Cricco et al114 found that there is a greater risk of Alzheimer's

disease in adults with self-reported insomnia. Osorio et al121 examined the relationship between insomnia

and AD in 346 cognitively normal older adults evaluated during multiple visits over 7.7 years of follow-

up. Insomnia was collected only during the normal cognition stage and screened using items 4, 5, and 6

from the Hamilton Depression Rating Scale (HAM-D). Participants who reported insomnia had an

increased odds of 2.39 of developing AD when compared to the non-insomnia group. (OR = 2.39, 95%

confidence interval (CI) = 1.03–5.55). Depression did not confound the association, and individuals with

insomnia showed a faster progression to dementia (chi-square = 3.94, P = .047). Cricco et al114 found in

their study sample that self-reported symptoms of insomnia independently predicted incident cognitive

decline in older men, but not in women. Compared to those with no insomnia at the third annual follow

up (FU3) those reporting symptoms of insomnia at both baseline and FU3 had an adjusted odds ratio

(AOR) of 1.49 (95% CI = 1.03–2.14) for cognitive decline among non-depressed men. Men with

insomnia at FU3 only were not at increased risk (OR = 1.16, 95% CI = 0.82–1.65). These relationships

were not found in women. Men and women with depressive symptoms at FU3 were at increased risk for

cognitive decline independent of insomnia. Participants included over 6,000 community-dwelling men

and women age 65 and older from the prospective cohort of the four sites of the Established Populations

Page 53: Omonigho A. Michael Bubu

41

for Epidemiologic Studies of the Elderly (EPESE) in Iceland. The authors noted that sleep complaints

data were only available at 3-year intervals, making misclassification of exposure in some cases more

likely, e.g., some participants classified as experiencing chronic sleep disturbances may have had

transient episodes of sleep difficulty during follow-up but may have had undisturbed sleep over the period

between follow-ups. Likewise, incident cases could have had symptoms for up to 3 years. Thus, the odds

ratios found in this study can be considered conservative estimates of risk.

Using the Maastricht Ageing Study (MAAS) cohort in Netherlands, Jelicic et al117 examined

whether subjective sleep complaints (i.e., extent to which an individual was distressed by difficulties with

falling asleep, waking up too early and by a restless or disturbed sleep) in a population-based sample of

838 middle aged and older adults (≥ 50 years) predicted cognitive decline over a period of 3 years.

Results showed that subjective sleep complaints were negatively associated with cognitive performance at

follow-up (β = -0.05 95% CI: -0.09 to -0.00, p = 0.03). Foley et al115 examined the longitudinal

association between sleep disturbances (insomnia and daytime sleepiness) and incidence of dementia and

cognitive decline in 2346 Japanese-American men aged 71 to 93 in the Honolulu-Asia Aging Study

(HAAS). Sleep disturbances were assessed by a questionnaire that asked whether participants were sleepy

during the day or had insomnia (defined as “usually” having trouble falling asleep or waking up far too

early and not going back to sleep). Individuals were screened for possible dementia using the CASI.

Dementia was diagnosed according to less-restrictive diagnostic criteria developed by Cummings et al.189

for very mild dementia and more-restrictive diagnostic criteria for mild or more severe dementia from the

Diagnostic and Statistical Manual of Mental Disorders, Third Edition, Revised (DSM-IIIR).190 Results

from the study showed that participants with excessive daytime sleepiness at baseline (8%) were twice as

likely to be diagnosed with incident dementia than were those not reporting daytime sleepiness [adjusted

odds ratio (OR) = 2.19, 95% confidence interval (CI) = 1.37–3.50] and about 40% more likely to have a 9

or more point drop in their CASI score between examinations [OR = 1.44, 95% CI =1.01–2.08]. Insomnia

was not associated with cognitive decline or incidence of dementia. The authors noted that there was lack

Page 54: Omonigho A. Michael Bubu

42

of information to identify persons with sleep disordered breathing, which has been shown to affect

cognitive performance191,192 and has a 25% prevalence in older persons.193

E. Restless Leg Syndrome and cognitive decline and/or Alzheimer’s disease

Table 1.11 presents the results of the descriptive study characteristics, statistical methods and main

findings of the 3 studies (i.e., 2 cross-sectional84,92 and 1 case-control104) that examined any association

between restless leg syndrome and cognitive decline and/or AD.

E.1 Cross-sectional

Restless legs syndrome (RLS) can inhibit falling asleep or being able to stay asleep. RLS

incidence is higher in women and increases with age.194 Depression, antidepressant drug intake, renal

failure, and peripheral neuropathy, are known risk factors for RLS and also increase with age. Guarnieri

et al84 reported a 6% prevalence of RLS in MCI and Demented (AD/non-AD patients) assessed by the

international RLS study group155,195 questionnaire. However, in cognitively impaired individuals,

assessing RLS by use of a questionnaire might be arduous. Another study196 reported an RLS prevalence

of 24% in 59 demented individuals using specific guidelines of probable RLS that emphasizes behavioral

indicators and supportive features such as dopaminergic responsiveness, patient’s past history, and

presence of periodic limb movements, in cognitively impaired elderly.197 Pistacchi et al92 enrolled 236

patients (78 men and 158 women) with different subtypes of dementia and showed that RLS was one of

the most common symptoms in MCI patients.

E.2 Case-control studies

Given the degree of chronic sleep deprivation that RLS patients’ experience, Pearson et al104

hypothesized that RLS patients would exhibit decreased cognitive performance particularly on the more

pre-frontal cortical functioning (PFC) focused cognitive tasks relative to age matched controls with

relatively less or no impairments on other cognitive tasks. Results showed that RLS patients compared to

controls showed significant (P<0.05) and sizeable (20–40%) deficits on Verbal Fluency tests and the Trail

Page 55: Omonigho A. Michael Bubu

43

Making tests. The authors concluded that RLS patients have statistically significant specific cognitive

deficits and show cognitive deficits similar to those seen with 36 h total sleep deprivation.

DISCUSSION

The purpose of this systematic review was to examine the association of sleep and its related

problems/disorders with Alzheimer’s disease, cognitive decline or dementia and to evaluate the evidence

for a causal association. We systematically and critically synthesized and evaluated data from 72 original

publications. Study quality was assessed using an adaptation from previous systematic reviews73,74 and the

modified version of the Newcastle-Ottawa scale (NOS) for quality assessment of the observational

studies,75,76 with addition of new items relevant to this review. Appropriateness of study design, study

populations, study samples, size of the study sample, exposure and outcome measurement, statistical

analytic design, adjustment of potential sources of confounding, variations in study populations, and

numerous sources of bias were considered. Epidemiological studies can assess risk and contribute to

understanding of possible causation but possess built-in limitations related to their design. Consistency of

results, dose-response relationships and temporality play a crucial role in determining causation; it is

however necessary to consider high quality studies when accepting or rejecting causal associations

between exposure and outcome. Out of the 72 studies reviewed, only 14 studies were assessed to be of

low quality, while 58 studies were of medium to high quality, signifying that the publications reviewed in

this systematic review were mostly satisfactory in their design.

In this comprehensive review of seventy-two studies, there is strong evidence that sleep and its

related abnormalities/disorders are associated with cognitive decline and/or Alzheimer’s disease

independent of other factors. Summarizations of sectional findings already discussed in the sections above

are provided below.

A. Sleep depth and Cognitive decline and/or Alzheimer’s disease

Page 56: Omonigho A. Michael Bubu

44

The vast majority of the cross-sectional studies reviewed demonstrated an association between

sleep depth and cognitive decline and/or AD and/or Dementia. The association between sleep quality and

cognition was not explained by confound factors such as cerebrovascular disease, depression, or

medication usage.88 Objectively measured disturbed sleep was consistently related to poorer cognition,

whereas total sleep time was not. Sleep duration during the 24-h day was positively correlated with the

severity of dementia in nursing home patients,82 however, in a community-dwelling of women 65 years

old or older, there was no significant relationship for total sleep time with poorer cognition. These

findings may suggest that it is disturbance of sleep rather than quantity that affects cognition in demented

patients, while both sleep disturbance and duration affect cognition in the healthy elderly.

It is important to note that one cross-sectional study93 reported no significant differences between

good sleepers (GS) (PSQI <= 5) and poor sleepers (PSQI >= 5) in any of the objective cognitive function

tests except for the Trail Making Test A (TMA-A), processing speed being longer in the PS group (p <

0.001), and as such concluded that in healthy elderly subjects subjective sleep quality and duration was

not associated with subjective and objective cognitive performances, except for attention. However, in

this study, sleep duration and quality were self-reported and the authors had no information on sleep

structure and sleep fragmentation, two factors implicated in self-reported cognitive deficits.

Cross-sectional studies examining the association between sleep depth and AD pathology also

demonstrate an association. Both Spira et al94 and Ju et al85 showed that amyloid deposition in the

preclinical stage of AD appears to be associated with worse sleep quality. Spira et al also demonstrated an

association with sleep duration, however, Ju et al did not. The contrasted findings could possibly be due

to the variations in Aβ42 burden assessment. In Ju et al Aβ42 was measured using the Alzheimer Disease

Research Center (ADRC) Biomarker Core using enzyme linked immunosorbent assay while Spira et al

measured β-amyloid burden, using carbon 11–labeled Pittsburgh compound B positron emission

tomography distribution volume ratios (DVRs).

Page 57: Omonigho A. Michael Bubu

45

Results from reviewed case-control studies examining sleep depth and cognitive decline and/or

AD suggest that sleep disruptions are evident years before diagnosis of AD and that sleep is significantly

impaired in patients with MCI at both the objective and subjective level.98,99 This may have implications

for sleep being used as a surrogate marker of preclinical Alzheimer disease and for early detection of

dementia and/or therapeutic management of sleep complaints in MCI patients.

Case-control studies reviewed show an association between sleep depth and AD pathology

demonstrating that increased plasma Aβ42 levels are significantly associated with fragmented SWS in

amnestic MCI subjects,105 thereby suggesting that sleep disruptions may signal Aβ burden in persons at

increased risk for AD, and that changes in theta rhythm during REM and SWS are a relevant index of

brain impairments in the early stage of AD, and that a deficiency of Ach may result in an increase of

sEMG activity in MCI patients. Since cholinesterase inhibitors are capable of suppressing sEMG activity

in AD patients, it is possible that an increase in sEMG activity is associated with a deficiency of Ach,

which could be an early indicator of dementia

All cohort studies (prospective and retrospective) that evaluated the relationship between sleep

depth parameters and cognitive decline and/or AD reveal that sleep depth parameters increase the risk of

cognitive decline and/or developing AD. Both subjective and objective sleep assessments yield similar

outcomes of a relationship between sleep depth parameters and cognitive decline and/or AD independent

of other factors including depression/depressive symptoms.109,111,122 These findings suggest that sleep

depth parameters such as daytime napping, and night-time sleep duration, may be modifiable behaviors

open to intervention strategies, or, clinical indicators of future decline in older individuals. However, after

adjusting for depressive symptoms, Hahn et al116 could no longer show that self-reported sleep problems

may increase the risk for dementia. However, the authors measured depressive symptoms with only one

self-reported item. They also noted that they did not include longitudinal data on sleep. Research with

longitudinal sleep data is definitely preferable. Also, the relatively small sample size (n=214) may have

biased the results toward a Type II error, particularly in the more refined analyses.

Page 58: Omonigho A. Michael Bubu

46

The only cohort study reviewed,120 examining the relationship between sleep depth parameters

and AD pathology show that better sleep consolidation attenuates the effect of APOE genotype on

incident AD and development of neurofibrillary tangle pathology. Assessment of sleep consolidation may

identify APOE+ individuals at high risk for incident AD, and interventions to enhance sleep consolidation

should be studied as potentially useful means to reduce the risk of AD and development of neurofibrillary

tangles in APOE ε4+ individuals.

Experimental studies and RCTs evaluated the relationship between sleep depth parameters and

AD pathology and all demonstrate that sleep deprivation, or prolonged wakefulness, interferes with a

physiological morning decrease in Aβ42 and that chronic sleep deprivation increases cerebral Aβ42

levels, which elevates the risk of Alzheimer disease. Interestingly, Kang et al138 showed that orexin may

play a role in the pathogenesis of Alzheimer’s disease and Rothman et al146 demonstrated significant

positive correlations between cortical Aβ and p -Tau levels and circulating corticosterone. The latter

finding might suggest a probable role for glucocorticoids in mediating behavioral and biochemical

changes observed after sleep restriction in a mouse model of AD.

B. Circadian Rhythm/sleep-wake abnormalities and cognitive decline and/or Alzheimer’s

disease

All cross-sectional studies that evaluated any association between circadian Rhythm/sleep-wake

abnormalities and cognitive decline and/or Alzheimer’s disease demonstrated that an association between

the rest-activity rhythm and cognitive performance was present in elderly people. They also showed that

with the progression of dementia, excessive daytime sleepiness, the capacity to maintain sleep and the

capacity to maintain wakefulness are impaired, and result in complete fragmentation of sleep/wakefulness

during the night and day.87,89,91 However, it is unlikely that a relationship with a clinically meaningful

correlation exists between the circadian rhythm-associated SNPs and WASO in individuals with AD.95

All case-control studies that examined any association between circadian Rhythm/sleep-wake

abnormalities and cognitive decline and/or Alzheimer’s disease showed that circadian misalignment and

Page 59: Omonigho A. Michael Bubu

47

sleep disruption was evident in patients with MCI, and was consistent with changes observed in

Alzheimer’s disease. Such findings could be a marker for disease trajectory, and may even be implicated

in disease pathogenesis.

Case-control studies that examined any association between circadian rhythm/sleep-wake

abnormalities and AD pathology demonstrated that the hypocretin system was affected in advanced AD,

and that CSF melatonin levels was lower in AD patients than in old control subjects. CSF melatonin

levels from APOE-e3/4 genotype patients were significantly higher than those from the APOE-e4/4

genotype, suggesting a relationship between melatonin levels and signs and symptoms of AD.97,101,107 An

interesting study by Schmidt et al106 reported that cerebrospinal fluid melanin concentrating hormone

(CSF-MCH) correlated with T-tau (p=0.01) and P-tau (p=0.05) in the AD and that CSF-MCH correlated

negatively with MMSE scores in the AD (p=.001). These results add to preclinical studies and may allow

further research on the role of the hypothalamus in pathology and occurrence of behavioral disturbances

in AD. The authors noted that future investigations, including repeated MCH/HCRT-1 level

measurements in combination with sleep-wake-actigraphy and assessment of sleep by questionnaires,

histochemical determinations of MCH in animal and post-mortem tissues are warranted to understand the

hypothalamic impact in AD in greater detail.

All prospective cohort studies in this systematic review that examined the relationship between

circadian rhythm/sleep-wake abnormalities and cognitive decline and/or AD revealed associations

between the two, with older, healthy individuals with decreased circadian activity rhythm amplitude and

robustness, and delayed rhythms demonstrating increased risks of developing dementia and MCI.

Yesavage et al129 also showed that sleep-wake APOE status is associated with the progression of

sleep/wake disturbances in AD

Most experimental studies demonstrated anti-amyloidogenic effects of melatonin either in-vitro

and/or in-vivo in animal studies. These results suggest that melatonin can provide a combination of

Page 60: Omonigho A. Michael Bubu

48

antioxidant and anti-amyloidogenic features that can be explored either as a preventive or therapeutic

treatment for AD or as a model for development of anti-amyloidogenic indole analogs.

It is important to note that Quinn et al145 examined the effect of chronic melatonin therapy

initiated after the appearance of plaques in Tg2576 mice and failed to demonstrate any effect, as chronic

melatonin therapy did not alter amyloid burden or oxidative damage after 4-months of treatment.

Variations in melatonin dose, age of treatment initiation, and species of Aβ measured could possibly have

accounted for the different results. Quinn et al. noted that one of the prior studies used a lower dose and

still found an effect at this dose could not have accounted for the different result. The age at treatment

initiation varied across the studies and might possibly explain the different results. Another study188 using

similar Tg2576 mice initiated treatment at 4 months of age (prior to the appearance of hippocampal and

cortical plaques) also found diminished Aβ and nitrotyrosine at 7–8 months, but no significant difference

in histologically detectable Aβ or brain nitrotyrosine in animals at 15 months of age. The Quinn et al

study initiated treatment at 14 months.

Melatonin administration improved cognitive and non-cognitive functions significantly, and

seemed to be useful for care of the Alzheimer type of dementia patients as shown in the RCT conducted

by Asayama et al.131

C. Hypoxia (Apnea Hypopnea Index) and Cognitive decline and/or Alzheimer’s disease

Cross-sectional studies in our review that examined the association between hypoxemia and

cognitive decline and/or AD all showed that an association exists between SDB/OSA and CSF

Alzheimer’s disease-biomarkers in cognitively normal elderly individuals, thereby suggesting that

existing therapies for SDB such as continuous positive airway pressure could delay the onset to MCI or

dementia in normal elderly individuals. Cross-sectional studies in our review also showed that the APOE

£4 allele was associated with increased risk of SDB/OSAH, particularly in individuals under age 65.83,86

Page 61: Omonigho A. Michael Bubu

49

The only case-control study that evaluated the association between hypoxemia and cognitive

decline and/or AD also demonstrated that nocturnal sleep apnea/hypopnea was associated with lower

memory performance in APOE e4 carriers.103

Most cohort studies (retrospective and prospective) evaluated in our study revealed that

OSA/SDB may be a risk factor for dementia, with some suggesting that this relationship could be gender-

dependent, age-dependent, and time-dependent.112,128,150 In contrast, Tworoger et al125 in their study

found no associations between snoring or any of the sleep variables examined (average sleep duration,

snoring frequency, and difficulty sleeping over the previous 4 weeks) and cognitive decline over 2 years.

All 5 experimental studies that examined an association between hypoxemia and AD pathology

demonstrated an association between hypoxia and AD biomarkers. Two studies148,149 found that hypoxia

significantly increased BACE1 gene expression, resulting in increased β-secretase activity and Aβ

production. Overexpression of HIF-1 α increased BACE1 mRNA and protein level, whereas down-

regulation of HIF-1 α reduced the level of BACE1. Hypoxic treatment failed to further potentiate the

stimulatory effect of HIF-1α overexpression on BACE1 expression, suggesting that hypoxic induction of

BACE1 expression is primarily mediated by HIF-1α. Furthermore, hypoxia treatment markedly increased

Aβ deposition and potentiated the memory deficit in AD transgenic mice. These results provide evidence

that hypoxia/HIF-1α plays a vital role in balancing the amyloidogenic processing of APP, that hypoxia

can facilitate AD pathogenesis, and they provide a molecular mechanism to link vascular factors to AD.

Two other studies141,147 demonstrated that repeated hypoxia increased β-amyloid (Aβ) generation

and neuritic plaque formation by elevating β-cleavage of AD transgenic mice. They also found that

hypoxia enhanced the expression of APH-1a, a component of ɣ-secretase complex, which in turn may

lead to increase in ɣ-cleavage activity. Furthermore, these authors demonstrated that repeated hypoxia

treatment can activate macroautophagy, which may contribute to the increases in Aβ production. Taken

together, the results suggest an important role of hypoxia in modulating the APP processing by

facilitating both β- and ɣ-cleavage which may result in a significant increase of Aβ generation

Page 62: Omonigho A. Michael Bubu

50

Both RCTs130,132 that evaluated the cognitive effects of CPAP treatment demonstrated that CPAP

improved both subjectively and objectively measured sleepiness, especially in individuals with severe

OSA (AHI >30). CPAP use resulted in mild, transient improvement in the most sensitive measures of

executive and frontal-lobe function for those with severe disease, which suggests the existence of a

complex OSA-neurocognitive relationship.

D. Insomnia and Cognitive decline and/or Alzheimer’s disease

All the 4 prospective studies reviewed that examined the association between insomnia

parameters and cognitive decline and/or AD revealed that there is a greater risk of cognitive decline

and/or Alzheimer's disease in adults with insomnia and/or insomnia like symptoms. The only study that

did not find an association was cross-sectional and assessment of sleep parameters was based on a

structured interview, obtaining self-reported description of sleep characteristics from subjects with

significant cognitive impairment.

E. Restless Leg Syndrome and Cognitive decline and/or Alzheimer’s disease

All 3 studies (2 cross-sectional and 1 case-control) that evaluated RLS and cognitive decline

and/or AD associations concluded that RLS patients have statistically significant specific cognitive

deficits. Pearson et al104 showed that RLS patients compared to controls showed significant (P<0.05) and

sizeable (20–40%) deficits on Verbal Fluency tests and the Trail Making tests.

Summary of evidence

1. Sleep is associated with cognitive decline and/or AD/AD pathology

Taken together, the cross-sectional studies reviewed in this study generally provide support for an

association of sleep and poor cognitive outcomes, including AD and dementia. They show that

individuals with AD , dementia or MCI are more likely to have several indicators of sleep problems,

including sleep depth problems such poor sleep quality, sleep efficiency, sleep latency and sleep

Page 63: Omonigho A. Michael Bubu

51

deprivation; sleep-wake/circadian rhythm abnormalities, excessive daytime sleepiness and OSA/SDB.

However a significant limitation of cross-sectional studies is that causal inferences cannot be made

because the temporal sequence of sleep and circadian disruption, neurodegeneration, and cognitive

decline cannot be assessed. In addition, depression or its symptoms may be important mediators or

confounders of the association between sleep variables and dementia, and should be considered in these

studies.116 However, not all studies clearly addressed the role of depression with certain studies having a

history of depression as an exclusion criteria without considering current depression which might well be

important when one is examining factors in relation to circadian rhythm. Some studies51,87,93 assessed the

presence of sleep disorders by means of subjective symptoms and not using objective instruments, such as

polysomnography, thereby limiting the credibility of information obtained from subjects with an

important cognitive impairment. Polysomnography is essential for a correct diagnosis of some sleep

disturbances. In particular, sleep disordered breathing can be diagnosed only by means of

polysomnographic recordings. Other studies made use of tools that provided limited information on sleep

maintenance, and no detail of sleep times or other sleep parameters and others had very small sample size.

Furthermore, the association between sleep and Alzheimer’s disease pathology, measured by

Alzheimer’s disease biomarkers such as Aβ and Phosphorylated Tau accumulations, has not been

extensively investigated in humans’ in-vivo. The above study findings linking sleep to AD pathology

definitely complement those linking poor sleep to cognitive decline and dementia, however, because of

the cross-sectional nature of the studies, we cannot establish temporality and consequently causality, as

we are unable to infer whether this pathology might cause or result from poor sleep. As such, Aβ

deposition may be a cause and/or occur as a complication during the progression of AD, and/or sleep

disturbance and Aβ plaques may have a shared cause. It is therefore evident that prospective

observational studies are needed to better understand the temporal order of sleep disturbance and AD

pathogenesis and/or biomarkers. This will aid efforts to better evaluate potential opportunities for targeted

and effective prevention.

Page 64: Omonigho A. Michael Bubu

52

Altogether, it can also be seen that studies suggest that sleep variables interact with the APOE

genotype to affect Alzheimer’s disease-related outcomes. Notably, an association between sleep

disturbance especially Sleep Disordered Breathing (SDB) and CSF Alzheimer’s disease-biomarkers in

cognitively normal elderly individuals is evident. Current treatments for SDB such as continuous positive

airway pressure (CPAP) could interrupt the onset of mild cognitive impairment (MCI) or dementia in

normal elderly individuals.

2. Sleep problems precede the onset of cognitive impairment

Taken together, these prospective studies provide support that sleep problems precede the onset

of cognitive impairment, including AD and dementia. Some of the findings suggest interactions by age,

and/or sex.112,114,126 These findings require replication, and if these associations are causal, prevention

efforts may need to be targeted differently at specific ages or in different populations. It is however

important to note that longitudinal studies of dementia are prone to miss both prevalent and incident cases

because full neurological evaluations are not conducted on all participants, especially when selected

participants refuse further neuropsychological testing and examination or die over the course of follow-

up.198-200 An important limitation of most of the prospective studies is the length of the follow up. Only 4

studies had greater than 5 years follow up. Although longitudinal studies can assess temporality, AD is a

lifelong disease process and preclinical AD is very much present prior to the onset of symptomatic AD,

therefore, reverse causation might have actually been the case in these studies since a <5 year follow-up

period may not be sufficient and thus may not reflect the true prospective nature of AD biomarkers and/or

cognitive decline, as certain participants may already have had the disease but were asymptomatic. Also

self-reported data on sleep duration may be subject to bias, including the existence of a change in the

typical sleep duration anticipating the onset of dementia. On the other hand, objective sleep measures are

particularly useful in studies of sleep, aging, and cognition since self-report sleep measures do not

necessarily correlate with objectively measured sleep, and discrepancies between these modes of

assessment may be more pronounced among older adults with cognitive impairment.108,201

Page 65: Omonigho A. Michael Bubu

53

3. Sleep problems may lead to increased AD pathology or may have their effects on dementia

through other pathways

Altogether, it can be seen that experimental studies provide the most compelling evidence of the

plausibility of causal associations between sleep deprivation and AD pathology and/or AD biomarkers,

but typically are performed in animals. The power of experimental mice models to mirror the extremely

complex physiological and biochemical processes in humans is a matter of dispute because of inherent

translational limitations of these models imposed by variations in size and physiology, as well as

differences in the homology of targets between mice and humans. However, from the above study

findings it is clear that alteration of the sleep-wake cycle and circadian rhythm regulation could be

important players in the onset and development of sporadic AD. Therefore, amending and/or

improvement of sleep-wake cycle abnormalities could be a viable therapeutic strategy for AD at risk

individuals. Both animal experimental studies and RCTs also demonstrate that hypoxemia might

contribute to cognitive dysfunction in severe cases, however, in mildly affected patients,192,202 the

interacting effect of cognitive reserve,203 intelligence level,204 and depression205,206 needs to be

considered. We also see from the above study findings that disrupted melatonin levels could be a

surrogate origin of sleep pathology, as decline in melatonin levels are known to occur with aging and this

decline is enhanced with AD, even in preclinical stages. Studies also show that melatonin’s cognitive

benefits could involve its anti-Aβ aggregation, anti-inflammatory, and/or antioxidant properties. Findings

provide reinforcement for long-term melatonin therapy as a primary or complementary strategy for

abating the progression of Alzheimer disease. Also as a result of the fact that the age at treatment

initiation of melatonin might play a role in its ability to suppress Aβ levels, the implication for human

studies could be that melatonin is probably unlikely to have a role in the treatment of established cerebral

amyloidosis characteristic of AD. If this is found to be true, then clinical trials of melatonin will have to

focus on prevention rather than treatment of established cerebral amyloidosis.

Page 66: Omonigho A. Michael Bubu

54

LIMITATIONS

Several methodological concerns must be considered when interpreting the findings of this

systematic review. First, as with any systematic review, one limitation relates to the potential bias

introduced in the paper selection process. To minimize this bias, we adhered strictly to the guidelines and

recommendations in the Cochrane handbook67 whenever possible. We also ensured that studies were not

restricted by language to guard against language bias. Secondly, the possible bias introduced by the

individual studies that were included is a further limitation. Various study designs were rated low for

assigning the grade of evidence.207 In addition to study design, conclusions made from the poor quality

studies could introduce possible bias. We included both high- and low-quality studies in the

summarization of findings because both mostly came to the same conclusion, therefore possibly

indicating that the study quality might not have influenced the overall conclusion. Thirdly, there was

considerable variation between the studies in the assessment methods used to verify sleep problems and

cognitive decline and/or AD/Dementia, suggesting heterogeneity that could possibly raise some questions

about the comparability of results across studies. Our research question focused on a broad concept of

sleep problems based on having any sleep problem symptoms. Thus, we assumed a common underlying

concept by making general conclusions regarding sleep problems and cognitive decline and/or AD.

Fourthly, as in any systematic review, publication bias may have affected the representativeness of the

included studies by over reporting significant findings. Non assessment of publication bias with a funnel

plot is a limitation. Lastly the possibility of a “file drawer” effect exists because exclusion and inclusion

criteria are always subjective.

CONCLUSION AND FUTURE DIRECTIONS

In summary, there is substantial evidence providing support for sleep and its related abnormalities

as a potential cause of cognitive decline, dementia, and Alzheimer’s disease pathology. Ample evidence

linking sleep impairments and circadian regulating mechanisms directly to clinical symptoms in AD

Page 67: Omonigho A. Michael Bubu

55

patients exist. Future research is needed to make clear the contribution of the different genetic,

neurodegenerative and environmental factors to sleep impairment. There is also growing experimental

and epidemiological evidence for close reciprocal interaction between cognitive decline and sleep

alteration, however, long-term longitudinal studies are required to better understand the relationship

between chronic sleep deprivation, circadian rhythm/sleep-wake abnormalities, sleep-disordered

breathing, sleep disorders like OSAS and cognitive decline. Future studies need to be able to define causal

pathways more definitively so we can be clear for example, whether neurodegeneration is leading to both

sleep changes and memory changes, or whether aging is contributing to OSA and/or sleep architecture

changes that in turn affect memory, and/or whether the effect of aging on sleep architecture predisposes to

both apnea and memory impairment. Thus, further research that examines these basic mechanisms as well

as employing clinical/translational approaches are needed. More research with objective sleep measures,

Alzheimer’s disease biomarkers, and cognitive measures are also needed. There is also significant

evidence from in vitro and animal studies for an important role of both melatonin and hypocretin in

modulating AD pathophysiology. Although the exact underlying mechanisms are unknown as of yet,

future studies examining the role of lack of sleep and circadian rhythm deterioration as causative factors

in the onset of AD, especially human studies, are needed, as their findings can be promising for the

development of new prevention and/or treatment opportunities.

RESEARCH AGENDA AND GAPS IN THE LITEARTURE I INTEND TO ADDRESS

The need for a meta-analysis to quantify the effect of sleep problems/disorders on cognitive

impairment and AD. This would allow for efficient integration of existing information from various

studies enabling us to establish whether the literature is consistent and can be generalized across

populations, settings, and/or whether findings vary significantly by particular subsets.

AIM I: To conduct a meta-analysis to quantify the aggregate effect of sleep problems and/or

disorders on cognitive decline and Alzheimer's disease.

Page 68: Omonigho A. Michael Bubu

56

The need to be able to define causal pathways more definitively so we can be clear for example,

whether neurodegeneration is leading to both sleep changes and memory changes, or whether aging is

contributing to OSA and/or sleep architecture changes that in turn affect memory, and/or whether the

effect of aging on sleep architecture predisposes to both apnea and memory impairment. One way to be

able to do this is to conduct more research examining relationships between OSA (preferably with

objective sleep measures) and Alzheimer’s disease biomarkers, and/or cognitive measures. This led to the

following study aims:

AIM IIa: To examine whether the presence of Obstructive Sleep Disordered Breathing is

associated with CSF P-Tau, Hippocampal atrophy and β-amyloid burden in older adults with

MCI and AD.

AIM IIb: To examine the effect of Obstructive Sleep Apnea on longitudinal changes of

Alzheimer's disease biomarkers in cognitive normal, mild cognitive impairment and Alzheimer's

disease patient

The need for clarity in understanding and appreciating the heterogeneity of OSA and its outcomes

in distinct age groups especially as it relates to cognitive function, subsequent cognitive decline and AD.

One way to do this is to integrate decades of research examining OSA and cognition; OSA and

subsequent cognitive decline; and OSA and AD; with particular focus in appreciating the heterogeneity of

OSA and its outcomes in distinct age groups.

AIM III: To conduct a Review titled "Obstructive Sleep Apnea: A Distinct Physiological

Phenotypic Risk Factor in older adults with Cognitive decline and Alzheimer’s disease"

References

1. Thies W, Bleiler L. 2013 Alzheimer's disease facts and figures. Alzheimer's & dementia : the

journal of the Alzheimer's Association. 2013;9(2):208-245.

Page 69: Omonigho A. Michael Bubu

57

2. Hebert LE, Beckett LA, Scherr PA, Evans DA. Annual incidence of Alzheimer disease in the

United States projected to the years 2000 through 2050. Alzheimer disease and associated

disorders. 2001;15(4):169-173.

3. Wimo A, Jonsson L, Bond J, Prince M, Winblad B. The worldwide economic impact of dementia

2010. Alzheimer's & dementia : the journal of the Alzheimer's Association. 2013;9(1):1-11 e13.

4. Brookmeyer R, Johnson E, Ziegler-Graham K, Arrighi HM. Forecasting the global burden of

Alzheimer's disease. Alzheimer's & dementia : the journal of the Alzheimer's Association.

2007;3(3):186-191.

5. Fratiglioni L, Ahlbom A, Viitanen M, Winblad B. Risk factors for late-onset Alzheimer's disease:

a population-based, case-control study. Annals of neurology. 1993;33(3):258-266.

6. Green RC, Cupples LA, Go R, et al. Risk of dementia among white and African American

relatives of patients with Alzheimer disease. JAMA : the journal of the American Medical

Association. 2002;287(3):329-336.

7. Mayeux R, Sano M, Chen J, Tatemichi T, Stern Y. Risk of dementia in first-degree relatives of

patients with Alzheimer's disease and related disorders. Archives of neurology. 1991;48(3):269-

273.

8. Farrer LA, Cupples LA, Haines JL, et al. Effects of age, sex, and ethnicity on the association

between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and

Alzheimer Disease Meta Analysis Consortium. JAMA : the journal of the American Medical

Association. 1997;278(16):1349-1356.

9. Saunders AM, Strittmatter WJ, Schmechel D, et al. Association of apolipoprotein E allele epsilon

4 with late-onset familial and sporadic Alzheimer's disease. Neurology. 1993;43(8):1467-1472.

10. Anstey KJ, von Sanden C, Salim A, O'Kearney R. Smoking as a risk factor for dementia and

cognitive decline: a meta-analysis of prospective studies. American journal of epidemiology.

2007;166(4):367-378.

Page 70: Omonigho A. Michael Bubu

58

11. Pendlebury ST, Rothwell PM. Prevalence, incidence, and factors associated with pre-stroke and

post-stroke dementia: a systematic review and meta-analysis. Lancet neurology.

2009;8(11):1006-1018.

12. Rusanen M, Kivipelto M, Quesenberry CP, Jr., Zhou J, Whitmer RA. Heavy smoking in midlife

and long-term risk of Alzheimer disease and vascular dementia. Archives of internal medicine.

2011;171(4):333-339.

13. Ohara T, Doi Y, Ninomiya T, et al. Glucose tolerance status and risk of dementia in the

community: the Hisayama study. Neurology. 2011;77(12):1126-1134.

14. Reitz C, Brayne C, Mayeux R. Epidemiology of Alzheimer disease. Nature reviews. Neurology.

2011;7(3):137-152.

15. Wu W, Brickman AM, Luchsinger J, et al. The brain in the age of old: the hippocampal formation

is targeted differentially by diseases of late life. Annals of neurology. 2008;64(6):698-706.

16. Fitzpatrick AL, Kuller LH, Lopez OL, et al. Midlife and late-life obesity and the risk of dementia:

cardiovascular health study. Archives of neurology. 2009;66(3):336-342.

17. Kivipelto M, Ngandu T, Fratiglioni L, et al. Obesity and vascular risk factors at midlife and the

risk of dementia and Alzheimer disease. Archives of neurology. 2005;62(10):1556-1560.

18. Raji CA, Ho AJ, Parikshak NN, et al. Brain structure and obesity. Human brain mapping.

2010;31(3):353-364.

19. Whitmer RA, Gustafson DR, Barrett-Connor E, Haan MN, Gunderson EP, Yaffe K. Central

obesity and increased risk of dementia more than three decades later. Neurology.

2008;71(14):1057-1064.

20. Xu WL, Atti AR, Gatz M, Pedersen NL, Johansson B, Fratiglioni L. Midlife overweight and

obesity increase late-life dementia risk: a population-based twin study. Neurology.

2011;76(18):1568-1574.

Page 71: Omonigho A. Michael Bubu

59

21. Solomon A, Kivipelto M, Wolozin B, Zhou J, Whitmer RA. Midlife serum cholesterol and

increased risk of Alzheimer's and vascular dementia three decades later. Dementia and geriatric

cognitive disorders. 2009;28(1):75-80.

22. Debette S, Seshadri S, Beiser A, et al. Midlife vascular risk factor exposure accelerates structural

brain aging and cognitive decline. Neurology. 2011;77(5):461-468.

23. Launer LJ, Ross GW, Petrovitch H, et al. Midlife blood pressure and dementia: the Honolulu-

Asia aging study. Neurobiol Aging. 2000;21(1):49-55.

24. Ninomiya T, Ohara T, Hirakawa Y, et al. Midlife and late-life blood pressure and dementia in

Japanese elderly: the Hisayama study. Hypertension. 2011;58(1):22-28.

25. Larson EB, Wang L, Bowen JD, et al. Exercise is associated with reduced risk for incident

dementia among persons 65 years of age and older. Annals of internal medicine. 2006;144(2):73-

81.

26. Laurin D, Verreault R, Lindsay J, MacPherson K, Rockwood K. Physical activity and risk of

cognitive impairment and dementia in elderly persons. Archives of neurology. 2001;58(3):498-

504.

27. Mortimer JA. Physical activity and cognitive function in Alzheimer disease. JAMA : the journal

of the American Medical Association. 2009;301(3):272-273; author reply 273-274.

28. Willis BL, Gao A, Leonard D, Defina LF, Berry JD. Midlife fitness and the development of

chronic conditions in later life. Archives of internal medicine. 2012;172(17):1333-1340.

29. Evans DA, Bennett DA, Wilson RS, et al. Incidence of Alzheimer disease in a biracial urban

community: relation to apolipoprotein E allele status. Archives of neurology. 2003;60(2):185-189.

30. Stern Y, Gurland B, Tatemichi TK, Tang MX, Wilder D, Mayeux R. Influence of education and

occupation on the incidence of Alzheimer's disease. JAMA : the journal of the American Medical

Association. 1994;271(13):1004-1010.

31. Graves AB, Mortimer JA, Larson EB, Wenzlow A, Bowen JD, McCormick WC. Head

circumference as a measure of cognitive reserve. Association with severity of impairment in

Page 72: Omonigho A. Michael Bubu

60

Alzheimer's disease. The British journal of psychiatry : the journal of mental science.

1996;169(1):86-92.

32. Mortimer JA, Snowdon DA, Markesbery WR. Small head circumference is associated with less

education in persons at risk for Alzheimer disease in later life. Alzheimer disease and associated

disorders. 2008;22(3):249-254.

33. Mortimer JA, French LR, Hutton JT, Schuman LM. Head injury as a risk factor for Alzheimer's

disease. Neurology. 1985;35(2):264-267.

34. Mortimer JA, van Duijn CM, Chandra V, et al. Head trauma as a risk factor for Alzheimer's

disease: a collaborative re-analysis of case-control studies. EURODEM Risk Factors Research

Group. Int J Epidemiol. 1991;20 Suppl 2:S28-35.

35. Mortimer JA. Brain reserve and the clinical expression of Alzheimer's disease. Geriatrics.

1997;52 Suppl 2:S50-53.

36. Hall CB, Lipton RB, Sliwinski M, Katz MJ, Derby CA, Verghese J. Cognitive activities delay

onset of memory decline in persons who develop dementia. Neurology. 2009;73(5):356-361.

37. Wilson RS, Bennett DA, Bienias JL, et al. Cognitive activity and incident AD in a population-

based sample of older persons. Neurology. 2002;59(12):1910-1914.

38. Wilson RS, Mendes De Leon CF, Barnes LL, et al. Participation in cognitively stimulating

activities and risk of incident Alzheimer disease. JAMA : the journal of the American Medical

Association. 2002;287(6):742-748.

39. Ballard C, Gauthier S, Corbett A, Brayne C, Aarsland D, Jones E. Alzheimer's disease. Lancet.

2011;377(9770):1019-1031.

40. Peter-Derex L, Yammine P, Bastuji H, Croisile B. Sleep and Alzheimer's disease. Sleep medicine

reviews. 2014.

41. Cooke JR, Ancoli-Israel S. Normal and abnormal sleep in the elderly. Handbook of clinical

neurology. 2011;98:653-665.

Page 73: Omonigho A. Michael Bubu

61

42. 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 : the journal of the

American Medical Association. 2000;284(7):861-868.

43. Bliwise DL. Sleep in normal aging and dementia. Sleep. 1993;16(1):40-81.

44. Touitou Y. Human aging and melatonin. Clinical relevance. Experimental gerontology.

2001;36(7):1083-1100.

45. Prinz PN, Peskind ER, Vitaliano PP, et al. Changes in the sleep and waking EEGs of

nondemented and demented elderly subjects. Journal of the American Geriatrics Society.

1982;30(2):86-93.

46. Prinz PN, Vitaliano PP, Vitiello MV, et al. Sleep, EEG and mental function changes in senile

dementia of the Alzheimer's type. Neurobiology of aging. 1982;3(4):361-370.

47. Vitiello MV, Prinz PN, Williams DE, Frommlet MS, Ries RK. Sleep disturbances in patients with

mild-stage Alzheimer's disease. Journal of gerontology. 1990;45(4):M131-138.

48. Hoch CC, Reynolds CF, 3rd, Kupfer DJ, Houck PR, Berman SR, Stack JA. Sleep-disordered

breathing in normal and pathologic aging. The Journal of clinical psychiatry. 1986;47(10):499-

503.

49. Mant A, Saunders NA, Eyland AE, Pond CD, Chancellor AH, Webster IW. Sleep-related

respiratory disturbance and dementia in elderly females. Journal of gerontology.

1988;43(5):M140-144.

50. McCurry SM, Reynolds CF, Ancoli-Israel S, Teri L, Vitiello MV. Treatment of sleep disturbance

in Alzheimer's disease. Sleep medicine reviews. 2000;4(6):603-628.

51. Moran M, Lynch CA, Walsh C, Coen R, Coakley D, Lawlor BA. Sleep disturbance in mild to

moderate Alzheimer's disease. Sleep medicine. 2005;6(4):347-352.

52. Vitiello MV, Prinz PN. Alzheimer's disease. Sleep and sleep/wake patterns. Clinics in geriatric

medicine. 1989;5(2):289-299.

Page 74: Omonigho A. Michael Bubu

62

53. Bianchetti A, Scuratti A, Zanetti O, et al. Predictors of mortality and institutionalization in

Alzheimer disease patients 1 year after discharge from an Alzheimer dementia unit. Dementia

(Basel, Switzerland). 1995;6(2):108-112.

54. Pollak CP, Perlick D. Sleep problems and institutionalization of the elderly. Journal of geriatric

psychiatry and neurology. 1991;4(4):204-210.

55. Pollak CP, Perlick D, Linsner JP, Wenston J, Hsieh F. Sleep problems in the community elderly

as predictors of death and nursing home placement. Journal of community health.

1990;15(2):123-135.

56. Singer C, Tractenberg RE, Kaye J, et al. A multicenter, placebo-controlled trial of melatonin for

sleep disturbance in Alzheimer's disease. Sleep. 2003;26(7):893-901.

57. Meeks TW, Ropacki SA, Jeste DV. The neurobiology of neuropsychiatric syndromes in

dementia. Current opinion in psychiatry. 2006;19(6):581-586.

58. Ju YE, Lucey BP, Holtzman DM. Sleep and Alzheimer disease pathology-a bidirectional

relationship. Nature reviews. Neurology. 2013.

59. Rauchs G, Schabus M, Parapatics S, et al. Is there a link between sleep changes and memory in

Alzheimer's disease? Neuroreport. 2008;19(11):1159-1162.

60. Rauchs G HC, Bertran F, Desgranges B, Eustache F. . Sleep and episodic memory: a review of

the literature in young healthy subjects and potential links between sleep changes and memory

impairment observed during aging and Alzheimer’s disease. . Rev Neurol (Paris) 2010;.

2010;166:(873e):81.

61. Bloom HG, Ahmed I, Alessi CA, et al. Evidence-based recommendations for the assessment and

management of sleep disorders in older persons. Journal of the American Geriatrics Society.

2009;57(5):761-789.

62. Palma JA, Urrestarazu E, Iriarte J. Sleep loss as risk factor for neurologic disorders: a review.

Sleep medicine. 2013;14(3):229-236.

Page 75: Omonigho A. Michael Bubu

63

63. Slats D, Claassen JA, Verbeek MM, Overeem S. Reciprocal interactions between sleep, circadian

rhythms and Alzheimer's disease: focus on the role of hypocretin and melatonin. Ageing research

reviews. 2013;12(1):188-200.

64. Spira AP, Chen-Edinboro LP, Wu MN, Yaffe K. Impact of sleep on the risk of cognitive decline

and dementia. Current opinion in psychiatry. 2014;27(6):478-483.

65. Pai M, McCulloch M, Gorman JD, et al. Systematic reviews and meta-analyses: an illustrated,

step-by-step guide. The National medical journal of India. 2004;17(2):86-95.

66. Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and

meta-analyses: the PRISMA statement. BMJ (Clinical research ed.). 2009;339:b2535.

67. Higgins JPT GSe. Cochrane handbook for systematic reviews of interventions. Version 5.1.0

[updated March 2011]. . http://www.cochranehandbook.org [accessed 11.23.14]. 2011.

68. Buysse DJ, Reynolds CF, 3rd, Monk TH, Berman SR, Kupfer DJ. The Pittsburgh Sleep Quality

Index: a new instrument for psychiatric practice and research. Psychiatry research.

1989;28(2):193-213.

69. Ancoli-Israel S, Cole R, Alessi C, Chambers M, Moorcroft W, Pollak CP. The role of actigraphy

in the study of sleep and circadian rhythms. Sleep. 2003;26(3):342-392.

70. Medicine. AAoS. The international classification of sleep disorders, revised: diagnostic and

coding manual. Chicago, USA: . American Academy of Sleep Medicine, ISBN 0-9657220-1-5;

2001. 2001.

71. Kling RN, McLeod CB, Koehoorn M. Sleep problems and workplace injuries in Canada. Sleep.

2010;33(5):611-618.

72. Uehli K, Mehta AJ, Miedinger D, et al. Sleep problems and work injuries: a systematic review

and meta-analysis. Sleep medicine reviews. 2014;18(1):61-73.

73. Bhui K, Stansfeld S, Hull S, Priebe S, Mole F, Feder G. Ethnic variations in pathways to and use

of specialist mental health services in the UK. Systematic review. The British journal of

psychiatry : the journal of mental science. 2003;182:105-116.

Page 76: Omonigho A. Michael Bubu

64

74. Dinos S, Khoshaba B, Ashby D, et al. A systematic review of chronic fatigue, its syndromes and

ethnicity: prevalence, severity, co-morbidity and coping. Int J Epidemiol. 2009;38(6):1554-1570.

75. Wells G SB, O’Connell D, Peterson J,Welch V, Losos M, et al. . The Newcastle-Ottawa scale

(NOS) for assessing the quality of nonrandomised studies in metaanalyses,.

http://www.ohri.ca/programs/clinical_epidemiology/oxford.asp/; 2011 [accessed 11.25.14].

2011.

76. D. P. Personal communication 2011. 2011.

77. Popay J RH, Sowden A et al. Developing guidance on the conduct of narrative synthesis in

systematic reviews. . J Epidemiol Community Health 2005;59:A7.

78. Moher D, Liberati A, Tetzlaff J, Altman DG, Grp P. Preferred Reporting Items for Systematic

Reviews and Meta-Analyses: The PRISMA Statement. Journal of Clinical Epidemiology.

2009;62(10):1006-1012.

79. Blackwell T, Yaffe K, Ancoli-Israel S, et al. Poor sleep is associated with impaired cognitive

function in older women: the study of osteoporotic fractures. The journals of gerontology. Series

A, Biological sciences and medical sciences. 2006;61(4):405-410.

80. Craig D, Hart DJ, Passmore AP. Genetically increased risk of sleep disruption in Alzheimer's

disease. Sleep. 2006;29(8):1003-1007.

81. Djonlagic I, Guo M, Matteis P, Carusona A, Stickgold R, Malhotra A. Untreated sleep-disordered

breathing: links to aging-related decline in sleep-dependent memory consolidation. PloS one.

2014;9(1):e85918.

82. Fetveit A, Bjorvatn B. Sleep duration during the 24-hour day is associated with the severity of

dementia in nursing home patients. International journal of geriatric psychiatry.

2006;21(10):945-950.

83. Gottlieb DJ, DeStefano AL, Foley DJ, et al. APOE epsilon4 is associated with obstructive sleep

apnea/hypopnea: the Sleep Heart Health Study. Neurology. 2004;63(4):664-668.

Page 77: Omonigho A. Michael Bubu

65

84. Guarnieri B, Adorni F, Musicco M, et al. Prevalence of Sleep Disturbances in Mild Cognitive

Impairment and Dementing Disorders: A Multicenter Italian Clinical Cross-Sectional Study on

431 Patients. Dementia and geriatric cognitive disorders. 2012;33(1):50-58.

85. Ju YE, McLeland JS, Toedebusch CD, et al. Sleep quality and preclinical Alzheimer disease.

JAMA neurology. 2013;70(5):587-593.

86. Kadotani H, Kadotani T, Young T, et al. Association between apolipoprotein E epsilon4 and

sleep-disordered breathing in adults. JAMA : the journal of the American Medical Association.

2001;285(22):2888-2890.

87. Merlino G, Piani A, Gigli GL, et al. Daytime sleepiness is associated with dementia and cognitive

decline in older Italian adults: A population-based study. Sleep medicine. 2010;11(4):372-377.

88. Nebes RD, Buysse DJ, Halligan EM, Houck PR, Monk TH. Self-reported sleep quality predicts

poor cognitive performance in healthy older adults. The journals of gerontology. Series B,

Psychological sciences and social sciences. 2009;64(2):180-187.

89. Oosterman JM, van Someren EJ, Vogels RL, Van Harten B, Scherder EJ. Fragmentation of the

rest-activity rhythm correlates with age-related cognitive deficits. Journal of sleep research.

2009;18(1):129-135.

90. Osorio RS, Ayappa I, Mantua J, et al. The interaction between sleep-disordered breathing and

apolipoprotein E genotype on cerebrospinal fluid biomarkers for Alzheimer's disease in

cognitively normal elderly individuals. Neurobiol Aging. 2014;35(6):1318-1324.

91. Pat-Horenczyk R, Klauber MR, Shochat T, Ancoli-Israel S. Hourly profiles of sleep and

wakefulness in severely versus mild-moderately demented nursing home patients. Aging (Milan,

Italy). 1998;10(4):308-315.

92. Pistacchi M, Gioulis M, Contin F, Sanson F, Marsala SZ. Sleep disturbance and cognitive

disorder: epidemiological analysis in a cohort of 263 patients. Neurological sciences : official

journal of the Italian Neurological Society and of the Italian Society of Clinical Neurophysiology.

2014.

Page 78: Omonigho A. Michael Bubu

66

93. Saint Martin M, Sforza E, Barthelemy JC, Thomas-Anterion C, Roche F. Does subjective sleep

affect cognitive function in healthy elderly subjects? The Proof cohort. Sleep medicine.

2012;13(9):1146-1152.

94. Spira AP, Gamaldo AA, An Y, et al. Self-reported sleep and beta-amyloid deposition in

community-dwelling older adults. JAMA neurology. 2013;70(12):1537-1543.

95. Yesavage JA, Noda A, Hernandez B, et al. Circadian clock gene polymorphisms and sleep-wake

disturbance in Alzheimer disease. The American journal of geriatric psychiatry : official journal

of the American Association for Geriatric Psychiatry. 2011;19(7):635-643.

96. Chen PC, Wu D, Chen CC, Chi NF, Kang JH, Hu CJ. Rapid eye movement sleep atonia in

patients with cognitive impairment. Journal of the neurological sciences. 2011;305(1-2):34-37.

97. Fronczek R, van Geest S, Frolich M, et al. Hypocretin (orexin) loss in Alzheimer's disease.

Neurobiology of aging. 2012;33(8):1642-1650.

98. Hita-Yanez E, Atienza M, Cantero JL. Polysomnographic and subjective sleep markers of mild

cognitive impairment. Sleep. 2013;36(9):1327-1334.

99. Hita-Yanez E, Atienza M, Gil-Neciga E, Cantero JL. Disturbed sleep patterns in elders with mild

cognitive impairment: the role of memory decline and ApoE epsilon4 genotype. Current

Alzheimer research. 2012;9(3):290-297.

100. Hot P, Rauchs G, Bertran F, et al. Changes in sleep theta rhythm are related to episodic memory

impairment in early Alzheimer's disease. Biological psychology. 2011;87(3):334-339.

101. Liu RY, Zhou JN, van Heerikhuize J, Hofman MA, Swaab DF. Decreased melatonin levels in

postmortem cerebrospinal fluid in relation to aging, Alzheimer's disease, and apolipoprotein E-

epsilon4/4 genotype. The Journal of clinical endocrinology and metabolism. 1999;84(1):323-327.

102. Naismith SL, Hickie IB, Terpening Z, et al. Circadian Misalignment and Sleep Disruption in Mild

Cognitive Impairment. Journal of Alzheimers Disease. 2014;38(4):857-866.

103. O'Hara R, Schroder CM, Kraemer HC, et al. Nocturnal sleep apnea/hypopnea is associated with

lower memory performance in APOE epsilon4 carriers. Neurology. 2005;65(4):642-644.

Page 79: Omonigho A. Michael Bubu

67

104. Pearson VE, Allen RP, Dean T, Gamaldo CE, Lesage SR, Earley CJ. Cognitive deficits

associated with restless legs syndrome (RLS). Sleep medicine. 2006;7(1):25-30.

105. Sanchez-Espinosa MP, Atienza M, Cantero JL. Sleep deficits in mild cognitive impairment are

related to increased levels of plasma amyloid-beta and cortical thinning. NeuroImage.

2014;98:395-404.

106. Schmidt FM, Kratzsch J, Gertz HJ, et al. Cerebrospinal fluid melanin-concentrating hormone

(MCH) and hypocretin-1 (HCRT-1, orexin-A) in Alzheimer's disease. PloS one.

2013;8(5):e63136.

107. Slats D, Claassen JA, Lammers GJ, Melis RJ, Verbeek MM, Overeem S. Association between

hypocretin-1 and amyloid-beta42 cerebrospinal fluid levels in Alzheimer's disease and healthy

controls. Current Alzheimer research. 2012;9(10):1119-1125.

108. Anderson KN, Catt M, Collerton J, et al. Assessment of sleep and circadian rhythm disorders in

the very old: the Newcastle 85+ Cohort Study. Age and ageing. 2014;43(1):57-63.

109. Benito-Leon J, Louis ED, Villarejo-Galende A, Romero JP, Bermejo-Pareja F. Long sleep

duration in elders without dementia increases risk of dementia mortality (NEDICES). Neurology.

2014;83(17):1530-1537.

110. Bidzan L, Grabowski J, Dutczak B, Bidzan M. [Sleep disorders in the preclinical period of the

Alzheimer's disease]. Psychiatria polska. 2011;45(6):851-860.

111. Blackwell T, Yaffe K, Laffan A, et al. Associations of objectively and subjectively measured

sleep quality with subsequent cognitive decline in older community-dwelling men: the MrOS

sleep study. Sleep. 2014;37(4):655-663.

112. Chang WP, Liu ME, Chang WC, et al. Sleep apnea and the risk of dementia: a population-based

5-year follow-up study in Taiwan. PloS one. 2013;8(10):e78655.

113. Cohen-Zion M SC, Marler, Shochat T, Kripke DF, Ancoli-Israel S. . Changes in cognitive

function associated with sleep disordered breathing in older people. . J Am Geriatr Soc. 2001;.

2001;49((12):):1622-1627.

Page 80: Omonigho A. Michael Bubu

68

114. Cricco M, Simonsick EM, Foley DJ. The impact of insomnia on cognitive functioning in older

adults. Journal of the American Geriatrics Society. 2001;49(9):1185-1189.

115. Foley D, Monjan A, Masaki K, et al. Daytime sleepiness is associated with 3-year incident

dementia and cognitive decline in older Japanese-American men. Journal of the American

Geriatrics Society. 2001;49(12):1628-1632.

116. Hahn EA, Wang HX, Andel R, Fratiglioni L. A change in sleep pattern may predict Alzheimer

disease. The American journal of geriatric psychiatry : official journal of the American

Association for Geriatric Psychiatry. 2014;22(11):1262-1271.

117. Jelicic M, Bosma H, Ponds RW, Van Boxtel MP, Houx PJ, Jolles J. Subjective sleep problems in

later life as predictors of cognitive decline. Report from the Maastricht Ageing Study (MAAS).

International journal of geriatric psychiatry. 2002;17(1):73-77.

118. Keage HA, Banks S, Yang KL, Morgan K, Brayne C, Matthews FE. What sleep characteristics

predict cognitive decline in the elderly? Sleep medicine. 2012;13(7):886-892.

119. 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(7):1027-

1032.

120. Lim AS, Yu L, Kowgier M, Schneider JA, Buchman AS, Bennett DA. Modification of the

Relationship of the Apolipoprotein E epsilon4 Allele to the Risk of Alzheimer Disease and

Neurofibrillary Tangle Density by Sleep. JAMA neurology. 2013;70(12):1544-1551.

121. Osorio RS, Pirraglia E, Aguera-Ortiz LF, et al. Greater risk of Alzheimer's disease in older adults

with insomnia. Journal of the American Geriatrics Society. 2011;59(3):559-562.

122. Song Y, Blackwell T, Yaffe K, Ancoli-Israel S, Redline S, Stone KL. Relationships Between

Sleep Stages and Changes in Cognitive Function in Older Men: The MrOS Sleep Study. Sleep.

2014.

123. Sterniczuk R, Theou O, Rusak B, Rockwood K. Sleep disturbance is associated with incident

dementia and mortality. Current Alzheimer research. 2013;10(7):767-775.

Page 81: Omonigho A. Michael Bubu

69

124. Tranah GJ, Blackwell T, Stone KL, et al. Circadian activity rhythms and risk of incident dementia

and mild cognitive impairment in older women. Annals of neurology. 2011;70(5):722-732.

125. 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

disease and associated disorders. 2006;20(1):41-48.

126. Virta JJ, Heikkila K, Perola M, et al. Midlife sleep characteristics associated with late life

cognitive function. Sleep. 2013;36(10):1533-1541, 1541A.

127. Walsh CM, Blackwell T, Tranah GJ, et al. Weaker Circadian Activity Rhythms are Associated

with Poorer Executive Function in Older Women. Sleep. 2014.

128. 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(6):613-619.

129. Yesavage JA, Friedman L, Kraemer H, et al. Sleep/wake disruption in Alzheimer's disease:

APOE status and longitudinal course. Journal of geriatric psychiatry and neurology.

2004;17(1):20-24.

130. Ancoli-Israel S, Palmer BW, Cooke JR, et al. Cognitive effects of treating obstructive sleep apnea

in Alzheimer's disease: a randomized controlled study. Journal of the American Geriatrics

Society. 2008;56(11):2076-2081.

131. Asayama K, Yamadera H, Ito T, Suzuki H, Kudo Y, Endo S. Double blind study of melatonin

effects on the sleep-wake rhythm, cognitive and non-cognitive functions in Alzheimer type

dementia. Journal of Nippon Medical School = Nippon Ika Daigaku zasshi. 2003;70(4):334-341.

132. Kushida CA, Nichols DA, Holmes TH, et al. Effects of continuous positive airway pressure on

neurocognitive function in obstructive sleep apnea patients: The Apnea Positive Pressure Long-

term Efficacy Study (APPLES). Sleep. 2012;35(12):1593-1602.

133. Ooms S, Overeem S, Besse K, Rikkert MO, Verbeek M, Claassen JA. Effect of 1 night of total

sleep deprivation on cerebrospinal fluid beta-amyloid 42 in healthy middle-aged men: a

randomized clinical trial. JAMA neurology. 2014;71(8):971-977.

Page 82: Omonigho A. Michael Bubu

70

134. Craig LA, McDonald RJ. Chronic disruption of circadian rhythms impairs hippocampal memory

in the rat. Brain research bulletin. 2008;76(1-2):141-151.

135. Deng YQ, Xu GG, Duan P, Zhang Q, Wang JZ. Effects of melatonin on wortmannin-induced tau

hyperphosphorylation. Acta pharmacologica Sinica. 2005;26(5):519-526.

136. Di Meco A, Joshi YB, Pratico D. Sleep deprivation impairs memory, tau metabolism, and

synaptic integrity of a mouse model of Alzheimer's disease with plaques and tangles.

Neurobiology of aging. 2014;35(8):1813-1820.

137. Jyoti A, Plano A, Riedel G, Platt B. EEG, activity, and sleep architecture in a transgenic

AbetaPPswe/PSEN1A246E Alzheimer's disease mouse. Journal of Alzheimer's disease : JAD.

2010;22(3):873-887.

138. Kang JE, Lim MM, Bateman RJ, et al. Amyloid-beta dynamics are regulated by orexin and the

sleep-wake cycle. Science (New York, N.Y.). 2009;326(5955):1005-1007.

139. Kaushal N, Ramesh V, Gozal D. Human apolipoprotein E4 targeted replacement in mice reveals

increased susceptibility to sleep disruption and intermittent hypoxia. American journal of

physiology. Regulatory, integrative and comparative physiology. 2012;303(1):R19-29.

140. Lahiri DK, Chen D, Ge YW, Bondy SC, Sharman EH. Dietary supplementation with melatonin

reduces levels of amyloid beta-peptides in the murine cerebral cortex. Journal of pineal research.

2004;36(4):224-231.

141. Li L, Zhang X, Yang D, Luo G, Chen S, Le W. Hypoxia increases Abeta generation by altering

beta- and gamma-cleavage of APP. Neurobiology of aging. 2009;30(7):1091-1098.

142. Mander BA, Rao V, Lu B, et al. Prefrontal atrophy, disrupted NREM slow waves and impaired

hippocampal-dependent memory in aging. Nature neuroscience. 2013;16(3):357-364.

143. Olcese JM, Cao C, Mori T, et al. Protection against cognitive deficits and markers of

neurodegeneration by long-term oral administration of melatonin in a transgenic model of

Alzheimer disease. Journal of pineal research. 2009;47(1):82-96.

Page 83: Omonigho A. Michael Bubu

71

144. Pappolla M, Bozner P, Soto C, et al. Inhibition of Alzheimer beta-fibrillogenesis by melatonin.

The Journal of biological chemistry. 1998;273(13):7185-7188.

145. Quinn J, Kulhanek D, Nowlin J, et al. Chronic melatonin therapy fails to alter amyloid burden or

oxidative damage in old Tg2576 mice: implications for clinical trials. Brain research.

2005;1037(1-2):209-213.

146. Rothman SM, Herdener N, Frankola KA, Mughal MR, Mattson MP. Chronic mild sleep

restriction accentuates contextual memory impairments, and accumulations of cortical Abeta and

pTau in a mouse model of Alzheimer's disease. Brain research. 2013;1529:200-208.

147. Shiota S, Takekawa H, Matsumoto SE, et al. Chronic intermittent hypoxia/reoxygenation

facilitate amyloid-beta generation in mice. Journal of Alzheimer's disease : JAD. 2013;37(2):325-

333.

148. Sun X, He G, Qing H, et al. Hypoxia facilitates Alzheimer's disease pathogenesis by up-

regulating BACE1 gene expression. Proceedings of the National Academy of Sciences of the

United States of America. 2006;103(49):18727-18732.

149. Zhang X, Zhou K, Wang R, et al. Hypoxia-inducible factor 1alpha (HIF-1alpha)-mediated

hypoxia increases BACE1 expression and beta-amyloid generation. The Journal of biological

chemistry. 2007;282(15):10873-10880.

150. Cohen-Zion M, Stepnowsky C, Marler, Shochat T, Kripke DF, Ancoli-Israel S. Changes in

cognitive function associated with sleep disordered breathing in older people. Journal of the

American Geriatrics Society. 2001;49(12):1622-1627.

151. Netzer NC, Stoohs RA, Netzer CM, Clark K, Strohl KP. Using the Berlin Questionnaire to

identify patients at risk for the sleep apnea syndrome. Annals of internal medicine.

1999;131(7):485-491.

152. Reisberg B, Borenstein J, Salob SP, Ferris SH, Franssen E, Georgotas A. Behavioral symptoms in

Alzheimer's disease: phenomenology and treatment. The Journal of clinical psychiatry. 1987;48

Suppl:9-15.

Page 84: Omonigho A. Michael Bubu

72

153. Levine DW, Kripke DF, Kaplan RM, et al. Reliability and validity of the Women's Health

Initiative Insomnia Rating Scale. Psychological assessment. 2003;15(2):137-148.

154. Allen RP PD, Hening WA, Trenkwalder C, Walters AS, Montplaisi J, . Restless Legs Syndrome

Diagnosis and Epidemiology Workshop at the National Institutes of Health, International Restless

Legs Syndrome Study Group: Restless legs syndrome: diagnostic criteria, special considerations,

and epidemiology. A report from the restless legs syndrome diagnosis and epidemiology

workshop at the National Institutes of Health. Sleep medicine. 2003;4:101–119.

155. Rothdach AJ, Trenkwalder C, Haberstock J, Keil U, Berger K. Prevalence and risk factors of RLS

in an elderly population: the MEMO study. Memory and Morbidity in Augsburg Elderly.

Neurology. 2000;54(5):1064-1068.

156. WHO. World Health Organization International statistical classification of diseases and related

health problems. http://www.who.int/classification/icd/en.

157. APA. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders.

4th ed. Text revised. . Washington, DC: American Psychiatric Association; 2000. 2000.

158. McKhann G DD, Folstein M, Katzman R, Price D, Stadlan EM Clinical diagnosis of

Alzheimer’s disease: report of the NINCDS-ADRDA work group under the auspices of

Department of Health and Human Services Task Force on Alzheimer’s Disease. . Neurology 34:.

1984;34:939-944.

159. Petersen RC, Smith GE, Waring SC, Ivnik RJ, Tangalos EG, Kokmen E. Mild cognitive

impairment: clinical characterization and outcome. Archives of neurology. 1999;56(3):303-308.

160. Folstein MF, Folstein SE, McHugh PR. "Mini-mental state". A practical method for grading the

cognitive state of patients for the clinician. J Psychiatr Res. 1975;12(3):189-198.

161. Delis DC, Kramer JH, Kaplan E, &, Ober BA. California Verbal Learning Test: Second Edition.

San Antonio, TX Psychological Corporation. 2000.

162. Shin MS, Park SY, Park SR, Seol SH, Kwon JS. Clinical and empirical applications of the Rey-

Osterrieth Complex Figure Test. Nature protocols. 2006;1(2):892-899.

Page 85: Omonigho A. Michael Bubu

73

163. Wechsler D. The Measurement of Adult Intelligence. Baltimore (MD). Williams & Witkins. 1939:

p. 229.

164. Yesavage JA, Brink TL, Rose TL, et al. Development and validation of a geriatric depression

screening scale: a preliminary report. J Psychiatr Res. 1982;17(1):37-49.

165. Lawton MP, Brody EM. Assessment of older people: self-maintaining and instrumental activities

of daily living. The Gerontologist. 1969;9(3):179-186.

166. Teng EL, Chui HC. The Modified Mini-Mental State (3MS) examination. The Journal of clinical

psychiatry. 1987;48(8):314-318.

167. Jack CR, Jr., Knopman DS, Jagust WJ, et al. Hypothetical model of dynamic biomarkers of the

Alzheimer's pathological cascade. Lancet neurology. 2010;9(1):119-128.

168. Mintun MA, Larossa GN, Sheline YI, et al. [11C]PIB in a nondemented population: potential

antecedent marker of Alzheimer disease. Neurology. 2006;67(3):446-452.

169. Meyer-Lindenberg A, Buckholtz JW, Kolachana B, et al. Neural mechanisms of genetic risk for

impulsivity and violence in humans. Proceedings of the National Academy of Sciences of the

United States of America. 2006;103(16):6269-6274.

170. Agnew HW, Jr., Webb WB, Williams RL. The first night effect: an EEG study of sleep.

Psychophysiology. 1966;2(3):263-266.

171. Browman CP, Cartwright RD. The first-night effect on sleep and dreams. Biological psychiatry.

1980;15(5):809-812.

172. Webb WB, Campbell SS. The first night effect revisited with age as a variable. Waking and

sleeping. 1979;3(4):319-324.

173. Locke PA, Conneally PM, Tanzi RE, Gusella JF, Haines JL. Apolipoprotein E4 allele and

Alzheimer disease: examination of allelic association and effect on age at onset in both early- and

late-onset cases. Genetic epidemiology. 1995;12(1):83-92.

174. Peacock ML, Fink JK. ApoE epsilon 4 allelic association with Alzheimer's disease: independent

confirmation using denaturing gradient gel electrophoresis. Neurology. 1994;44(2):339-341.

Page 86: Omonigho A. Michael Bubu

74

175. Poirier J. Apolipoprotein E in animal models of CNS injury and in Alzheimer's disease. Trends in

neurosciences. 1994;17(12):525-530.

176. Schmechel DE, Saunders AM, Strittmatter WJ, et al. Increased amyloid beta-peptide deposition

in cerebral cortex as a consequence of apolipoprotein E genotype in late-onset Alzheimer disease.

Proceedings of the National Academy of Sciences of the United States of America.

1993;90(20):9649-9653.

177. Strittmatter WJ, Saunders AM, Schmechel D, et al. Apolipoprotein E: high-avidity binding to

beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease.

Proceedings of the National Academy of Sciences of the United States of America.

1993;90(5):1977-1981.

178. Henderson AS, Easteal S, Jorm AF, et al. Apolipoprotein E allele epsilon 4, dementia, and

cognitive decline in a population sample. Lancet. 1995;346(8987):1387-1390.

179. Reisberg B, Ferris SH, de Leon MJ, Crook T. The Global Deterioration Scale for assessment of

primary degenerative dementia. The American journal of psychiatry. 1982;139(9):1136-1139.

180. Jin X, Shearman LP, Weaver DR, Zylka MJ, de Vries GJ, Reppert SM. A molecular mechanism

regulating rhythmic output from the suprachiasmatic circadian clock. Cell. 1999;96(1):57-68.

181. Reppert SM, Weaver DR. Coordination of circadian timing in mammals. Nature.

2002;418(6901):935-941.

182. Peyron C, Tighe DK, van den Pol AN, et al. Neurons containing hypocretin (orexin) project to

multiple neuronal systems. The Journal of neuroscience : the official journal of the Society for

Neuroscience. 1998;18(23):9996-10015.

183. Saper CB, Fuller PM, Pedersen NP, Lu J, Scammell TE. Sleep state switching. Neuron.

2010;68(6):1023-1042.

184. Bhat RV, Budd Haeberlein SL, Avila J. Glycogen synthase kinase 3: a drug target for CNS

therapies. Journal of neurochemistry. 2004;89(6):1313-1317.

Page 87: Omonigho A. Michael Bubu

75

185. Bhat RV, Budd SL. GSK3beta signalling: casting a wide net in Alzheimer's disease. Neuro-

Signals. 2002;11(5):251-261.

186. Li X, Bijur GN, Jope RS. Glycogen synthase kinase-3beta, mood stabilizers, and neuroprotection.

Bipolar disorders. 2002;4(2):137-144.

187. Pei JJ BE, Braak H, Grundke-Iqbal I, Iqbal K, Winblad B, et al. . Distribution of active glycogen

synthase kinase 3beta (GSK- 3beta) in brains staged for Alzheimer disease neurofibrillary

changes. . J Neuropathol Exp Neurol 1999; . 1999;58::1010–1019.

188. Matsubara E, Bryant-Thomas T, Pacheco Quinto J, et al. Melatonin increases survival and

inhibits oxidative and amyloid pathology in a transgenic model of Alzheimer's disease. Journal of

neurochemistry. 2003;85(5):1101-1108.

189. Cummings JL BD. Dementia: A Clinical Approach, 2nd Ed. Boston: Butterworth, . 1992:pp 1–

17.

190. Association. AP. Diagnostic and Statistical Manual of Mental Disorders, 3rd Ed., Revised.

Washington, DC: . American Psychiatric Association,. 1987.

191. Bliwise DL. Is sleep apnea a cause of reversible dementia in old age? Journal of the American

Geriatrics Society. 1996;44(11):1407-1409.

192. Redline S, Strauss ME, Adams N, et al. Neuropsychological function in mild sleep-disordered

breathing. Sleep. 1997;20(2):160-167.

193. Ancoli-Israel S, Kripke DF, Klauber MR, Mason WJ, Fell R, Kaplan O. Sleep-disordered

breathing in community-dwelling elderly. Sleep. 1991;14(6):486-495.

194. Tison F, Crochard A, Leger D, Bouee S, Lainey E, El Hasnaoui A. Epidemiology of restless legs

syndrome in French adults: a nationwide survey: the INSTANT Study. Neurology.

2005;65(2):239-246.

195. Boeve BF, Molano JR, Ferman TJ, et al. Validation of the Mayo Sleep Questionnaire to screen

for REM sleep behavior disorder in an aging and dementia cohort. Sleep medicine.

2011;12(5):445-453.

Page 88: Omonigho A. Michael Bubu

76

196. Rose KM, Beck C, Tsai PF, et al. Sleep disturbances and nocturnal agitation behaviors in older

adults with dementia. Sleep. 2011;34(6):779-786.

197. Allen RP, Kushida CA, Atkinson MJ. Factor analysis of the International Restless Legs

Syndrome Study Group's scale for restless legs severity. Sleep medicine. 2003;4(2):133-135.

198. Beckett LA. Community-based studies of Alzheimer's disease: statistical challenges in design and

analysis. Statistics in medicine. 2000;19(11-12):1469-1480.

199. Clayton D SD, Dunn G et al. . Analysis of longitudinal binary data from multi-phase sampling. . J

R Stat Soc Series B 1998;. 1998;60::71–87.

200. Gao S, Hui SL. Estimating the incidence of dementia from two-phase sampling with non-

ignorable missing data. Statistics in medicine. 2000;19(11-12):1545-1554.

201. Van Den Berg JF, Van Rooij FJ, Vos H, et al. Disagreement between subjective and actigraphic

measures of sleep duration in a population-based study of elderly persons. Journal of sleep

research. 2008;17(3):295-302.

202. Lim DC, Veasey SC. Neural injury in sleep apnea. Curr Neurol Neurosci Rep. 2010;10(1):47-52.

203. McCoy JG, Strecker RE. The cognitive cost of sleep lost. Neurobiology of learning and memory.

2011;96(4):564-582.

204. Quan SF, Wright R, Baldwin CM, et al. Obstructive sleep apnea-hypopnea and neurocognitive

functioning in the Sleep Heart Health Study. Sleep medicine. 2006;7(6):498-507.

205. Cross RL, Kumar R, Macey PM, et al. Neural alterations and depressive symptoms in obstructive

sleep apnea patients. Sleep. 2008;31(8):1103-1109.

206. Sachs-Ericsson N, Joiner T, Plant EA, Blazer DG. The influence of depression on cognitive

decline in community-dwelling elderly persons. The American journal of geriatric psychiatry :

official journal of the American Association for Geriatric Psychiatry. 2005;13(5):402-408.

Page 89: Omonigho A. Michael Bubu

77

207. Atkins D, Best D, Briss PA, et al. Grading quality of evidence and strength of recommendations.

BMJ (Clinical research ed.). 2004;328(7454):1490.

Page 90: Omonigho A. Michael Bubu

78

Table 1.1 Literature Search Terms for the Systematic Review of Sleep and Alzheimer’s disease

Sleep Related Alzheimer's Disease Related Alzheimer's disease pathology related Study Types

Sleep (MeSH) Alzheimer's disease Dementia Amyloid beta Cross sectional

Sleep loss Cognitive Decline Amyloid beta deposition/generation Prevalence studies

Sleep deprivation Cognitive Impairment Alzheimer's disease pathogenesis Experimental studies

Sleep disruption Mild Cognitive Impairment (MeSH) Amyloid Precursor Protein Randomized Control Trials

Sleep apnea MCI Amyloid cascade hypothesis Incidence studies

Obstructive sleep apnea aMCI Inflammation Epidemiologic studies

Sleep Disordered Breathing Cognition P-Tau Case control

Excessive Daytime sleepiness Alzheimer (MeSH) Phosphorylated Tau Prospective Cohort

Sleep Disorders (MeSH) Hippocampal atrophy Retrospective cohort

Rapid Eye Movement Behavioral Disorder Apolipoprotein E Historical cohort

NREM Slow wave sleep APOE4 Closed cohort

Hypoxia Case-Comparison

Sleep Dependent Memory Nested case control

Sleep quality Case-Referent Study

Sleep function Matched case control

Wakefulness Cochrane database of systematic reviews

Circadian Rhythm (MeSH) Meta-analysis

Melatonin Synthesis

Hypocretin Research integration

Page 91: Omonigho A. Michael Bubu

79

Table 1.2: Scoring system for quality of paper of the Systematic Review of Sleep and Alzheimer's disease

· Hypotheses (maximum possible=1 star)

o The hypothesis/research question of the study not clearly described

o The hypothesis/research question of the study clearly described ¨

· Sampling (maximum possible= 2 stars)a

o Sample source and size

Ø Routine data=0

Ø Sample size: less than 50 per comparison groups

Ø Sample size: more than 50 per comparison groups ¨

o Type of sampling

Ø Consecutive patients or opportunistic

Ø Random representative of populations studied ¨

· Type of Study (maximum possible=3 stars)

o Ecological Study

o Case-control/Cross-sectional ¨

o Retrospective/Prospective Cohort ¨¨

o Randomized Control Trial/Experimental ¨¨¨

· Selection (Cohort studies) (maximum possible=4 stars)

o Representativeness of the exposed cohort

Ø Truly representative of the average distribution of sleep problems in the community ¨

Ø Somewhat representative of the average distribution of sleep problems in the community ¨

Ø Selected group of users e.g., nurses, volunteers

Ø No description of the derivation of the cohort

o Selection of the non-exposed cohort

Ø Drawn from the same community as the exposed cohort ¨

Ø Drawn from a different source

Ø No description of the derivation of the non-exposed cohort

o Length of follow up of cohorts to determine outcome

Ø Follow up period greater than 5 years ¨

Ø Follow up period less than 5 years

o Adequacy of follow up of cohorts (Likelihood to introduce bias)

Ø Complete follow up- all subjects accounted for ¨

Ø Follow up rate ≥ 50% and description of loss to follow up subjects

Ø Follow up rate < 50% and no description of loss to follow up subjects

Ø Follow up rate not mentioned

Table continues

Page 92: Omonigho A. Michael Bubu

80

Table 1.2 continued

· Selection (Case-control studies) (maximum possible=4 stars)

o Is the case definition adequate?

Ø Yes, with independent validation ¨

Ø Yes, e.g., record linkage or based on self-reports ¨

Ø No description

o Representativeness of the cases

Ø Consecutive or obviously representative series of cases ¨

Ø Potential for selection biases or not stated

o Selection of controls

Ø Community controls ¨

Ø Hospital controls

Ø No description of source

o Non response rate

Ø Same rate for both groups ¨

Ø Non respondents described

Ø Rate different and no designation

· Selection (Experimental studies) (maximum possible=4 stars)

o Appropriate description of experimental mice/population

Ø Yes, with transgenic status described ¨

Ø No description

o Appropriate description of experimental procedures

Ø Yes, with application of standard protocols ¨

Ø Standardized protocols not adhered to.

o Controls or comparison group

Ø Comparison group present ¨

Ø No controls used

o Comparability of experimental conditions

Ø Mice/population in both groups exposed to similar experimental conditions except for exposure/intervention ¨

Ø Experimental conditions not described

Ø Conditions different and no designation

Table continues

Page 93: Omonigho A. Michael Bubu

81

Table 1.2 continued

· Methodology (maximum possible=5 stars)

o Definition/ Operationalization of Alzheimer's Disease/Dementia/Cognitive Decline(2)b

Ø No standardized measures (e.g. self-reported diagnosis)

Ø Standardized Operationalized criteria (e.g. ICD-9, ICD-10, DSM-IV)* ¨

Ø Objective measures (e.g., PET scan, MRI) * ¨¨

o Definition/Operationalization of Sleep and its related abnormalities/disorders

Ø No standardized measures (e.g. self-reported diagnosis) =0

Ø Standardized Operationalized criteria/questionnaire (e.g. ICD-9, ICD-10, DSM-IV, PSQI, ESS)^ ¨

Ø Objective measures (e.g., Actigraph, Polysomnography)^ ¨¨

o Study Population (Not applicable to Animal Experimental studies)

Ø No appropriate description of study population

Ø In-patient non-population based

Ø Study population is population based ¨

· Statistical analyses and results (maximum possible=5 stars)

o Statistical analysis

Ø Statistical methods not described =0

Ø Statistical methods clearly described ¨

o Statistical outcomes

Ø Statistical outcome not clearly reported

Ø Statistical outcomes clearly reported (e.g. OR, RR, CIs) ¨

o Other explanatory variables in the analysis (e.g. confounders) (4)c

Ø No explanatory variables involved

Ø Age and/or gender controlled for ¨

Ø Age, gender, SES and risk factors (sleep related and AD related covariates) ¨¨

o Results

Ø Research questions/hypotheses not clearly answered/tested

Ø Research questions/hypotheses clearly answered/tested ¨

a: Sample size values. The sample size value of 50 participants per group has been chosen as these permits reasonable estimates of rates of CF. Not Applicable to Animal Experimental studies

b: ICD-9/10 International Classification of Disease version 9/10; DSM-IV Diagnostic Statistical Manual-IV; PQSI Pittsburg Sleep Quality Index; ESS Epworth Sleepiness Scale c: Adjustment for confounding variables. From an overview of studies on sleep and Alzheimer's Disease; Age, years of education, marital status, forced expiratory volume in one second based on spirometry, self-reported history of physician’s diagnosis of asthma, number of hours of sleep and napping, and use of benzodiazepines. Chronic obstructive pulmonary disease, Nonfatal coronary heart disease including myocardial infarction, Stroke, The ankle-brachial index calculated as the mean of the posterior tibial pressure on each side divided by the mean of the two right brachial pressures. Apolipoprotein E genotype, BMI and Depression were all-important covariates to adjust for. Not Applicable to Animal Experimental studies

* Must be independent blind assessment or made use of record linkage to receive a star

^ Must be use of secure record (eg surgical records) or made use of structured interview where blind to case/control status to receive a star

Page 94: Omonigho A. Michael Bubu

82

Table 1.3 : Quality Assessment of Cross-sectional studies

First Author, Country, Year Published

(Reference)

Hypotheses

(maximum

possible=1 star)

Sampling

(maximum

possible=2 stars)

Type of Study

(maximum

possible=3

stars)

Methodology

(maximum

possible=5 stars)

Statistical

Analysis

(maximum

possible=5 stars)

Total (maximum

possible=16

stars)

Quality Rating

Blackwell, United States, 2006 () ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 13 High

Nebes, United States, 2009 () ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 10 Medium

Saint Martin, France, 2012 () ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 10 Medium

Guarnieri, Italy, 2012 () ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 9 Medium

Moran, Ireland, 2005 () ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 9 Medium

Pistacchi, Italy, 2014 () ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 8 Low

Fetveit, Norway, 2006 () ♦

♦ ♦ ♦ ♦ ♦ ♦ 7 Low

Ju, United States, 2013 () ♦

♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 9 Medium

Spira, United States, 2013 () ♦

♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 10 Medium

Craig, Northern Ireland, UK, 2006 () ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 8 Low

Oosterman, Netherlands, 2009 () ♦

♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 9 Medium

Merlino, Italy, 2010 () ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 8 Low

Pat-Horenczyk, United States, 1998 () ♦

♦ ♦ ♦ ♦ ♦ ♦ 7 Low

Yesavage, United States & France, 2011 () ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 8 Low

Gottlieb, United States, 2004 () ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 13 High

Kadotani, United States, 2001 () ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 12 High

Osorio, United States, 2014 () ♦

♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 10 Medium

Djonlagic, United States, 2014 () ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ 8 Low

Page 95: Omonigho A. Michael Bubu

83

Table 1.4: Quality Assessment Scores of Case-control studies

First Author, Country, Year

Published (Reference)

Hypotheses

(maximum

possible=1 star)

Sampling

(maximum

possible=2 stars)

Type of Study

(maximum

possible=3

stars)

Selection (Case-

control study)

(maximum

possible=4 stars)

Methodology

(maximum

possible=5

stars)

Statistical

Analysis

(maximum

possible=5

stars)

Total

(maximum

possible=20

stars)

Quality Rating

Chen, Taiwan, 2011 () ♦

♦ ♦♦♦ ♦♦♦ ♦♦ 10 Low

Hita-Yañez, Spain, 2013 () ♦

♦ ♦♦♦ ♦♦♦ ♦♦♦ 11 Medium

Hita-Yañez, Spain, 2012 () ♦

♦ ♦♦♦ ♦♦♦♦ ♦♦♦ 12 Medium

Hot, France, 2011 () ♦

♦ ♦♦ ♦♦♦ ♦♦ 9 Low

Sanchez-Espinosa, Spain, 2014 () ♦

♦ ♦♦♦♦ ♦♦♦♦ ♦♦♦ 13 Medium

Naismith, Australia, 2014 () ♦

♦ ♦♦♦ ♦♦♦ ♦♦ 10 Low

Fronczek, Netherland, 2012 () ♦

♦ ♦♦♦ ♦♦♦♦ ♦♦ 11 Medium

Slats, Netherlands, 2012 () ♦

♦ ♦♦ ♦♦♦♦ ♦♦♦♦ 12 Medium

Schmidt, Germany, 2013 () ♦

♦ ♦♦ ♦♦♦♦ ♦♦♦ 11 Medium

Liu, Netherlands and China, 1999 () ♦ ♦ ♦ ♦♦ ♦♦♦♦ ♦♦♦ 12 Medium

O’Hara, United States, 2005 () ♦

♦ ♦♦ ♦♦♦♦ ♦♦ 10 Low

Pearson, United States, 2006 () ♦ ♦ ♦♦ ♦♦♦ ♦♦♦ 10 Low

Page 96: Omonigho A. Michael Bubu

84

Table 1.5: Quality Assessment Scores of Cohort studies/Randomized Clinical Trials

First Author, Country, Year

Published (Reference)

Hypotheses

(maximum

possible=1 star)

Sampling

(maximum

possible=2

stars)

Type of

Study

(maximum

possible=3

stars)

Selection

(Cohort studies)

(maximum

possible=4 stars)

Methodology

(maximum

possible=5 stars)

Statistical

Analysis

(maximum

possible=5

stars)

Total

(maximum

possible=20

stars)

Quality

Rating

Keage, Australia, & United Kingdom

2012 () ♦ ♦♦ ♦♦ ♦♦♦♦ ♦♦♦ ♦♦♦♦ 16 High

Tworoger, United States, 2006 () ♦ ♦♦ ♦♦ ♦♦ ♦♦♦ ♦♦♦♦ 14 Medium

Virta, Finland, 2013 () ♦ ♦♦ ♦♦ ♦♦♦♦ ♦♦♦ ♦♦♦ 15 High

Song, United States, 2014 () ♦ ♦♦ ♦♦ ♦♦♦ ♦♦♦ ♦♦♦♦ 15 High

Blackwell, United States, 2014 () ♦ ♦♦ ♦♦ ♦♦♦ ♦♦♦ ♦♦♦♦♦ 16 High

Sterniczuk, Canada, 2013 () ♦ ♦♦ ♦♦ ♦♦♦ ♦♦ ♦♦♦♦ 14 Medium

Benito-León, Spain, 2014 () ♦ ♦♦ ♦♦ ♦♦♦♦ ♦♦ ♦♦♦♦♦ 16 High

Hahn, United States, 2013 () ♦ ♦ ♦♦ ♦♦♦♦ ♦♦ ♦♦♦♦♦ 15 High

Bidzan, Poland, 2011 () ♦

♦♦ ♦♦ ♦♦ ♦♦ 9 Low

Lim, Canada, 2013 () ♦ ♦ ♦♦ ♦♦♦ ♦♦♦ ♦♦♦ 13 Medium

Ooms, Netherlands, 2014 () ♦ ♦ ♦♦♦ ♦♦ ♦♦♦♦ ♦♦♦♦ 15 High

Walsh, United States 2014 () ♦ ♦♦ ♦♦ ♦♦♦ ♦♦♦ ♦♦♦♦ 15 High

Anderson, United Kingdom, 2014 () ♦ ♦♦ ♦♦ ♦♦ ♦♦♦ ♦♦♦ 13 Medium

Tranah, United States, 2011 () ♦ ♦♦ ♦♦ ♦♦♦ ♦♦♦ ♦♦♦♦♦ 16 High

Lim, United States, 2013 () ♦ ♦♦ ♦♦ ♦♦ ♦♦♦ ♦♦♦♦♦ 15 High

Yesavage, United States, 2004 () ♦

♦♦ ♦♦ ♦♦♦♦ ♦♦ 11 Medium

Asayama, Japan, 2003 () ♦ ♦ ♦♦♦ ♦♦ ♦♦♦ ♦♦♦♦ 14 Medium

Cohen-Zion, United States, 2001 () ♦

♦♦ ♦♦ ♦♦♦ ♦♦♦ 10 Low

Chang, Taiwan, 2013 () ♦ ♦♦ ♦♦ ♦♦♦ ♦♦♦ ♦♦♦♦ 15 High

Page 97: Omonigho A. Michael Bubu

85

Yaffe, United States, 2011 () ♦ ♦ ♦♦ ♦♦ ♦♦♦♦ ♦♦♦♦♦ 15 High

Kushida, United States, 2012 () ♦ ♦ ♦♦♦ ♦♦ ♦♦♦♦ ♦♦♦♦ 15 High

Ancoli-Israel, United States, 2008 () ♦ ♦ ♦♦♦ ♦♦ ♦♦♦♦ ♦♦♦♦ 15 High

Cricco, Iceland, 2001 () ♦ ♦♦ ♦♦ ♦♦ ♦♦♦ ♦♦♦♦♦ 15 High

Jelicic, The Netherlands, 2002 () ♦ ♦♦ ♦♦ ♦♦ ♦♦♦ ♦♦♦ 13 Medium

Foley, United States, 2001 () ♦ ♦♦ ♦♦ ♦♦ ♦♦♦ ♦♦♦♦♦ 15 High

Osorio, United States, 2011 () ♦ ♦♦ ♦♦ ♦♦♦ ♦♦♦ ♦♦♦♦ 15 High

Table 1.6: Quality Assessment Scores of Experimental studies

First Author, Country, Year Published

(Reference)

Hypotheses

(maximum

possible=1

star)

Type of Study

(maximum

possible=3 stars)

Selection

(Experimental

studies)

(maximum

possible=4 stars)

Methodology

(maximum

possible=4

stars)

Statistical

Analysis

(maximum

possible=3

stars)

Total

(maximum

possible=15

stars)

Quality Rating

Rothman, United States, 2013 () ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

Di Meco, United States, 2014 () ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

Kang, United States, 2009 () ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

Mander, United States, 2013 ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦♦♦♦ ♦ ♦ 13 High

Craig, Canada, 2008 () ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

Jyoti, Scotland, UK, 2010 () ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

Quin, United States, 2005 () ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

DENG. China, 2005 () ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

Papolla, United States, 1998 () ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

Olcese, United States, 2009 () ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

Lahiri, United States, 2004 () ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

Page 98: Omonigho A. Michael Bubu

86

Shiota, Japan, 2013 () ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

Kaushal, United States, 2012 () ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

Sun, Canada, China, United States, 2006

() ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

Li, China, 2009 () ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

Zhang, China, 2007 () ♦ ♦ ♦ ♦ ♦♦♦♦ ♦♦♦♦ ♦ 13 High

Table 1.7. Sleep Depth and Cognitive/Alzheimer's Disease Measures: Descriptive Study Characteristics, and Main Findings

First Author,

Country, Year

Published (Reference)

Study Design Sample size N Study Population Sleep Depth

Assessment

Cognitive/AD Measures

Assessment Conclusion

Blackwell, United States, 2006 ()

Cross sectional N=2,242 Community-dwelling women 65 years old or older

Actigraphy

Mini-Mental State Examination (MMSE) and the Trail Making B Test (Trails B)

Objectively measured disturbed sleep was consistently related to poorer cognition, whereas total sleep time was not.

Nebes, United States, 2009 ()

Cross-sectional N=157 Participants were 65–80 years old community volunteers

Pittsburgh Sleep Quality Index

[PSQI]

Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). Test of Nonverbal Intelligence (TONI))

Sleep problems may contribute to performance variability between elderly individuals but only in certain cognitive domains.

Page 99: Omonigho A. Michael Bubu

87

Guarnieri, Italy, 2012 () Cross-sectional N= 431

Participants were patients enrolled consecutively in 10 Italian neurological centers: 204 had Alzheimer’s disease (AD), 138 mild cognitive impairment (MCI), 43 vascular dementia (VaD), 25 frontotemporal dementia and 21 Lewy body dementia (LBD) or Parkinson’s disease dementia (PDD).

PSQI, Berlin questionnaire and the International

Restless Leg Syndrome (RLS)

Study Group questionnaire

DSM-IV-TR criteria. AD and Vascular Dementia (VaD) were diagnosed respectively according to NINCS-ARDRA and NINDS-AIREN criteria. LBD and PDD, frontotemporal dementia (FTD) and MCI were diagnosed according to specific consensus guidelines

A careful clinical evaluation of sleep disorders should be performed routinely in the clinical setting of persons with cognitive decline. Instrumental supports should be used only in selected patients

Pistacchi, Italy, 2014 () Cross-sectional N= 236

236 patients (78 men and 158 women) were enrolled with different subtypes of dementia: (AD), (VaD), mixed dementia (MCI), dementia with Lewy bodies (DLB), (PDD), and frontotemporal lobar degeneration (FTLD)

PSQI, Berlin questionnaire and the International

Restless Leg Syndrome (RLS)

Study Group questionnaire

DSM-IV-TR criteria . AD and VaD were diagnosed respectively according to NINCS-ARDRA and NINDS-AIREN criteria . LBD and PDD, frontotemporal dementia (FTD) and MCI were diagnosed according to specific consensus guidelines .

Findings demonstrate that sleep disturbance was related to dementia

Fetveit, Norway, 2006 ()

Cross-sectional N=23 Nursing home residents with varying degree of dementia

Actigraphy

Grades of dementia were assessed by Mini-Mental Status Examination Score (MMSE) and Clinical Dementia Rating (CDR).

Sleep duration during the 24-h day was positively correlated with the severity of dementia in nursing home patients.

Page 100: Omonigho A. Michael Bubu

88

Ju, United States, 2013 ()

Cross-sectional N=142

Longitudinal studies of memory and aging at the Washington University Knight Alzheimer Disease Research Center (ADRC). The Adult Children Study, in which all were age 45-75 years at baseline and 50% have a parental history of sAD.

Actigraphy

Aβ42 was measured by the Alzheimer's Disease Research Center (ADRC) Biomarker Core using enzyme linked immunosorbant assay (INNOTEST; Innogenetics, Ghent, Belgium).

Amyloid deposition in the preclinical stage of AD appears to be associated with worse sleep quality, but not with changes in sleep quantity.

Spira, United States, 2013 ()

Cross-sectional N= 70

Participants were adults (mean age, 76 [range, 53-91] years) from the neuroimaging sub-study of the Baltimore Longitudinal Study of Aging, a normative aging study.

Self-reported sleep variables: The 5-item Women’s

Health Initiative Insomnia Rating Scale (WHIIRS)

β-Amyloid burden, measured by carbon 11–labeled Pittsburgh compound B positron emission tomography distribution volume ratios (DVRs).

Among community-dwelling older adults, reports of shorter sleep duration and poorer sleep quality are associated with greater Aβ burden.

Craig, Northern Ireland, UK, 2006 ()

Cross-sectional N=426

Subjects were identified from outpatient memory-clinic records based at the Belfast City Hospital Trust, Mater Infirmorum, and Holywell Hospital. Patients fulfilled the criteria for probable AD.

A validated neuropsychological

assessment tool, the

Neuropsychiatric Inventory with

Caregiver Distress (NPI-D), was

employed to record sleep disturbances during the course of the dementing

illness.

Outpatient memory-clinic records. DNA extracted from peripheral blood leukocytes was analyzed for the APOE and MAO-A 30 bp VNTR polymorphisms

Sleep disturbance in AD is common and distressing and is associated with genetic variation at MAO-A.

Page 101: Omonigho A. Michael Bubu

89

Hita-Yañez, Spain, 2012 ()

Case-control N=50

Twenty-five MCI patients (7 females, mean age: 70.5 ± 6.8 yr) and 25 healthy old (HO) volunteers (13 females, mean age: 67.1 ± 5.3 yr)

Polysomnography (PSG)

ApoE genotype was determined by conventional PCR (polymerase chain reaction) methods. The diagnosis of MCI was based on consensus criteria. Global cognitive status was assessed with the Mini Mental State Examination (MMSE).

Sleep disruptions are evident years before diagnosis of AD, which may have implications for early detection of dementia and/or therapeutic management of sleep complaints in MCI patients.

Chen, Taiwan, 2011 () Case-control N=21

Authors recruited 9 controls without dementia, 6 patients with mild cognitive impairment (MCI), and 6 patients with mild Alzheimer's disease (AD). None of the participants had sleep complaints, and all AD patients were receiving cholinesterase inhibitors.

Polysomnography (PSG)

MMSE. NINCDS-ADRDA. All patients with AD were taking acetyl cholinesterase inhibitors (AChEI)

A deficiency of Ach may result in an increase of sEMG activity in MCI patients.

Hita-Yañez, Spain, 2013 ()

Case-control N=50

Twenty-five patients with MCI (7 females, mean age: 70.5 ± 6.8 y) and 25 HE subjects (13 females, mean age: 67.1 ± 5.3 y) were enrolled in the study,

Overnight PSG recordings and

self-reported sleep measures were

obtained

The diagnosis of MCI was based on consensus criteria. Global cognitive status was assessed with the Mini Mental State Examination (MMSE).

Sleep is significantly impaired in patients with mild cognitive impairment at both the objective and subjective level, which may be used as a surrogate marker of preclinical Alzheimer disease.

Page 102: Omonigho A. Michael Bubu

90

Hot, France, 2011 () Case-control N=28

Fourteen un-medicated AD patients (seven men and seven women; mean age (±SD): 76.7 ± 3.8 years) participated in this study

Polysomnography (PSG)

Episodic memory was assessed with an original task derived from Grober and Buschke’s procedure (1987).This task consisted in learning 15 words, presented by series of three on separate cards

Changes in theta rhythm during REM and SWS are a relevant index of brain impairments in the early stage of AD, and are related to episodic memory performance.

Sanchez-Espinosa, Spain, 2014 ()

Case-control N=21

Participants were primarily recruited from older people's associations, normal community health screening, and hospital outpatient services.

Polysomnography (PSG)

Neuropsychological testing; diagnostic criteria of aMCI proposed by Petersen et al. Plasma Aβ levels. Cerebral MRI acquisition, image preprocessing and cortical thickness estimation was done for each participant

Increased plasma Aβ42 levels are significantly associated with fragmented SWS in aMCI subjects, suggesting that sleep disruptions may signal Aβ burden in persons at increased risk for AD. Reduced REM sleep and plasma Aβ levels in aMCI subjects were significantly related to thinning of cortical regions targeted by AD neuropathology.

Keage, Australia, & United Kingdom 2012 ()

Prospective cohort N= 2012

The Medical Research

Council Cognitive Function

and Ageing Study (CFAS), a longitudinal multi-center population-based study of ageing and dementia (www.cfas.ac.uk). . Follow-up

time: 10-years

Self-reported measures .

MMSE

Daytime napping, night-time sleep duration, and excessive daytime sleepiness may be modifiable behaviors open to intervention strategies, or, clinical indicators of future decline in older individuals.

Page 103: Omonigho A. Michael Bubu

91

Tworoger, United States, 2006 ()

Prospective cohort N= 1844

Participants (all women) were members of the Nurses’

Health Study cohort aged 70 to 81 years at initial cognitive interview in 2000. Follow-up

period: 2years

Self-reported measures .

Women completed six tests of cognitive function encompassing general cognition, verbal memory, category fluency, and attention. Tests were repeated 2 years later.

Diminished cognition is a risk factor for dementia. However, the lack of an association with prospective cognitive decline warrants further investigation

Virta, Finland, 2013 () Prospective cohort N= 2,336

The subjects of the current study were members of the older Finnish Twin Cohort, consisting of same-sex twin pairs born in Finland prior to 1958 with both co-twins alive in 1967. Follow-up time: 18-

26years

Self-reported measures .

Between 1999 and 2007, participants were assigned a linear cognitive score with a maximum score of 51 based on a telephone interview (mean score 38.3, Standard Deviation (SD) 6.1).

This is the first study indicating that midlife sleep length, sleep quality, and use of hypnotics are associated with late life cognitive function. .

Song, United States, 2014 ()

Prospective cohort N = 2,601

The Osteoporotic Fractures in Men Study (MrOS). Participants were age 65 and older and free of probable dementia at sleep visit. Follow-up averaged 3.4 years

Sleep stages were identified by in-

home polysomnography at the initial sleep visit (2003-2005).

Cognitive outcomes were assessed with the Trail Making Test Part B and Modified Mini-Mental State Examination (3MS) at sleep visit and two follow-up

Increased time in Stage N1 and less time in Stage R are associated with worsening cognitive performance in older men over time

Blackwell, United States, 2014 ()

Prospective Cohort N=2822

The Osteoporotic Fractures

in Men Study (MrOS). Participants were age 65 and older (mean age 76.0 ± 5.3 y)and free of probable dementia at sleep visit. Follow-up averaged 3.4 ± 0.5

y.

Actigraphy

Clinically significant cognitive decline: five-point decline on the Modified Mini-Mental State examination (3MS), change score for the Trails B test time in the worse decile.

Among older community-dwelling men, reduced sleep efficiency, greater nighttime wakefulness, greater number of long wake episodes, and poor self-reported sleep quality were associated with subsequent cognitive decline.

Page 104: Omonigho A. Michael Bubu

92

Sterniczuk, Canada, 2013 ()

Retrospective Cohort

N= 28,697

The Survey of Health,

Ageing and Retirement in

Europe (SHARE) (N = 30,038). Follow-up period:

4.3years

Self-reported sleep measures

Alzheimer’s disease/Dementia was reported by the participants themselves and/or by a proxy respondent .Cognition was assessed using performance-based cognitive tests

These findings indicate that sleep disturbance may exist prior to the manifestation of other typical symptoms observed in AD (e.g., memory loss).

Benito-León, Spain, 2014 ()

Prospective cohort N= 3,857

Neurological Disorders in

Central Spain (NEDICES) Study, a longitudinal, population-based survey followed for a median of 12.5

years

Self-reported sleep measures

ICD-9 codes for deaths that occurred before 1999 and ICD-10 codes for deaths occurring thereafter.

Self-reported long sleep duration was associated with 58% increased risk of dementia-specific mortality in this cohort of elders without dementia.

Hahn, United States, 2013 ()

Prospective cohort N= 214

The population-based

Kungsholmen Project, a longitudinal study of aging and dementia among adults aged 75 years and older residing in Kungsholmen district, Stockholm, Sweden. Follow-

up time: Up to 9-years

Comprehensive Psychopathological

Rating Scale (CPRS)

DSM-III-Revised and AD diagnosis was made using the NINCDS-ADRDA criteria

Self-reported sleep problems may increase the risk for dementia, and depressive symptoms may explain this relationship.

Bidzan, Poland, 2011 () Prospective cohort N=150

Care centers residents age 55 and older, who had had the presence of dementia excluded (n = 2910).. Follow-up time:

7-years

The Neuropsychiatric

Inventory-Nursing Home (NPI-NH)

scale and the Association for

Methodology and Documentation in

Psychiatry (AMDP) scale.

Alzheimer’s disease Assessment Scale Cognitive (ADAS-cog) scale. Dementia in the Alzheimer’s disease was diagnosed based on the NINCDS-ADRDA criteria.

The patients in the preclinical period of dementia experienced sleep disturbances more frequently and with greater intensity, which in combination with other factors may have some prognostic value.

Page 105: Omonigho A. Michael Bubu

93

Lim, Canada, 2013 () Prospective cohort N= 698

Participants were community-dwelling older adults without dementia (mean age, 81.7 years; 77% women) in the Rush Memory and Aging

Project. follow-up period of

up to 6 years.

Actigraphy

Neuropsychometric tests. NINCDS-ADRDA criteria. Autopsies were performed on 201 participants who died, and β-amyloid (Aβ) and neurofibrillary tangles were identified by immunohistochemistry and quantified. Peripheral blood lymphocytes were used for DNA extraction, and APOE genotype was determined

Better sleep consolidation attenuates the effect of APOE genotype on incident AD and development of neurofibrillary tangle pathology.

Rothman, United States, 2013 ()

Experimental Not Applicable

Male 3xTgAD mice were subjected to sleep restriction(SR) for 6h/day for 6 weeks using the modified multiple platform technique, and behavioral(Morris water maze, fear conditioning, open field)

Mice were tested in the Morris water

maze (MWM)

Hippocampal and cortical Aβ and P-Tau concentrations were obtained

Significant positive correlations between cortical Aβ and p -Tau levels and circulating corticosterone indicate a potential role for GCs in mediating behavioral and biochemical changes observed after sleep restriction in a mouse model of AD

Di Meco, United States, 2014 ()

Experimental Not Applicable

A total of 18 mice were kept in a pathogen-free environment, on a 12-hour light and/or dark cycle and had access to food and water ad libitum. Starting at the age of 8 months, mice were randomized to 2 groups, one underwent a 20/ 4-hour light and/or dark cycle for 2 months (4 males and 5 females), the other was kept on a normal light and/or dark cycle, as controls (5 males and 4 females).

The locomotor cage test was used to record the sleep-wake cycle of the animals. The fear conditioning tests. The Morris water-

maze test

Mouse brain homogenates were extracted in radio-immuno-precipitation assay (RIPA) buffer and were prepared for immunohistochemistry. Cd-5 kinase (Cdk-5) activity was carried out.

Authors conclude that alteration of the sleep-wake cycle and circadian rhythm regulation could be important players in the onset and development of sporadic AD. Correction of sleep-wake cycle aberrations could be a viable therapeutic strategy for individuals bearing this risk factor.

Page 106: Omonigho A. Michael Bubu

94

Kang, United States, 2009 ()

Experimental Not Applicable Both wild-type mice and human APP transgenic (Tg2576) mice

Mice were forced into wakefulness for 6 hours at the beginning of the second 12-hour

light period when they would

naturally be asleep.

Authors monitored hippocampal Aβ levels using in vivo micro dialysis in both wild-type mice and human APP transgenic (Tg2576) mice, which express a mutated form of human amyloid precursor protein (APP). ISF Aβ was assessed in Tg2576 mice at 3 months of age, several months earlier than Aβ deposition begins.

Sleep-wake cycle and orexin may play a role in the pathogenesis of Alzheimer’s disease.

Mander, United States, 2013

Experimental N=33

A group of cognitively normal older adults (n=18 mean age 20.4 ± 2.1) and a group of healthy young adults (n=15 mean age 72.1 ± 6.6)

Measured with Polysomnogram (PSG), starting at their habitual bed time to minimize the influence of

age-related circadian

differences.

Approximately 2 h post-awakening, participants performed an event-related fMRI scanning session while performing a long-delay (10 h) recognition test

Together, these data support a model in which age-related mPFC atrophy diminishes SWA, the functional consequence of which is impaired long-term memory.

Ooms, Netherlands, 2014 ()

Randomized Controlled Trial

N=26

The Alzheimer,

Wakefulness, and Amyloid

Kinetics (AWAKE) study at the Radboud Alzheimer Center, a randomized clinical trial that took place between

June 1, 2012, and October 1,

2012.

Sleep was monitored using

continuous polysomnographic recording from 3 PM until 10 AM.

Cerebrospinal fluid, Aβ42, P-tau, and T-tau were deter-mined using the xMAP-based Innobia assay (Innogenetics) and CSF Aβ40 was measured using an enzyme-linked immuno-sorbent assay

Sleep deprivation, or prolonged wakefulness, interferes with a physiological morning decrease in Aβ42. Chronic sleep deprivation increases cerebral Aβ42 levels, which elevates the risk of Alzheimer disease.

Page 107: Omonigho A. Michael Bubu

95

Table 1.8. Sleep-wake cycle abnormalities and Cognitive/Alzheimer's Disease Measures: Descriptive Study Characteristics, and Main

Findings

First Author,

Country, Year

Published

(Reference)

Study Design Sample

size N Study Population

Sleep-wake cycle

Assessment

Cognitive/AD Measures

Assessment Conclusion

Oosterman, Netherlands, 2009 ()

Cross-sectional (Correlational)

N= 144

Study was part of a larger study on the effects of cardiovascular risk factors, and participants (90 males and 54 females, aged 69.5 ± 8.5 years (mean ± SD), range 50–91) were recruited among people visiting the outpatient clinic (of cardiology, internal medicine, or neurology) of the Saint Lucas Andreas Hospital in Amsterdam, the Netherlands.

Wrist Actigraphy

Mental speed: Stroop test (Stroop, 1935). The Word (W) and Color (C) cards of the Stroop test Memory: 15-words test. The Dutch version (Saan and Deelman, 1986) of the Auditory Verbal Learning Test (Rey, 1964) .Digit span forward (Wechsler, 1987).

An association between the rest-activity rhythm and cognitive performance is present in elderly people.

Merlino, Italy, 2010 ()

Cross-sectional N=750

Subjects aged 65 years or older, living independently or in an institution, who resided in the seventh district of Udine, Italy were recruited.

Sleep disorders were investigated with a battery of standardized questions and questionnaires

The diagnosis of dementia was made according to the DSM-IV-TR criteria . AD and VaD were diagnosed respectively according to NINCS-ARDRA and NINDS-AIREN criteria . LBD and PDD, frontotemporal dementia (FTD) and MCI were diagnosed according to specific consensus guidelines .

Excessive daytime sleepiness was significantly related to dementia. Insomnia, was not associated with the presence of cognitive decline.

Pat-Horenczyk, United States, 1998 ()

Cross-sectional N= 67

Nursing home residents (mean age = 85.7 years). There were 46 severely demented patients, and 21 mild-moderately demented patients

Actigraphy Levels of dementia were assessed by Mini-Mental State Examination (MMSE)

With the progression of dementia, both the capacity to maintain sleep and the capacity to maintain wakefulness are impaired, and result in complete fragmentation of sleep/wakefulness during the night and day.

Yesavage, United States & France, 2011 ()

Cross-sectional N=300

Two cohorts of AD participants. Cohort 1 (n=124): individuals with probable AD recruited from the Stanford/Veterans Affairs NIA Alzheimer’s Disease Core Center (n=81) and the Memory Disorders Clinic at the University of Nice School of Medicine (n=43). Cohort 2 (n=176): individuals with probable AD derived from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) data set.

Wrist actigraphy data for seven days in Cohort 1 and by the Neuropsychiatric Inventory (NPI-Q) for Cohort 2.

Diagnosis of probable AD by NINCDS-ADRDA criteria based on relevant neurological, medical, neuroimaging, and neuropsychological assessments.

It is unlikely that a relationship with a clinically meaningful correlation exists between the circadian rhythm-associated SNPs and WASO in individuals with AD.

Page 108: Omonigho A. Michael Bubu

96

Naismith, Australia, 2014 ()

Case-control N=58

Participants (30 patients with MCI and 28 age-matched controls) were recruited from a specialist ‘Healthy Brain Ageing’ Clinic, at the Brain & Mind Research Institute, Sydney, Australia.

Actigraphy. Polysomnographic (PSG) assessments, on separate nights. Salivary melatonin was assessed to determine dim light melatonin onset (DLMO)

A clinical diagnosis of MCI was obtained using Petersen’s criteria

Circadian misalignment and sleep disruption is evident in patients with MCI, and is consistent with changes observed in Alzheimer’s disease. Such findings could be a marker for disease trajectory, and may even be implicated in disease pathogenesis.

Fronczek, Netherland, 2012 ()

Case-control N=49

Ten patients with AD (Braak 5 or 6) and 10 non-demented controls (Braak 0 or 1) were matched for sex, age, postmortem delay (PMD), and fixation time (hypothalamus group).

Hypocretin-1 immunocytochemistry and neuron quantification and measurement in ventricular CSF was done.

Clinical diagnosis and Neuropathological examination according to van de Nes et al. 1993 and Braak et al. 1998

Hypocretin system is affected in advanced AD. This is reflected in a 40% decreased cell number, and 14% lower CSF hypocretin-1 levels.

Slats, Netherlands, 2012 ()

Case-control N=12 Six patients with AD (59-85 yrs, MMSE 16-26) and six healthy volunteers (64-77 yrs).

CSF hypocretin-1 was measured using a commercially available RIA kit (Phoenix Pharmaceuticals, Belmont, CA).

CSF Aβ42 was determined using the xMAP-based Innobia assay (Innogenetics NV, Ghent, Belgium) CSF Aβ40 using ELISA (the Genetics Company, Schlieren, Switzerland).Total protein level in CSF was analyzed using the Lowry method.

Mean Aβ42 levels and mean hypocretin-1 levels and amplitude may suggest a relationship between AD pathology and hypocretin disturbance, which could hold possibilities for treatment of AD related sleep disorders.

Schmidt, Germany, 2013 ()

Case-control N=66

33 patients with mild to severe AD and 33 subjects without any psychiatric or neurological disorder (HS) that were consecutively recruited to participate in the study.

Melanin-concentrating hormone (MCH) and hypocretin-1 (HCRT-1, orexin-A) were measured using a fluorescence immunoassay (FIA)

Consortium to Establish a Registry for Alzheimer’s Disease (CERAD), Wechsler Memory Scale-Revision (WMS-R), clock drawing test, and Trail Making Test (TMT), a MRI head-scan, genotyping of ApoE4 and determination of Ab42, T-tau and P-tau in the CSF.

This report on MCH in patients with AD adds to preclinical studies and may allow further research on the role of the hypothalamus in pathology and occurrence of behavioral disturbances in AD.

Page 109: Omonigho A. Michael Bubu

97

Liu, Netherlands and China, 1999 ()

Case-control N=167

Autopsies were performed within the framework of The Netherlands Brain Bank. Ventricular postmortem CSF was obtained at autopsy, 1–12 h after death, from 85 patients with AD (mean age, 75 ± 1.1 yr) and 82 age-matched controls (mean age, 76 ± 1.4 yr) without a primary neurological or psychiatric disease.

Melatonin in postmortem CSF was measured by a direct RIA

ApoE genotyping. The genotype of each extracted DNA sample was determined by polymerase chain reaction (PCR)

CSF melatonin levels fromApoE-€3/4 genotype patients were significantly higher thanthose from the ApoE-€4/4 genotype, suggesting a relationship between melatonin levels and signs and symptoms of AD. CSF melatonin levels was lower in AD patients than in old control subjects.

Walsh, United States 2014 ()

Prospective Cohort

N=1287

1,287 community-dwelling older women (82.8 ± 3.1 y) participating in an ongoing prospective study who were free of dementia at the baseline visit. Follow-up

time: 5 years

Actigraphy

Modified Mini-Mental Status Examination (3MS), California Verbal Learning Task (CVLT), digit span, Trail Making Test B (Trails B), categorical fluency, and letter fluency.

Weaker Circadian Activity Rhythm patterns are associated with worse cognitive function, especially executive function, in older women without dementia.

Keage, Australia, & United Kingdom 2012 ()

Prospective cohort

N= 2012

The Medical Research Council

Cognitive Function and Ageing Study (CFAS), a longitudinal multi-center population-based study of ageing and dementia (www.cfas.ac.uk). Follow-up

time: 10-years

Self-reported sleep measures

MMSE

Excessive daytime sleepiness may be modifiable behaviors open to intervention strategies, or, clinical indicators of future decline in older individuals.

Anderson, United Kingdom, 2014 ()

Prospective Cohort

N= 421

The study was nested in the Newcastle

85+ Study, a population-based longitudinal study of health and ageing in the very old. Follow-up time: 2 years

Pittsburgh Sleep Questionnaire Inventory (PSQI) and the Epworth Sleepiness Score (ESS) as subjective measures of sleep and wake.

Cognition assessed by the Mini-Mental State Examination (MMSE)

Abnormal sleep–wake patterns are associated with cognitive impairment

Page 110: Omonigho A. Michael Bubu

98

Tranah, United States, 2011 ()

Prospective Cohort

N=1282

Women were participants of the Study of

Osteoporotic Fractures (SOF), a longitudinal epidemiologic study of 10,366 community-dwelling women age 65 years or older. Follow-up time: 4.9

years

Circadian activity rhythms were measured with the Sleep-Watch-O (Sleep-Watch-OVR, Ambulatory Monitoring, Inc., Ardsley, NY) actigraph.

Modified Mini-Mental State Examination (3MS); California Verbal Learning Test (CVLT) delayed recall; Informant Questionnaire on Cognitive Decline in the Elderly (IQCODE); self-reported previous dementia diagnosis

Older, healthy women with decreased circadian activity rhythm amplitude and robustness, and delayed rhythms have increased odds of developing dementia and MCI

Lim, United States, 2013 ()

Prospective Cohort

N=737

Rush Memory and Aging Project (MAP)which is an ongoing community-based cohort study of aging which began in 1997. Follow-up period was up to 6

years (mean 3.3 years).

Actigraphy Battery of Neuropsychological Tests

Sleep fragmentation in older adults is associated with incident AD and the rate of cognitive decline.

Yesavage, United States, 2004 ()

Prospective Cohort

N=44

To be included in the current analysis, the patient must have been assessed during at least 2 stages of cognitive impairment, defined as a decline from one stage of AD to another.

Actigraphy

APOE genotypes were then determined. Diagnosis of probable AD by NINCDS-ADRDA criteria and a score of 15 or higher at entry on the Mini-Mental State Exam (MMSE). .

APOE status is associated with the progression of sleep/wake disturbances in AD

Craig, Canada, 2008 ()

Experimental Not

Applicable

Twenty-four male Long Evans hooded rats (300–400 g) obtained from Charles River (Saint-Constant, PQ) were used for all studies. They were divided into three groups: acutely phase shifted rats (acute; n = 8), chronically phase shifted rats (chronic; n = 8), control rats for water maze testing (control; n=7) and control rats for fear conditioning (control; n = 14).

In order to create a state of circadian disruption a two-stage phase shifting session which lasted 16 days was used.

Rats underwent Behavioral Testing on a 6 day version of the water maze task, followed by 3 days of testing on a fear conditioning task. They then underwent a Rapid Acquisition Task training in three stages.

Authors propose that chronic circadian disruption may play a role in the development of age-related cognitive deficits and dementia in the elderly

Page 111: Omonigho A. Michael Bubu

99

Jyoti, Scotland, UK, 2010 ()

Experimental Not

Applicable

APP/PSEN1 and PSEN1 Transgenic mice. Transgenic mice were singly housed and assigned to the following groups, depending upon age (5 or 20 months) and genotype: 1) APP/PSEN1 [5 months: n = 8; 20 months: n = 6]; 2) PSEN1 [5 months: n = 8; 20 months: n = 11]; 3) WT [5 months: n = 9; 20 months: n = 8].

Circadian activity was recorded in PhenoTyper home cages (Noldus, The Netherlands) through video observation techniques and XY coordinates recorded over 7 consecutive days.

Authors characterized sleep, electroencephalogram (EEG) disturbances i.e., endophenotypes of Alzheimer’s disease (AD) patients alongside cognitive dysfunction in transgenic mice carrying transgenes for amyloid-protein precursor (APPswe) and presenilin 1 (PSEN1A246E) at 5 (pre-plaque) and 20 months, relative to PSEN1 and wild-type (WT) mice, using a novel wireless microchip device.

APP/PSEN1 mice exhibit abnormalities in activity and sleep architecture preceding amyloid plaque deposition as well as age-related changes in cortical EEG power.

Quinn, United States, 2005 ()

Experimental Not

Applicable Transgenic mice that were bred from a breeding pair of Tg2576 mice

Plasma melatonin

Soluble and formic acid extractable beta amyloid determination was done and Aβ burden in hippocampus and cerebral cortex was quantified with NIH image.

Melatonin failed to produce anti-amyloid or antioxidant effects when initiated after the age of amyloid plaque deposition. These findings diminish the possibility that melatonin will be useful for the treatment of established Alzheimer’s disease.

DENG. China, 2005 ()

Experimental Not

Applicable

N2a cells were grown and differentiated in 96-well culture plates at density of 1.5×105 cells in 100 μL. Cells were exposed to various concentrations of wortmannin for 2 h at 37 ºC in the presence or absence of a 12-h-preincubation with melatonin 25, 50, and 100 μmol/L or Vitamin E (VE). Dimethylsulfoxide (Me2SO, 0.01%), in which wortmannin, melatonin and VE were dissolved, served as a vehicle control.

Western blot, P-labeling and the detection of malondialdehyde level and superoxide dismutase activity

MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl-tetrazolium bromide), crystal violet assay, phase-contrast, dead end colorimetric apoptosis detection system (TUNEL) and electron microscopy were used to detect cell viability, morphology and apoptosis

Wortmannin is an effective tool for reproducing Alzheimer-like tau hyperphosphorylation cell model and melatonin/vitamin E can effectively protect the cells from wortmannin-induced impairments

Page 112: Omonigho A. Michael Bubu

100

Papolla, United States, 1998 ()

Experimental Not

Applicable

Aliquots of Aβ1–40 and Aβ1–42 at a concentration of 250 μm in 5 mm Tris-HC1, pH 7.4, were incubated at room temperature

Melatonin or the melatonin analog 5-hydroxy-N-acetyl-tryptamine (NAT) (Sigma) orN-t-butyl-α-phenylnitrone (PBN) (Sigma),

Peptides Aβ1–40 and Aβ1–42 were synthesized in the W. M. Keck Foundation (Yale University, CT), and their purity was evaluated by amino acid sequence and laser desorption mass spectrometry.

Melatonin can provide a combination of antioxidant and anti-amyloidogenic features that can be explored either as a preventive or therapeutic treatment for AD or as a model for development of anti-amyloidogenic indole analogs.

Olcese, United States, 2009 ()

Experimental Not

Applicable APP + PS1 double transgenic (Tg) mice

Authors administered melatonin (100 mg/L drinking water) to APP + PS1 double transgenic (Tg) mice from 2–2.5 months of age to their killing at age 7.5 months.

Mice were initially genotyped at the time of weaning and then had confirmatory genotyping within several months thereafter, with further confirmation via plasma Ab40 expression.

Results are consistent with a melatonin-facilitated removal of Aβ from the brain. Inflammatory cytokines such as tumor necrosis factor (TNF)-α were decreased in hippocampus (but not plasma) of Tg+ melatonin mice.

Lahiri, United States, 2004 ()

Experimental Not

Applicable

Male B6C3F1 mice, a hybrid between C57BL/6 and C3H from Harlan Labs (Indianapolis, IN, USA), aged 6 months (young group), 12 months (middle-aged group) and 27 months (old group), were housed two or three per cage and were maintained on a 12 hr light/dark cycle in a temperature controlled (22 ± 1°C) room.

An ELISA procedure was used for the quantitative measurement of melatonin in serum

A sensitive enzyme-linked immunosorbent assay (ELISA) test for measuring levels of Aβ-40 was performed in the murine brain homogenates. ELISA test was also performed using the IBL assay reagents for the quantitative determination of Aβ-42 in the murine brain homogenates.

Melatonin supplementation may retard neurodegenerative changes associated with brain aging. Depletion of melatonin in the brain of aging mice may in part account for this adverse change.

Asayama, Japan, 2003 ()

Randomized Controlled Trial

N=20

The subjects were 9 persons given a placebo (PLA), and 11 given melatonin (3 mg)(MLT). The drugs were given at 20: 30 each day for 4 weeks.

Actigraph data from 18 patients (PLA8, MLT10)

Diagnosis of AD via CT or MRI and a clinical diagnosis based on DSM IV criteria and the NINCDS-ADRDA criteria, Dementia severity was assessed by the Clinical Dementia Rating Scale (CDR), Cognitive funcion was assessed by the Alzheimer's Disease Assessment Scale (ADAS) the MMSE

Melatonin administration improved sleep time and night activity significantly, but no significant effect with regard to improving daytime naps and activity. Melatonin administration improved cognitive and non-cognitive functions significantly.

Page 113: Omonigho A. Michael Bubu

101

Table 1.9. Sleep Disordered Breathing/Sleep Apnea and Cognitive/Alzheimer's Disease Measures: Descriptive Study

Characteristics, Statistical Methods and Main Findings

First Author, Country, Year

Published (Reference) Study Design

Sample

size N Study Population Hypoxia Assessment

Cognitive/AD Measures

Assessment Conclusion

Gottlieb, United States, 2004 ()

Cross sectional N=1,775 .

Participants were aged 40 to 100 years from the Sleep

Heart Health Study (SHHS) a longitudinal cohort study of the relation of sleep-disordered breathing to CVD.

Obstructive Sleep Apnea/Hypopnea (OSAH) was defined as an Apnea Hypopnea Index (AHI) ≥ 15.

APOE was genotyped

The APOE £4 allele is associated with increased risk of OSAH, particularly in individuals under age 65.

Kadotani, United States, 2001 ()

Cross-sectional N=791 Probability based sampling of 791 middle-aged adults, mean age of 49 and age range 32-68

Nocturnal Polysomnography with Sleep Disordered Breathing defined as AHI ≥ 15

APOE was genotyped

A significant proportion of Sleep Disordered Breathing is associated with APOE £4 allele in the general population

Osorio, United States, 2014 () Cross-sectional N=95

Ninety-five cognitively normal elderly participants were recruited at NYU Center for Brain Health ongoing between 2009 and 2013.

Among the 95 participants, 25 were considered free of SDB (AHI4% < 5) and were included as normal controls, 51 had mild SDB (AHI4% 5-14.99), and 19 had moderate to severe SDB (AHI4% > 15).

CSF measures of phosphorylated-tau (p-tau), total-tau (t-tau), and amyloid beta 42 (Ab-42), as well as ApoE allele status

There is an association between SDB and CSF Alzheimer’s disease-biomarkers in cognitively normal elderly individuals..

Djonlagic, United States, 2014 ()

Cross-sectional N=44

Participants (19–68 years) who had been referred by a physician for a baseline polysomnography (PSG) evaluation.

Based on their PSG, patients were assigned either to the OSA group (AHI>5/h), or control (Non-OSA) group (AHI<5/h).

Psychomotor Vigilance Task (PVT) and the Motor Sequence Learning Task (MST)

The presence of untreated obstructive sleep apnea is associated with an aging-related cognitive deficit, otherwise not present in individuals without OSA.

Page 114: Omonigho A. Michael Bubu

102

O’Hara, United States, 2005 ()

Case-control N=36

Twelve men and 24 women participated. All were over age 60, with a mean age of 70.6 (SD = 8.1), a mean of 16.1 (SD = 2.4) years of education, and a mean Mini-Mental State Examination (MMSE) of 28.7 (SD = 1.2; range 27 to 30). Eighteen had an APOE €4 allele, and 18 were non-carriers

The apnea-hypopnea index (AHI), i.e., the average number of apneas or hypopneas per hour of estimated sleep measured OSAH severity in each subject.

APOE genotyping. MMSE; the Rey Auditory Verbal Learning Test of immediate verbal recall (RAVLT1); short-term free recall (RAVLT6) and 30-minute delayed free-recall (RAVLT Delayed); the Stroop Color and Word (SCW) measure of attention and cognitive flexibility; and the Symbol-Digit Modalities Test (SDMT) of information processing speed.

Nocturnal sleep apnea/hypopnea is associated with lower memory performance in APOE €4 carriers

Cohen-Zion, United States, 2001 ()

Prospective cohort

N=46

Community-dwelling people age 65 and older with high risk for SDB were originally studied from 1981 through 1985 and then followed every 2 years. Follow-up period:

2years

Subjects’ sleep was recorded using the Modified Respitrace/Medilog system, which measured thoracic and abdominal respiration, tibialis electromyogram (EMG), and wrist activity (to discriminate sleep/wake states).

MMSE

The results of this study could theoretically suggest that any relationship between SDB and cognitive function may be mediated by the effect of SDB on daytime sleepiness.

Tworoger, United States, 2006 ()

Prospective cohort

N = 1844

Participants were women aged 70 to 81 years at initial cognitive interview in 2000 and members of the Nurses’

Health Study cohort. Follow-

up period: 2years

Authors collected information about average sleep duration and snoring frequency on questionnaires in 1986 and 2000;

Women completed six tests of cognitive function encompassing general cognition, verbal memory, category fluency, and attention. Tests were repeated 2 years later.

Associations between sleep patterns and initial cognitive function may be clinically relevant given that diminished cognition is a risk factor for dementia. However, the lack of an association with prospective cognitive decline was seen

Page 115: Omonigho A. Michael Bubu

103

Chang, Taiwan, 2013 () Retrospective cohort

N=8484

Longitudinal Health

Insurance Database 2005 (LHID2005) in Taiwan, 1414 patients with Sleep Apnea (SA) aged 40 years Follow-

up period: 5years

Diagnoses of SA [ICD-9-Codes 327.23, 780.51, 780.53, 780.57] between January 2003 and December 2005. Clinically assessed including only cases in which patients obtained ≥2 SA diagnoses in outpatient visits or ≥1 inpatient service.

ICD-9 code

SA may be a gender-dependent, age-dependent, and time-dependent risk factor for dementia

Yaffe, United States, 2011 () Prospective cohort

N=298 Study of Osteoporotic

Fractures. Follow-up

period: 2.3 years

Sleep disordered breathing was defined as an apnea-hypopnea index of 15 or more events per hour of sleep.

Diagnostic and Statistical Manual of Mental Disorders (Fourth Edition) criteria. MCI was diagnosed using the modified criteria by Peterson et al

Among older women, those with sleep disordered breathing, compared with those without SDB, were associated with an increased risk of developing cognitive impairment

Shiota, Japan, 2013 () Experimental Not Applicable

In vivo, 15 male triple transgenic AD mice were exposed to either Chronic Intermittent Hypoxia (CIH) or normoxia (5% O2 and 21% O2 every 10 min, 8 h/day for 4 weeks). In vitro, human neuroblastoma SH-SY5Y cells stably expressing wild-type amyloid-β protein precursor were exposed to either IH (8 cycles of 1% O2 for 10 min followed by 21% O2 for 20 min) or normoxia

CIH was applied (n = 9) by exposing to alternating 5% O2 and 21% O2 every 10 min for 8 h per day during daytime for 8 weeks in a chamber (370×260×250 mm, 26 L, Sibata Scientific Technology Ltd, Tokyo, Japan). Control mice (n = 6) were kept under normoxia and touched by human hands once a day to balance out their stress through direct human contact.

Amyloid-β (Aβ) profile, cognitive brain function, and brain pathology were evaluated.

Results suggest that Obstructive Sleep Apnea (OSA) would aggravate AD. Early detection and intervention of OSA in AD may help to alleviate the progression of the disease.

Kaushal, United States, 2012 ()

Experimental Not Applicable

Adult male human ApoE4-targeted replacement mice [B6.129P2-Apoetm3(APOE*4)MaeN8] (hApoE4) and C57BL/6NTac (wild-type, WT) mice,

HYPOXIA MEASURE O2 concentration was continuously measured by an O2 analyzer and was changed from 1:00 PM until 7:00 PM by a computerized system controlling the gas valve outlets.

SLEEP INTEGRITY

MEASURE Polysomnography

The increased susceptibility and limited recovery ability of hApoE4 mice to sleep apnea suggests that early recognition and treatment of the latter in AD patients may restrict the progression and clinical manifestations of this frequent neurodegenerative disorder

Page 116: Omonigho A. Michael Bubu

104

Sun, Canada, China, United States, 2006 ()

Experimental Not Applicable

APP23 Transgenic mice

APP23 mice (8 months of age, n = 20, 50% female) were assigned randomly to hypoxia and control groups. The hypoxia group was treated in a hypoxia chamber at 8% O2 for 16 h/day for 1 month.

The water maze test. Neuritic Plaque Staining was done.

The results demonstrate that hypoxia can facilitate AD pathogenesis, and they provide a molecular mechanism to link vascular factors to AD

Li, China, 2009 () Experimental Not Applicable

Transgenic mice (The Jackson Lab, No. 003378, APPSwe + PS1A246E)

The mice were treated with hypoxia once a day and repeated for 60 days. Mice were assigned randomly to hypoxia and control groups (9 months old female, 10 mice in each group).

Neuritic plaques staining was performed.

Taken together, the results suggest an important role of hypoxia in modulating the APP processing by facilitating both β- and ɣ-cleavage which may result in a significant increase of Aβ generation.

Zhang, China, 2007 () Experimental Not Applicable

Mouse neuroblastoma N2a cells stably expressing human APP695 (N2a-APP) were maintained in cell culture medium containing 50% DME high glucose medium

Cell culture, Transfection and Hypoxic treatment was done.

Immunoblotting and Antibodies, Aβ ELISA Assay, In Vitro β-Secretase Activity Assay, Pulse-chase experiments, Quantitative Real-time Polymerase Chain Reaction (RT-PCR), HIF-1α RNA Interference, Tissue Isolation from HIF-1α Conditional Knock-out Mice were also carried out on the mice models

The results demonstrate an important role for hypoxia/HIF-1α in modulating the amyloidogenic processing of APP and provide a molecular mechanism for increased incidence of AD following cerebral ischemic and stroke injuries

Kushida, United States, 2012 ()

Randomized Controlled Trial

N=1,098

The Apnea Positive Pressure

Long-term Efficacy Study (APPLES) was a 6-month, randomized, double-blind, 2-arm, sham-controlled, multicenter trial conducted at 5 U.S. University, hospital, or private practices.

Diagnosis of Obstructive Sleep Apnea (OSA) with an apnea-hypopnea index (AHI) ≥ 10 and age ≥ 18 years

Three neurocognitive variables, each representing a neurocognitive domain: Pathfinder Number Test-Total Time (attention and psychomotor function [A/P]), Buschke Selective Reminding Test-Sum Recall (learning and memory [L/M]), and Sustained Working Memory Test-Overall Mid-Day Score (executive and frontal-lobe function [E/F])

CPAP treatment improved both subjectively and objectively measured sleepiness, especially in individuals with severe OSA (AHI >30).

Page 117: Omonigho A. Michael Bubu

105

Ancoli-Israel, United States, 2008 ()

Randomized Controlled Trial

N=52

Participants were men and women with mild-moderate AD and OSA recruited from the University of California San Diego (UCSD) Alzheimer's Disease Research Center.

Diagnosis of Obstructive Sleep Apnea (OSA) with an apnea-hypopnea index (AHI) ≥ 10.CPAP unit pressure was set at 8 cm H2O pressure while the system mask's pressure varied from 0.5 cm H2O at end-expiration to 0.0 cm H2O during inspiration

NINCS-ARDRA and a CT or MRI of the brain consistent with AD done within 24 months Mini Mental Status Examination (MMSE) score greater than 17.

OSA may aggravate cognitive dysfunction in dementia and thus may be a reversible cause of cognitive loss in AD patients.

Table 1.10. Insomnia and Cognitive/Alzheimer's Disease Measures: Descriptive Study Characteristics, and Main Findings

First Author,

Country, Year

Published

(Reference)

Study Design Sample size

N Study Population Insomnia Assessment Cognitive/AD Measures Assessment Conclusion

Merlino, Italy, 2010 ()

Cross-sectional N=750

Subjects aged 65 years or older, living independently or in an institution, who resided in the seventh district of Udine, Italy were recruited.

Insomnia and other sleep abnormalities were investigated with a battery of standardized questions and questionnaires

DSM-IV-TR criteria . AD and VaD were diagnosed respectively according to NINCS-ARDRA and NINDS-AIREN criteria . LBD and PDD, frontotemporal dementia (FTD) and MCI were diagnosed according to specific consensus guidelines .

Insomnia was not associated with the presence of cognitive decline. As opposed to insomnia, excessive daytime sleepiness was significantly related to dementia.

Cricco, Iceland, 2001 ()

Prospective cohort N=6444

The four sites of the Established Populations

for Epidemiologic Studies of the Elderly (EPESE). Follow-up period: 3 years

Insomnia was defined as self-report of trouble falling asleep or waking up too early most of the time.

Cognitive decline was defined as two or more errors on the Short Portable Mental Status Questionnaire (SPMSQ) at FU3.

Chronic insomnia independently predicts incident cognitive decline in older men.

Jelicic, The Netherlands, 2002 ()

Prospective Cohort

N=838

Maastricht Ageing Study

(MAAS), a prospective cohort study of cognitive ageing based in the Netherlands. Follow-up

period: 3 years

Self-reported sleep measures

Cognitive performance at follow-up, measured with the Mini Mental Status Examination (MMSE)

Subjective sleep complaints predict cognitive decline in middle aged and older adults

Page 118: Omonigho A. Michael Bubu

106

Foley, United States, 2001 ()

Prospective Cohort

N=2346

Japanese-American men age 71 to 93 of The

Honolulu-Asia Aging

Study (HAAS)

Sleep disturbances were assessed by questionnaire.

Dementia was diagnosed according to less-restrictive diagnostic criteria developed by Cummings et al. for very mild dementia and more-restrictive diagnostic criteria for mild or more severe dementia from the Diagnostic and Statistical Manual of Mental Disorders, Third Edition, Re-vised (DSM-IIIR).

Daytime sleepiness in older adults may be an early indicator of decline in cognitive functioning and onset of dementia.

Osorio, United States, 2011 ()

Prospective Cohort

N=346

Brain aging studies at New York University’s Alzheimer’s Disease Research Center. Follow-

up period: over 7.7 years.

Insomnia was collected only during the normal cognition stage and screened using items 4, 5, and 6 from the Hamilton Depression Rating Scale (HAM-D)

Mini-Mental State examination There is a greater risk of Alzheimer's disease in adults with Insomnia

Table 1.11 Restless Less Syndrome (RLS) and Cognitive/Alzheimer's Disease Measures: Descriptive Study Characteristics, and Main

Findings

First Author,

Country, Year

Published

(Reference)

Study Design Sample size

N Study Population RLS Assessment Cognitive/AD Measures Assessment Conclusion

Pearson, United States, 2006 ()

Case-control N=31

Sixteen patients off RLS treatment for at least 2 weeks and 15 age- and gender-matched control subjects

RLS patients were required to have daily symptoms and to have periodic leg movements in sleep (PLMS) of greater than 20/h on a screening nocturnal Polysomnogram (PSG).

Cognitive Tests consisted of the following: the Stroop color-word test, Trail Making Test (A and B), the Porteus Maze Test (Vineland Revision), the Colored Progressive Matrices Test, and two verbal fluency tests (words beginning with a fixed first letter and words in categories).

RLS patients have statistically significant specific cognitive deficits and show cognitive deficits similar to those seen with 36 h total sleep deprivation.

Page 119: Omonigho A. Michael Bubu

107

Figure 1.1 Flow Diagram of Systematic Review of Sleep and Alzheimer’s disease

Studies identified through database searching (n = 2341)

(PubMed=904, Embase=1026, Web of Science=305, Cochrane library=106)

Studies after duplicates removed (n = 1221)

Studies screened for titles (n = 1221)

Studies excluded (n = 180)

Full-text articles assessed for eligibility

(n = 201)

Full-text articles excluded, with reasons (n = 130)

- Review article (n = 7) - No risk estimate (n =

17) - Sleep not assessed (n =

18) - Relationship not

studied (n = 53) - Not peer reviewed (n =

10) - Alzheimer not

examined (n = 25)

Studies included in systematic review

(n = 72)

Article identified from reference search

(n = 1)

Studies screened for abstracts

(n = 1041)

Studies excluded (n = 840)

Duplicates removed (n = 1120)

Page 120: Omonigho A. Michael Bubu

108

SECTION 2

SLEEP, COGNITIVE IMPAIRMENT AND ALZHEIMER’S DISEASE: A SYSTEMATIC REVIEW

AND META-ANALYSIS

Note to the Reader:

This entire chapter has been previously published in SLEEP journal, 2017, Jan 1; 40(1). doi:

10.1093/sleep/zsw032 and have been reproduced with permission from Oxford University Press.

List of Authors: Omonigho M Bubu MD, MPH1*, Michael Brannick PhD2, James Mortimer PhD1, Ogie

Umasabor-Bubu MD MPH1, Yuri V. Sebastião MPH1, Yi Wen MS3 , Skai Schwartz PhD1, Amy R.

Borenstein PhD1, Yougui Wu PhD1, David Morgan PhD4,5, William M. Anderson PhD6

1. University of South Florida, College of Public Health, Department of Epidemiology &

Biostatistics, 13201 Bruce B. Downs Avenue MDC56, Tampa, FL 33612

2. University of South Florida, College of Arts and Sciences, Psychology Department, 4202 East

Fowler Ave, PCD4118G, Tampa, FL 33620

3. University of South Florida, College of Engineering, Department of Chemical and Biomedical

Engineering, 4202 East Fowler Ave, PCD4118G, Tampa, FL 33620

4. University of South Florida, Morsani College of Medicine, Molecular Pharmacology and

Physiology, 12901 Bruce B. Downs Avenue MDC 8, Tampa, FL 33612

5. Byrd Alzheimer Institute, 4001 E. Fletcher Ave, Tampa, FL 33613

6. University of South Florida, Morsani College of Medicine, Sleep Medicine, Internal

Medicine,12901 Bruce B. Downs Avenue MDC 8, Tampa, FL 33612

Page 121: Omonigho A. Michael Bubu

109

Author contributions:

Bubu OM designed and performed the study and wrote all sections of the manuscript under the

supervision of committee members, Mortimer J, Morgan D, Anderson W, Borenstein AR, Wu Y and

Schwartz S. Brannick M supervised the statistical analysis. Umasabor-Bubu OQ, Sebastiao Y, and Wen Y

assisted in writing, editing and proofreading the article.

ABSTRACT

Study Objectives: Mounting evidence implicates disturbed sleep or lack of sleep as one of the risk

factors for Alzheimer’s disease (AD) but the extent of the risk is uncertain. We conducted a broad

systematic review and meta-analysis to quantify the effect of sleep problems/disorders on cognitive

impairment and AD.

Methods: Original published literature assessing any association of sleep problems or disorders with

cognitive impairment or AD was identified by searching PubMed, Embase, Web of Science, and the

Cochrane library. Effect estimates of individual studies were pooled and relative risks (RR) and 95%

confidence intervals (CI) were calculated using random effects models. We also estimated the population

attributable risk (PAR).

Results: Twenty-seven observational studies (n = 69,216 participants) that provided 52 RR estimates

were included in the meta-analysis. Individuals with sleep problems had a 1.55 (95% CI: 1.25-1.93), 1.65

(95% CI: 1.45-1.86) and 3.78 (95% CI: 2.27-6.30) times higher risk for AD, cognitive impairment and

preclinical AD than individuals without sleep problems respectively. The overall meta-analysis revealed

that individuals with sleep problems had a 1.68 (95% CI: 1.51-1.87) times higher risk for the combined

outcome of cognitive impairment and/or AD. Approximately 15% of AD in the population may be

attributed to sleep problems.

Conclusion: This meta-analysis confirmed the association between sleep and cognitive impairment or

AD and, for the first time, consolidated the evidence to provide an “average” magnitude of effect. As

Page 122: Omonigho A. Michael Bubu

110

sleep problems are of a growing concern in the population, these findings are of interest for potential

prevention of AD.

Statement of Significance: This study systematically and efficiently integrated existing information from

various studies, and for the first time, consolidated the evidence of the association between sleep and

Alzheimer’s disease (AD) and/or cognitive impairment by providing an “average” magnitude of the

effect. Subgroup meta-analysis of various aspects of sleep problems examined in this analysis each

significantly increased the risk for AD. The public health relevance of these findings is buttressed by the

population attributable risk percent, suggesting that approximately 15% of AD may be prevented should

interventions be implemented to reduce sleep problems and disorders. These novel findings are of

pertinent interest to further understand the etiology of AD and are likely to be of interest to researchers,

clinicians’ and public health policy makers.

Page 123: Omonigho A. Michael Bubu

111

INTRODUCTION

Alzheimer’s disease (AD) is a neurodegenerative disorder afflicting approximately five million

older Americans, with a projected annual incidence of one million cases in the United States by 2050.1, 2

AD and other dementias are associated with significant morbidity and mortality, with worldwide costs

estimated at US$604 billion in 2010.3 Preventive and/or therapeutic measures targeted at delaying or

curing AD are therefore imperative to mitigate this societal burden.

Mounting evidence implicates disturbed sleep or lack of sleep as a risk factor for AD. Demented

older adults exhibit significant sleep disturbance and unhealthy sleep patterns, including shorter sleep

duration and fragmented sleep,4-7 altered circadian rest/activity patterns,5 and elevated rates of sleep

disordered breathing (SDB).8, 9 It is estimated that about 45% of AD patients have sleep disturbances.10, 11

However, the ultimate etiologic role of sleep in AD pathogenesis remains unknown. Sources of sleep

disturbances in AD are thought to include degeneration of neural pathways that regulate sleep-wake

patterns and sleep architecture.10, 12 Sleep disorders also may exacerbate cognitive symptoms through

impairment of sleep-dependent memory consolidation processes.13, 14 It is possible that circadian rhythms

and sleep disorders have a causal role in the pathophysiology of AD. If this is true, they could be critical

to the goal of preventing this disease, because effective interventions exist to improve sleep.15 Narrative

review articles examining this association16-19 reflect the opinion and consensus among specialists that

there is growing experimental and epidemiologic evidence for a close reciprocal association of cognitive

impairment and AD with sleep problems and disorders. However, to date, no review has quantified the

effect of having sleep problems and disorders on cognitive impairment and AD. In this context, we

conducted a broad systematic review and meta-analysis to quantify the effect between sleep problems and

disorders on cognitive impairment and AD.

METHODS

Page 124: Omonigho A. Michael Bubu

112

We conducted this review in accordance with the step-by-step guide for systematic reviews and

meta-analysis by Pai et al.,20 and adhered to the preferred reporting items for systematic reviews and

meta-analyses (PRISMA) statement by Moher et al.,21 and the Cochrane handbook.22

Identification of eligible studies

Electronic search

A broad search strategy was employed to identify all eligible published articles in scientific

(medical, psychological and epidemiological) journals up to November 2014. This strategy combined

terms characterizing clinical AD or AD pathology as the outcome variable, sleep, as the exposure

variable, and a third set of terms specifying specific study types including cross-sectional, case control,

cohort (retrospective and prospective), Randomized Clinical Trials (RCTs), literature reviews, systematic

reviews, and ecological studies. All terms within each set were combined with the Boolean operator OR,

and the three sets were combined using AND. Articles were identified by searching four bibliographic

databases on November 21, 2014:

• Medline (through PubMed; all years 1946 - present)

• Embase (through www.embase.com by Elsevier B.V. 2011; all years from 1947 - present)

• Web of ScienceTM (Thomson Reuters 2014; all years from 1900 – present)

• Cochrane library (i.e., the Cochrane Central Register of Controlled Trials; all years from 1900 -

present).

Articles written in other languages other than English were also considered and screened with the help of

Google translator. The PubMed search (see supplemental Table 2A in appendix) was adapted to

searching the other databases.

Searching other sources

To locate additional relevant studies, the reference lists of articles identified through database

searches and the bibliographies of narrative review articles were examined. We also hand-searched the

previous year’s issues (November 2013 to November 2014) of high ranking journals that published more

Page 125: Omonigho A. Michael Bubu

113

than one relevant identified article during the initial search, for example Sleep Medicine Reviews,

Neurology, JAMA (journal of American medical association), Sleep Medicine, Sleep, Journal of Sleep

Research and Journal of Clinical Sleep Medicine.

Selection criteria

Type of studies

Original articles from observational studies (cross-sectional, case-control and

retrospective/prospective cohort) and randomized controlled trials (RCT) were included in this review.

Eligible studies for the meta-analysis were required to: i) have reported a primary or secondary data

analysis using any of the above mentioned study design of any association of sleep problems or disorders

with cognitive impairment or AD; ii) have an explicit measure of cognitive impairment or AD (e.g.

performance on individual cognitive tests, international classification of disease-9 (ICD -9), diagnostic

statistical manual-IV text revision (DSM-IV TR)) or objective measures of Alzheimer pathology (e.g.,

positron emission tomography (PET) scan); iii) have an explicit or obvious measure of sleep problems or

disorders (e.g., Pittsburgh sleep quality index (PQSI)), Epworth sleepiness scale (ESS)) or objective

measures (e.g., wrist actigraphy or polysomnography (PSG)); iv) study a comparison group without sleep

problems or disorders in cohort studies; and v) provide sufficient data to quantify the measure of

association of sleep problems or disorders with cognitive impairment or AD. The following studies were

excluded: i) case series, case reports, narrative reviews, abstracts or editorials, and ii) any RCT that did

not examine the cognitive effects of treating sleep and its related problems or disorders in AD.

Sleep problems and/or disorders

Sleep problems and/or disorders were considered as the risk factor of interest in this systematic

review. All sleep disorders described in the international classification of sleep disorders (ICSD-2)23 were

considered. Previous studies24, 25 employed various concepts to define sleep problems in order to fit their

Page 126: Omonigho A. Michael Bubu

114

study design. Accordingly, for sub-group analysis, we grouped eligible studies in this review measuring

sleep using parameters such as quality, efficiency and/or disturbance as sleep quality.

Alzheimer’s disease/cognitive impairment

The outcomes of interest in this review were cognitive impairment or AD. AD is the most

common form of dementia accounting for up to 50-80% of dementia cases.2 Studies that solely looked at

specific forms of dementia other than AD, e.g., vascular, Lewy body, or frontotemporal dementia, were

excluded.

Participants and setting

Participants of the included studies were middle aged and elderly adults of both sexes with age

ranging from 40 – 91 years. Studies were considered from multiple continents and no geographic

restrictions were imposed.

Data Extraction

An independent assessment of all titles and abstracts of eligible studies identified by the search

strategy was performed by two authors (OB, OU) using EndNote X6. Potential articles for the study were

retrieved and examined for possible inclusion by the same two authors. Discordant decisions regarding

inclusion were resolved by two other authors (YS and YW). Data extraction was performed by two

authors (OB, OU). Extracted fields included study design, study population, exposure and outcome

assessment, statistical methods and tests used, covariates, and the main results of the study. Differences in

the information extracted were resolved by two other authors (YS and YW). Reviewers were not blinded

to the funding, authors, or institutions.

Assessment of study quality

Study quality was assessed using an adaptation from previous systematic reviews26, 27 and the

modified version of the Newcastle-Ottawa scale (NOS) for quality assessment of the observational

studies,28, 29 with addition of new items relevant to this review. Parameters used for the quality assessment

Page 127: Omonigho A. Michael Bubu

115

included well-specified hypothesis, study design type, appropriately described sample, sample size,

definition of sleep and its related abnormalities, definition of cognitive impairment or AD, statistical

analytic methods, and inclusion of or approach used to adjust for potential confounders. For each

assessment category, a star rating system was used with specific maximum number of stars awarded (see

supplemental table 2). Studies were divided into three categories: low quality (< 50% of the maximum

number of stars), medium quality (≈ 55-70% of the maximum number of stars), and high quality (70% or

more of the maximum number of stars).

Statistical analysis

This systematic review consisted of both binary and continuous outcomes. Effect sizes from the

reviewed studies included odds ratios (OR), hazard ratios (HR), Pearson’s correlation coefficient (r), beta

estimates (β) and standardized mean differences (d). To proceed with the meta-analysis, we converted the

different indices to a common index: odds ratios (meta-analyses were computed using log (OR) but were

converted back to odds for presentation) (see Table 2B in supplemental material). Conversions

included converting from d to the log(OR) (Log(OR) = dπ

√� , where π is the mathematical constant

(approximately 3.14159)), converting from r to d (d = d��

√���� ), and converting from β to OR (exp(β) (see

Table 2C in supplemental material).30 The question of whether or not it is appropriate to combine effect

sizes from studies using varying metrics is under debate. We considered the fact that studies included in

the review and subsequent analyses had to meet certain inclusion and exclusion criteria and were judged

to be comparable in relevant ways. Since our aim was to compute a summary effect size of the impact of

sleep problems on AD, we accounted for multiple outcome measures on the same subjects by computing

the mean of the effect sizes for each study and used the average effect size as the unit of analysis (see

Table 2D in supplemental material). For example, we pooled the effect estimates in a single study that

had separate outcome measures using the mini mental state examination (MMSE) and the trail making B

test (Trails B) on the same subjects.31 This influenced the calculation of the variance of the sum and mean

effects of the correlated variables (see Table 2D in supplemental material). For our analysis, we worked

Page 128: Omonigho A. Michael Bubu

116

with a plausible range of correlations ranging from 0.5-1.0 and performed sensitivity analyses to find the

most appropriate standard error range. Our method could only have overestimated the variance and

underestimated the precision of our summary effect. Individual studies that examined the effect of sleep

on AD on different populations were treated as independent.32-38 Re-analyses of reported effect estimates

was conducted where appropriate in order to maintain consistency of definitions. For example, effect

estimates for sleep duration of ≤ 5 h, 6 h, and for <= 6.5 h were pooled to a single estimate for sleep

duration of <= 6.5 h.32, 38, 39 Furthermore, we converted the risk estimate for better sleep consolidation to a

risk estimate for poor sleep consolidation.40 Participants in the reviewed studies were between the ages of

40 -91. Because the prevalence of AD in this population and in the general population is just under 5%,2

no relevant artificial bias was introduced by pooling effects from cohort studies with effects from case-

control and cross-sectional studies. Therefore, we will refer to the pooled effect estimates as relative

risks, understanding that these are estimated in some studies. Effect estimates of all individual studies

were pooled and relative risks (RR) and 95% confidence intervals (CI) were calculated using random

effects models. Heterogeneity of results across studies was assessed using chi-square test and quantified

using I-square statistics.41 Subgroup analyses were conducted to explore potential causes of heterogeneity.

We anticipated heterogeneity and as such restricted subgroup analyses to situations where four or more

studies were available per subgroup, because heterogeneity is better assessed with multiple studies.41

Multivariate meta-regression analysis was also performed to examine the effect of possible influential

factors (i.e., sex, mean age, sample size and study quality). We assessed for publication bias using a

funnel plot asymmetry test and trim-and-fill. Additionally, the population attributable risk percent (PAR

%), which estimates the percentage of the outcome (AD) that would be prevented in the population if the

exposure (sleep problems or disorders) was eliminated, was calculated. All statistical analysis were

conducted using metaphor in R42 and SAS® 9.4.43

RESULTS

Study Selection

Page 129: Omonigho A. Michael Bubu

117

Figure 1 shows the study retrieval and selection process for the effects of sleep on cognitive

impairment and/or AD. Of the 2,341 publications identified initially from the literature search, 27 studies

were included in the meta-analysis (Figure 1). A total of 1,120 duplicates were removed. The remaining

1,221 publications were screened for titles resulting in the exclusion of 180 publications. After this

exclusion, 1,041 publications were screened using published abstracts, resulting in the exclusion of 840

publications that were either case series/reports, literature reviews, abstracts or editorials. Two hundred

and one full text articles were assessed for eligibility and 130 publications (105 observational studies and

25 RCTs) were further excluded because they were either review articles, did not provide any measure of

association, were not peer reviewed or because sleep and AD were not assessed. One study was included

from a search of the reference lists of articles identified through database searches. The remaining 72

studies were selected for the systematic review. For the meta-analysis, 45 publications were further

excluded because they lacked sufficient information needed for a meta-analysis, were duplicate studies

presenting similar or exact information from same study population or had an experimental study design,

leaving 27 studies with 52 effect estimates.

Study characteristics

Design and Quality

Among the 27 studies identified for the meta-analysis, there were eight cross-sectional31, 37, 44-49

one case-control.50 two retrospective cohort33, 51 and sixteen prospective cohort,32, 34, 36, 38-40, 52-61 (Table 1).

Of these, 15 were considered to be of high quality, ten were of medium quality and two were of low

quality. Selection bias related to sampling, measurements of sleep and/or AD solely based on self-report,

and insufficient adjustment for core confounders were the main limitations. All of the included articles in

the meta-analysis were written in English and published in peer-reviewed journals.

Participants and setting

Page 130: Omonigho A. Michael Bubu

118

The 27 studies included in the meta-analysis were published between 2001 and 2014 and included

a total of 69,216 participants from four continents (Table 1). Pooled relative risks included 25 estimates

from studies conducted in North America, 14 estimates from Europe, one from Australia, and four from

Asia (Table 2). Twenty-one studies included both sexes, of which three had separate analysis for men and

women, five were limited to women, and one was limited to men (Table 1).

Sleep problems/disorders

Sleep problems were based on a physician diagnosis (e.g. ICD-9 codes and apnea hypopnea index

(AHI) scores for sleep apnea), or were assessed using self-reported questionnaires or interviews (Table

1). There was considerable variability in the methods used to define and validate sleep problems. Nine

studies utilized objective measures (e.g., wrist actigraphy or PSG), while seven studies utilized

standardized questionnaires such as the PQSI or ESS. Five studies utilized self-constructed questionnaires

and one study made use of fluorescence immune-assay (FIA) to determine melatonin/hypocretin

concentrations.50 Most studies reported risk estimates for individual symptoms. Multiple symptoms were

also assessed in some studies. Sleep parameters assessed included (but were not limited to) sleep

duration, sleep quality, daytime sleepiness, sleep latency, sleep efficiency, wake after sleep onset, sleep

fragmentation, circadian rhythm abnormalities, obstructive sleep apnea, and insomnia. The interval over

which sleep problems was assessed extended from about a week to a median of 12.5 years.32

Cognitive impairment and/or Alzheimer’s disease

There also was significant variability in the methods used to diagnose cognitive impairment or

AD. Most studies used self-reported diagnoses with information collected to determine whether they

likely met standardized criteria (e.g. (DSM-IV TR) (Table 2.1). Cognitive function was assessed by

various global cognitive function and neuropsychological tests. Individuals were regarded as cognitively

normal with Mini-Mental State Examination (MMSE) scores between 27 and 30, and a Clinical Dementia

Rating of 0. For "Mild Cognitive Impairment," MMSE scores ranged from 22-27 according to a DSM-III-

Page 131: Omonigho A. Michael Bubu

119

R definition (ICD-10: 23-28). Studies utilizing objective measures of Alzheimer pathology (e.g., PET

scan for amyloid load (Aβ)) to examine associations between sleep and preclinical AD were also

considered because we intended to also describe possible potential mechanisms showing that sleep is

possibly associated with changes in AD biomarkers predictive of individuals who develop AD in

subsequent sub-group analyses..

Meta-analysis

Overall meta-analysis based on risk estimates for the effect of sleep on cognitive impairment and

Alzheimer’s disease

Sleep problems including short and long sleep duration, poor sleep quality, circadian rhythm

abnormality, insomnia and obstructive sleep apnea, were associated with a significantly increased relative

risk (RR) for the combined outcome of cognitive impairment and AD of 1.68 (95% CI: 1.51-1.87) (Fig.

2.2). The relative risks ranged from 1.0734 to 4.2549 with two low outliers; 0.56,61 and 0.76,32 and five

high outliers 5.10,50 5.13, and 5.85,59 6.08,33 and 8.17.37 Removing these 7 outliers from the analysis had

no significant effect on the pooled RR (RR: 1.62, 95% CI: 1.47-1.79). There was significant heterogeneity

among the effect sizes (I-square 64.50%, p < 0.001). The random-effects variance component was .05

(95% CI: .023-.195). A prediction interval that considers both sampling error and random-effects

variance showed relative risk values ranging from 1.05 to 2.50. Removal of the 7 outliers reduced

heterogeneity but it remained statistically significant (I-square 54.36%, p < 0.001), indicating that these

outliers were not the sole source of heterogeneity.

Sub-group meta-analysis based on risk estimates for the effect of sleep on cognitive impairment, Pre-

clinical and ICD9/DSMIV Diagnoses of Alzheimer’s disease

Separate analyses of the studies in terms of the outcome measures were also conducted. Our

outcome measures included the use of cognitive tests by studies examining sleep and the risks of

cognitive impairment; the use of AD biomarkers or abnormal proteins by studies examining sleep and the

Page 132: Omonigho A. Michael Bubu

120

risk of preclinical AD; and the use of ICD9/DSMIV diagnoses of AD by studies examining the risk of

sleep and symptomatic AD respectively. Sleep problems including short and long sleep duration, poor

sleep quality, circadian rhythm abnormality, insomnia and obstructive sleep apnea, were associated with

significantly increased relative risks of cognitive impairment (RR: 1.64, 95% CI: 1.45-1.87) ), preclinical

AD (RR: 3.78, 95% CI: 2.27-6.30) and ICD9/DSMIV diagnoses of AD (RR: 1.55, 95% CI: 1.25-1.93)

respectively (Fig. 2.3) These results highlight potential mechanistic relationships suggesting that sleep is

not only associated with cognitive impairment, or symptomatic AD but is also associated with changes in

AD biomarkers predictive of persons that ultimately develop AD.

Funnel Plot Asymmetry Test and Trim and Fill Analysis Assessment of Publication Bias

Regarding possible publication bias, a funnel plot asymmetry test was significant (z = 5.5109, p <

.0001),62 indicating that some form of publication bias exist. Furthermore, a trim-and-fill analysis63

(which aims both to identify and correct for funnel plot asymmetry arising from publication bias by

essentially ‘trimming’ or removing the smaller studies causing funnel plot asymmetry, and then using the

trimmed funnel plot to estimate the true ‘center’ of the funnel, while replacing the omitted studies and

their missing ‘counterparts’ around the center (filling)) imputed 17 unpublished studies and confirmed

that in smaller studies, weaker effects were less often published (Fig. 2.4). The trim and fill method does

not require any assumptions regarding mechanisms leading to bias in publication, rather it provides an

estimate of the number of missing studies, and calculates an estimated effect ‘adjusted’ for the publication

bias (based on the filled studies).

Sub-group meta-analysis based on risk estimates for the effect of different sleep problems and

disorders on cognitive impairment and Alzheimer’s disease

Analyzing various sleep problems and disorders (Fig. 2.5), the highest relative risk for AD and/or

cognitive impairment was noted for obstructive sleep apnea (OSA) (RR: 2.37, 95% CI: 1.82-3.08). The

relative risk for cognitive impairment and/or AD associated with sleep quantity was 1.86 (95% CI: 1.34-

Page 133: Omonigho A. Michael Bubu

121

2.57), while sleep quality was 1.62 (95% CI: 1.34-1.97). Insomnia (RR: 1.38 95% CI: 1.13-1.67) and

circadian rhythm abnormalities (RR: 1.38, 95% CI: 1.18-1.61) were associated with the lowest relative

risks. After excluding the outliers, the relative risk for cognitive impairment or AD associated with OSA,

sleep quantity and sleep quality remained statistically significant (RR: 2.30, 95% CI: 1.59-2.41; RR:

1.72, 95% CI: 1.24-2.61 and RR: 1.51, 95% CI: 1.244-1.81 respectively).

Subgroup analyses (Table 2.2) investigating various sleep parameters revealed that poor sleep

efficiency was associated with the highest risk for cognitive impairment or AD, followed by sleep latency

and wake after sleep onset (WASO). Nighttime problems were associated with the strongest measure of

effect for cognitive impairment or AD, with smaller effects seen for night- and daytime problems. When

examined separately, men and women with sleep problems including short and long sleep duration, poor

sleep quality, circadian rhythm abnormality, insomnia and obstructive sleep apnea, each had similar

relative risks. The risk for cognitive impairment or AD due to sleep duration was higher in long sleepers

(> 8 h/day) when compared to normal sleepers (7 – 8 h/day), than in short sleepers (≤ 7 h/day). Studies

with ≥ 5 years of follow-up reported higher relative risks due to sleep problems including short and long

sleep duration, poor sleep quality, circadian rhythm abnormality, insomnia and obstructive sleep apnea,

than those with < 5 years of follow-up. Studies from North America and Europe reported similar relative

risks due to sleep problems. Prospective studies showed slightly lower relative risks than non-prospective

studies, perhaps indicating a problem with temporality. Finally, the estimated risk for cognitive

impairment or AD due to sleep problems was lower in the high-quality studies than in the low-quality

studies.

Sub-group meta-analysis based on risk estimates for the effect of self-report and actigraphic data of

poor sleep on cognitive impairment and/or Alzheimer’s disease

Furthermore, we analyzed studies that generated data on total sleep times (measured

actigraphically) and examined these effects separately in relation to cognition from more generally

defined poor sleep or self-reported sleep durations (Fig. 2.5). The relative risk for cognitive impairment

Page 134: Omonigho A. Michael Bubu

122

or AD associated with self-reported poor sleep was 1.49 (95% CI: 1.26-1.76), while actigraphic report

was 1.80 (95% CI: 1.56-2.09). These findings somewhat corroborate effects of self-reported sleep

durations in relation to cognition examined in terms of actigraphic data.

Exploring Potential Causes of Heterogeneity

The overall meta-analysis included a wide array of studies including different measures with

varying neurobiological substrates (breathing control in sleep, circadian rhythmicity, sleep durations,

perceived sleep depth) as well as varying outcome measures of AD, including ICD9/DSM diagnoses,

cognitive tests and abnormal proteins; and this could possibly be a potential source of heterogeneity.

Therefore, we explored this and other potential causes for heterogeneity and conducted several subgroup

meta-analyses (Figures 2.4-2.6 and Table 2.2) (also see Figures 2A-2C in supplemental material). The

differences seen across the subgroups failed to account for significant amounts of heterogeneity except for

the varying outcome measures of AD and sample size. However, tendencies could be observed. As

sample size increased, the effect size decreased. This agreed with the funnel plot. The varying outcome

measures of AD accounted for 12.78% heterogeneity. Several of the other subgroup analyses were close

to being statistically significant; however, there were only 52 effect sizes in the analysis, so power may

have been an issue for some sub-analyses.

Sensitivity Analyses

Sensitivity analysis incorporating only studies that examined the relationship between sleep and

cognitive impairment and/or symptomatic AD for the overall meta-analysis yielded a RR of 1.62 (95%

CI: 1.45-1.80), suggesting that exclusion of studies that examined sleep associations with preclinical AD

could reduce the overall effect size, however, the change in effect size was not statistically significant.

Similarly, removing effect estimates of outliers from the analysis had no significant effect on the pooled

RR (RR: 1.62, 95% CI: 1.47-1.79). Furthermore, exclusion of studies examining sleep associations with

preclinical AD for the sub-group meta-analysis based on risk estimates for the effect of different sleep

Page 135: Omonigho A. Michael Bubu

123

problems and disorders on cognitive impairment and Alzheimer’s disease produced similar relative risks

for the relationships between OSA, sleep quantity, sleep quality, insomnia and cognitive impairment

and/or AD.

Sensitivity analyses incorporating the 17 studies imputed by trim-and-fill showed an estimated

relative risk of 1.42 (95% CI: 1.28-1.58), suggesting that bias could reduce the size of the effect, but is

unlikely to explain it entirely. It should also be noted that the small sample, large effect size studies were

mostly based on results of studies in which correlations were converted to odds ratios, so there may be a

methodological explanation of funnel asymmetry other than bias. Altogether, the results support an

association between sleep problems and cognitive impairment or AD. Despite possible publication bias

and evidence of heterogeneity in the effect sizes, the overall association between sleep problems and

cognitive impairment or AD remained positive in all sensitivity analyses.

Furthermore, we clearly understand that testing for heterogeneity may not explain the variability

in the data because its basis may not simply be empirical but more fundamentally conceptual, therefore

we conducted several sensitivity analyses on both our exposure and outcome. Sensitivity analyses

examining the effect of omitting one study revealed no effect as the estimated relative risk remained

stable (See Fig. 2D in supplemental material). Sensitivity analyses were also conducted to see if all of

the subcategories of exposure can be combined into one exposure. We examined the effect of the

exposure on cognitive impairment and AD initially combined as presented in the overall meta-analysis

(Fig. 2.2) and while excluding each subcategory (Sleep Quality, Sleep Quantity, Obstructive Sleep

Apnea, Circadian Rhythm Abnormalities and Insomnia) from the exposure one at a time. No significant

change in the effect size was seen. We also conducted separate analyses of studies in terms of the

outcome measure since different outcome measures of cognitive impairment and AD was used. We

conducted analyses with the combined outcome of cognitive impairment and AD and examined the

change in the estimate while excluding each subcategory (cognitive tests, ICD-9/DSM IV and abnormal

Page 136: Omonigho A. Michael Bubu

124

proteins). No statistically significant difference was observed in the change in effect estimate. (Type I

error rate (α=0.05)).

Meta-regression analysis

A multivariate meta-regression analysis (Table 2.3) was conducted to examine the potential

influence of different factors on the natural logarithm of the odds ratio of sleep problems with cognitive

impairment or AD. None of the studied factors was statistically significant except for sample size (p =

.01), with larger samples showing smaller effects.

Population attributable risk percent

The PAR% suggested that approximately 15% of AD may be attributed to sleep problems, using

a conservative estimate of 30% as the prevalence of sleep problems in the populations of the included

studies and the pooled relative risk (RR: 1.55). By comparison, the prevalence of sleep problems in AD

ranges between 25% – 50%.11

DISCUSSION

Summary of the main results

In the present meta-analysis comprising 27 studies, individuals with sleep problems including

short and long sleep duration, poor sleep quality, circadian rhythm abnormality, insomnia and obstructive

sleep apnea, had a 1.68 times higher risk for the combined outcome of cognitive decline and AD

compared to individuals without sleep problems. Individuals with sleep problems including short and

long sleep duration, poor sleep quality, circadian rhythm abnormality, insomnia and obstructive sleep

apnea, had a 1.55, 1.65 and 3.78 times higher risk for AD, cognitive impairment and preclinical AD than

individuals without sleep problems respectively. A subgroup meta-analysis of classified sleep disorders

revealed a relative risk of 2.37 for obstructive sleep apnea, 1.86 for sleep duration problems, 1.62 for

sleep quality problems, and 1.38 for both insomnia and circadian rhythm abnormalities. Various aspects

of sleep problems examined in this analysis each significantly increased the risk for cognitive impairment

Page 137: Omonigho A. Michael Bubu

125

or AD. Approximately 15% of cognitive impairment or AD may be attributed to sleep problems including

short and long sleep duration, poor sleep quality, circadian rhythm abnormality, insomnia and obstructive

sleep apnea.

Effect of sleep problems on cognitive impairment or Alzheimer’s disease (meta-analysis)

Altogether, sleep problems including short and long sleep duration, poor sleep quality, circadian

rhythm abnormality, insomnia and obstructive sleep apnea, significantly increased cognitive impairment

or AD risk. Our findings show a RR increase seemingly inverse to diagnostic confidence (e.g., 1.55, 1.65

and 3.78 for AD, MCI and preclinical AD). These findings may reflect the use of more objective

investigative or diagnostic measures to identify individuals with preclinical AD in our study, however it

highlights potential mechanistic relationships suggesting that sleep is not only associated with cognitive

impairment, or symptomatic AD but is also associated with changes in AD biomarkers predictive of

persons that ultimately develop AD. Although there was heterogeneity in effect sizes, the variables

included in the meta-analysis failed to explain significant amounts of this variability. OSA appeared to be

a strong risk factor for AD. This may be explained by the fact that hypoxia plays a vital role and may

contribute to increases in Aβ production64, 65 thereby facilitating AD pathogenesis.66 Insomnia and

circadian rhythm abnormalities were each associated with an intermediate risk for cognitive impairment

or AD. Sleep quantity and quality seemed more important than insomnia and circadian activity rhythm as

a risk factor for cognitive impairment or AD. This could be because individuals with insomnia and/or

circadian rhythm abnormalities tend to have poor sleep quantity and quality, and as such the effect seen

could possibly reflect other underlying sleep problems such that individuals in the sleep quantity or

quality groups may also have associated insomnia and/or circadian rhythm abnormalities. Both long sleep

(> 8 h/day) and short sleep duration (< 7 h/day) increased the risk for cognitive impairment or AD when

compared to normal sleepers (7 – 8 h/day), with a higher risk seen in long sleepers. Although certain

prospective studies have shown associations between long sleep and cognitive impairment,67, 68 it is highly

plausible that this association is as a result of a general decline with old age and maybe reflective of other

Page 138: Omonigho A. Michael Bubu

126

disease conditions such as depression or other neurological disorders. AD patients, like in Parkinson’s

disease, may also exhibit a more bimodal distribution of sleep patterns (i.e. short or long) possibly

reflective of some pre-existing genetic predisposition that develops at disease inception.

Potentially influencing factors (meta-regression analysis)

The heterogeneity seen across studies could not be explained by our subgroup meta-analyses,

except for sample size and the different AD measures. It is likely that much of the heterogeneity can be

explained by the conceptual differences in exposure and/or outcome assessment. We explored this further

by conducting several sensitivity analyses on both our exposure and outcome, each initially combined as

presented in the overall meta-analysis and also with exclusion of each subcategory one at a time. No

significant change in the effect size was seen. Sensitivity analyses examining the effect of omitting one

study revealed no effect as the estimated relative risk remained stable. Several of the subgroup analyses

were close to being statistically significant. However, since there were only 52 effect sizes in the analysis,

power may have been an issue. Pooled relative risks from sub-group meta-analyses for the effect of sleep

problems on cognitive impairment or AD revealed consistent moderate to strong relative risks and these

relationships could possibly contribute to a causal interpretation of the findings. No RCT was included,

suggesting that additional work examining the effect of sleep therapy on cognitive impairment or AD

would be useful.

Among the factors tested only sample size significantly influenced the effect size between sleep

problems and AD such that larger sample size studies tended to result in smaller risk estimates for

cognitive impairment or AD while smaller sample size studies tended to result in larger risk estimates for

cognitive impairment or AD (Table 3). Overall, the results indicate that the meta-analyzed estimate

reflects an appropriate estimate of the general association. The need for proper multivariate modeling

approaches is underscored by the fact that the heterogeneity due to adjustment was close to statistical

significance in some instances, although power was limited.

Study results in comparison with other reviews on the topic

Page 139: Omonigho A. Michael Bubu

127

The results of this systematic review and meta-analysis are largely consistent with previous

reviews. Our study is the first systematic review and meta-analysis to quantify an “average” magnitude of

the association between sleep problems and cognitive impairment or AD. Previous narrative review

articles that have examined this association did so with a focus on different aspects of sleep

disturbances/disorders16-19. Palma et al.,16 focusing on sleep deprivation, reviewed how sleep loss

constitutes a risk factor for various neurologic diseases including AD. Slats et al.,18 focusing on the role of

hypocretin and melatonin, concluded that there is substantial evidence for impairments in both sleep and

circadian regulating mechanisms resulting from AD pathology, which may be linked to clinical

symptoms. Peter-Derex et al.,17 performed a thorough clinical review of sleep and AD, observing that

micro-architectural sleep alterations, nocturnal sleep fragmentation, decreased sleep duration, diurnal

napping and inversion of the sleep-wake cycle are the main disorders observed in patients with AD. Spira

et al.,19 concluded that poor sleep is a risk factor for cognitive impairment and AD and that healthy sleep

appears to play an important role in maintaining brain health with age, and could play a key role in AD

prevention. Various reviews69, 70 on the effect of OSA on AD agree on a bidirectional relationship

between these entities.

Strengths and Limitations of the study

Our systematic review allows for a systematic, scientific, constructive and yet replicable review

of the association between sleep and AD with a pre-specified hypothesis and specific inclusion and

exclusion criteria. We were able to systematically and efficiently integrate existing information from

various studies enabling us to establish whether the literature is consistent and can be generalized across

populations, settings, and/or whether findings vary significantly by particular subsets. The meta-analyses

in particular increased the power and precision of estimates of the risk of cognitive impairment or AD

from sleep problems. The explicit methods employed in our meta-analysis limit bias and, hopefully,

improved reliability and accuracy of our conclusions. Lastly, the generalizability of our findings can be

Page 140: Omonigho A. Michael Bubu

128

established as this review included a diversity of studies across various continents, providing an

interpretive context not available in any one study.

Several methodological concerns must be considered when interpreting the findings of this meta-

analysis. First, one potential limitation relates to the possible bias introduced in the paper selection

process. To minimize this bias, we adhered strictly to the guidelines and recommendations in the

Cochrane handbook 22 whenever possible. We also ensured that studies were not restricted by language to

guard against language bias. Second, the possible bias introduced by the individual studies that were

included is a further limitation. Various study designs were rated low for assigning the grade of

evidence71. With a scarcity of controlled studies, this analysis relied mostly on observational studies.

However, the subgroup meta-analysis revealed no significant difference between the prospective and non-

prospective studies, suggesting that our average estimate adequately reflects the general effect. In addition

to study design, conclusions made from the poor quality studies could introduce possible bias. We

included high, medium and low-quality studies in the summarization of findings. Our meta-regression

analysis indicated that study quality might not have influenced the overall conclusion. Third, there was

considerable variability between the studies in the assessment methods used to verify sleep problems and

AD or cognitive impairment, revealing moderate heterogeneity that could possibly raise questions about

the comparability of results across studies. Our research question focused on a broad concept of sleep

problems based on having any symptoms. Thus, we assumed a common underlying concept by making

general conclusions regarding sleep problems and cognitive impairment or AD. We based our assumption

on findings from reviews that consistently found associations of various sleep problems or disorders such

as sleep deprivation/loss,16, 19 sleep-wake abnormalities,17, 18 or sleep apnea,69, 70 with cognitive

impairment or AD. Moreover, our sensitivity analyses on the exposure and outcome measures which

involved examining the effect of sleep problems on cognitive impairment and AD with the exposure or

outcome measures combined and separated into subcategories did not reveal any statistical significant

change in the estimate. The persistence of heterogeneity in the subgroup meta-analysis of sleep

Page 141: Omonigho A. Michael Bubu

129

parameters and various sleep problems and disorders suggests within-study variation, possibly due to lack

of objective assessment, standardization or success in controlling for psychiatric and somatic co-

morbidities. However, the prediction interval that considered both sampling error and study heterogeneity

suggested that although the association may vary across studies, it appears to be positive in at least 97.5

percent of them. Finally, although funnel plot asymmetry was consistent with publication bias, trim-and-

fill results were also positive; suggesting that our overall result might be an overestimate, but that the

association does not appear to be explained by biased reporting of studies.

Future directions

Existing reviews note growing experimental and epidemiologic evidence for close reciprocal

associations between cognitive impairment and sleep alteration. However, longer-term longitudinal

studies are required to better understand whether sleep problems precede the onset of cognitive

impairment or AD by many years. Future studies need to be able to define causal pathways more

definitively so we can test whether neurodegeneration is leading to both sleep changes and memory

changes, or whether aging is contributing to OSA and/or sleep architecture changes that in turn affect

memory, or whether the effect of aging on sleep architecture predisposes to both apnea and memory

impairment. More research with objective sleep measures, AD biomarkers, and cognitive measures also

are needed. To facilitate appropriate comparisons among studies, it would be expedient to have greater

standardization of sleep and measures of cognitive impairment or AD. Future research is needed to make

clear the contribution of different genetic, neurodegenerative and environmental factors to sleep

impairment, as their findings can be promising for the development of new prevention and/or treatment

opportunities.

Conclusion

This meta-analysis confirmed the association between sleep and cognitive impairmentor AD and,

for the first time, consolidated the evidence by providing an “average” magnitude of the effect (RR=1.55,

95% CI: 1.25-1.93). The public health relevance of this finding is buttressed by the PAR%, suggesting

Page 142: Omonigho A. Michael Bubu

130

that approximately 15% of cognitive impairment or AD might be prevented if effective interventions

could be implemented to reduce sleep disorders.

References

1. Hebert LE, Beckett LA, Scherr PA, Evans DA. Annual incidence of Alzheimer disease in the

United States projected to the years 2000 through 2050. Alzheimer disease and associated disorders

2001;15:169-73.

2. Thies W, Bleiler L. 2013 Alzheimer's disease facts and figures. Alzheimer's & dementia : the

journal of the Alzheimer's Association 2013;9:208-45.

3. Wimo A, Jonsson L, Bond J, Prince M, Winblad B. The worldwide economic impact of dementia

2010. Alzheimer's & dementia : the journal of the Alzheimer's Association 2013;9:1-11 e3.

4. Bliwise DL. Sleep in normal aging and dementia. Sleep 1993;16:40-81.

5. Prinz PN, Peskind ER, Vitaliano PP, et al. Changes in the sleep and waking EEGs of

nondemented and demented elderly subjects. Journal of the American Geriatrics Society 1982;30:86-93.

6. Prinz PN, Vitaliano PP, Vitiello MV, et al. Sleep, EEG and mental function changes in senile

dementia of the Alzheimer's type. Neurobiol Aging 1982;3:361-70.

7. Vitiello MV, Prinz PN, Williams DE, Frommlet MS, Ries RK. Sleep disturbances in patients with

mild-stage Alzheimer's disease. Journal of gerontology 1990;45:M131-8.

8. Hoch CC, Reynolds CF, 3rd, Kupfer DJ, Houck PR, Berman SR, Stack JA. Sleep-disordered

breathing in normal and pathologic aging. The Journal of clinical psychiatry 1986;47:499-503.

9. Mant A, Saunders NA, Eyland AE, Pond CD, Chancellor AH, Webster IW. Sleep-related

respiratory disturbance and dementia in elderly females. Journal of gerontology 1988;43:M140-4.

10. McCurry SM, Reynolds CF, Ancoli-Israel S, Teri L, Vitiello MV. Treatment of sleep disturbance

in Alzheimer's disease. Sleep medicine reviews 2000;4:603-28.

11. Moran M, Lynch CA, Walsh C, Coen R, Coakley D, Lawlor BA. Sleep disturbance in mild to

moderate Alzheimer's disease. Sleep Med 2005;6:347-52.

Page 143: Omonigho A. Michael Bubu

131

12. Meeks TW, Ropacki SA, Jeste DV. The neurobiology of neuropsychiatric syndromes in

dementia. Current opinion in psychiatry 2006;19:581-6.

13. Rauchs G, Schabus M, Parapatics S, et al. Is there a link between sleep changes and memory in

Alzheimer's disease? Neuroreport 2008;19:1159-62.

14. Rauchs G HC, Bertran F, Desgranges B, Eustache F. . Sleep and episodic memory: a review of

the literature in young healthy subjects and potential links between sleep changes and memory

impairment observed during aging and Alzheimer’s disease. . Rev Neurol (Paris) 2010; 2010;166::81.

15. Bloom HG, Ahmed I, Alessi CA, et al. Evidence-based recommendations for the assessment and

management of sleep disorders in older persons. Journal of the American Geriatrics Society 2009;57:761-

89.

16. Palma JA, Urrestarazu E, Iriarte J. Sleep loss as risk factor for neurologic disorders: a review.

Sleep medicine 2013;14:229-36.

17. Peter-Derex L, Yammine P, Bastuji H, Croisile B. Sleep and Alzheimer's disease. Sleep medicine

reviews 2014.

18. Slats D, Claassen JA, Verbeek MM, Overeem S. Reciprocal interactions between sleep, circadian

rhythms and Alzheimer's disease: focus on the role of hypocretin and melatonin. Ageing research reviews

2013;12:188-200.

19. Spira AP, Chen-Edinboro LP, Wu MN, Yaffe K. Impact of sleep on the risk of cognitive decline

and dementia. Current opinion in psychiatry 2014;27:478-83.

20. Pai M, McCulloch M, Gorman JD, et al. Systematic reviews and meta-analyses: an illustrated,

step-by-step guide. The National medical journal of India 2004;17:86-95.

21. Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and

meta-analyses: the PRISMA statement. BMJ (Clinical research ed.) 2009;339:b2535.

22. Higgins JPT GSe. Cochrane handbook for systematic reviews of interventions. Version 5.1.0

[updated March 2011]. . http://www.cochranehandbook.org [accessed 11.23.14]. 2011.

Page 144: Omonigho A. Michael Bubu

132

23. Medicine. AAoS. The international classification of sleep disorders, revised: diagnostic and

coding manual. Chicago, USA: . American Academy of Sleep Medicine, ISBN 0-9657220-1-5; 2001.

2001.

24. Kling RN, McLeod CB, Koehoorn M. Sleep problems and workplace injuries in Canada. Sleep

2010;33:611-8.

25. Uehli K, Mehta AJ, Miedinger D, et al. Sleep problems and work injuries: a systematic review

and meta-analysis. Sleep medicine reviews 2014;18:61-73.

26. Bhui K, Stansfeld S, Hull S, Priebe S, Mole F, Feder G. Ethnic variations in pathways to and use

of specialist mental health services in the UK. Systematic review. The British journal of psychiatry : the

journal of mental science 2003;182:105-16.

27. Dinos S, Khoshaba B, Ashby D, et al. A systematic review of chronic fatigue, its syndromes and

ethnicity: prevalence, severity, co-morbidity and coping. Int J Epidemiol 2009;38:1554-70.

28. Wells G SB, O’Connell D, Peterson J,Welch V, Losos M, et al. . The Newcastle-Ottawa scale

(NOS) for assessing the quality of nonrandomised studies in metaanalyses,.

http://www.ohri.ca/programs/clinical_epidemiology/oxford.asp/; 2011 [accessed 11.25.14]. 2011.

29. D. P. Personal communication 2011. 2011.

30. Borenstein Michael HLV HJPT, Rothstein H.R. Introduction to Meta-Analysis. Chichester, U.K.:

John Wiley & Sons, 2009. , © 2009:45-9.

31. Blackwell T, Yaffe K, Ancoli-Israel S, et al. Poor sleep is associated with impaired cognitive

function in older women: the study of osteoporotic fractures. The journals of gerontology. Series A,

Biological sciences and medical sciences 2006;61:405-10.

32. Benito-Leon J, Bermejo-Pareja F, Vega S, Louis ED. Total daily sleep duration and the risk of

dementia: a prospective population-based study. Eur J Neurol 2009;16:990-7.

33. Chang WP, Liu ME, Chang WC, et al. Sleep apnea and the risk of dementia: a population-based

5-year follow-up study in taiwan. PLoS One 2013;8:e78655.

Page 145: Omonigho A. Michael Bubu

133

34. Cricco M, Simonsick EM, Foley DJ. The impact of insomnia on cognitive functioning in older

adults. Journal of the American Geriatrics Society 2001;49:1185-9.

35. Gottlieb DJ, DeStefano AL, Foley DJ, et al. APOE epsilon4 is associated with obstructive sleep

apnea/hypopnea: the Sleep Heart Health Study. Neurology 2004;63:664-8.

36. 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.

37. Osorio RS, Ayappa I, Mantua J, et al. The interaction between sleep-disordered breathing and

apolipoprotein E genotype on cerebrospinal fluid biomarkers for Alzheimer's disease in cognitively

normal elderly individuals. Neurobiol Aging 2014;35:1318-24.

38. 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 disease and

associated disorders 2006;20:41-8.

39. Keage HA, Banks S, Yang KL, Morgan K, Brayne C, Matthews FE. What sleep characteristics

predict cognitive decline in the elderly? Sleep medicine 2012;13:886-92.

40. Lim AS, Yu L, Kowgier M, Schneider JA, Buchman AS, Bennett DA. Modification of the

Relationship of the Apolipoprotein E epsilon4 Allele to the Risk of Alzheimer Disease and

Neurofibrillary Tangle Density by Sleep. JAMA neurology 2013;70:1544-51.

41. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses.

BMJ (Clinical research ed.) 2003;327:557-60.

42. Viechtbauer W. Conducting meta-analyses in R with the metafor package. . Journal of Statistical

Software, (2010). ;36:1–48. .

43. Inc. SI. SAS® 9.4 System Guide to Software Updates Cary NC, USA. SAS Institute Inc. 2013.

44. Ju YE, McLeland JS, Toedebusch CD, et al. Sleep quality and preclinical Alzheimer disease.

JAMA neurology 2013;70:587-93.

45. Merlino G, Piani A, Gigli GL, et al. Daytime sleepiness is associated with dementia and cognitive

decline in older Italian adults: A population-based study. Sleep medicine 2010;11:372-7.

Page 146: Omonigho A. Michael Bubu

134

46. Nebes RD, Buysse DJ, Halligan EM, Houck PR, Monk TH. Self-reported sleep quality predicts

poor cognitive performance in healthy older adults. The journals of gerontology. Series B, Psychological

sciences and social sciences 2009;64:180-7.

47. Oosterman JM, van Someren EJ, Vogels RL, Van Harten B, Scherder EJ. Fragmentation of the

rest-activity rhythm correlates with age-related cognitive deficits. Journal of sleep research 2009;18:129-

35.

48. Saint Martin M, Sforza E, Barthelemy JC, Thomas-Anterion C, Roche F. Does subjective sleep

affect cognitive function in healthy elderly subjects? The Proof cohort. Sleep medicine 2012;13:1146-52.

49. Spira AP, Gamaldo AA, An Y, et al. Self-reported sleep and beta-amyloid deposition in

community-dwelling older adults. JAMA neurology 2013;70:1537-43.

50. Schmidt FM, Kratzsch J, Gertz HJ, et al. Cerebrospinal fluid melanin-concentrating hormone

(MCH) and hypocretin-1 (HCRT-1, orexin-A) in Alzheimer's disease. PloS one 2013;8:e63136.

51. Sterniczuk R, Theou O, Rusak B, Rockwood K. Sleep disturbance is associated with incident

dementia and mortality. Current Alzheimer research 2013;10:767-75.

52. Blackwell T, Yaffe K, Laffan A, et al. 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.

53. Grandner MA, Drummond SP. Who are the long sleepers? Towards an understanding of the

mortality relationship. Sleep medicine reviews 2007;11:341-60.

54. Foley D, Monjan A, Masaki K, et al. Daytime sleepiness is associated with 3-year incident

dementia and cognitive decline in older Japanese-American men. Journal of the American Geriatrics

Society 2001;49:1628-32.

55. Hahn EA, Wang HX, Andel R, Fratiglioni L. A Change in Sleep Pattern May Predict Alzheimer

Disease. The American journal of geriatric psychiatry : official journal of the American Association for

Geriatric Psychiatry 2013.

Page 147: Omonigho A. Michael Bubu

135

56. Jelicic M, Bosma H, Ponds RW, Van Boxtel MP, Houx PJ, Jolles J. Subjective sleep problems in

later life as predictors of cognitive decline. Report from the Maastricht Ageing Study (MAAS).

International journal of geriatric psychiatry 2002;17:73-7.

57. Osorio RS, Pirraglia E, Aguera-Ortiz LF, et al. Greater risk of Alzheimer's disease in older adults

with insomnia. Journal of the American Geriatrics Society 2011;59:559-62.

58. Tranah GJ, Blackwell T, Stone KL, et al. Circadian activity rhythms and risk of incident dementia

and mild cognitive impairment in older women. Annals of neurology 2011;70:722-32.

59. Virta JJ, Heikkila K, Perola M, et al. Midlife sleep characteristics associated with late life

cognitive function. Sleep 2013;36:1533-41, 41A.

60. Walsh CM, Blackwell T, Tranah GJ, et al. Weaker circadian activity rhythms are associated with

poorer executive function in older women. Sleep 2014;37:2009-16.

61. Yaffe K, Laffan AM, Harrison SL, et al. Sleep-disordered breathing, hypoxia, and risk of mild

cognitive impairment and dementia in older women. JAMA : the journal of the American Medical

Association 2011;306:613-9.

62. Viechtbauer W. Conducting meta-analyses in R with the metafor package. Journal of Statistical

Software 2010;36:1-48.

63. Duval S, Tweedie R. Trim and fill: A simple funnel-plot-based method of testing and adjusting

for publication bias in meta-analysis. Biometrics 2000;56:455-63.

64. Li L, Zhang X, Yang D, Luo G, Chen S, Le W. Hypoxia increases Abeta generation by altering

beta- and gamma-cleavage of APP. Neurobiology of aging 2009;30:1091-8.

65. Zhang X, Zhou K, Wang R, et al. Hypoxia-inducible factor 1alpha (HIF-1alpha)-mediated

hypoxia increases BACE1 expression and beta-amyloid generation. The Journal of biological chemistry

2007;282:10873-80.

66. Sun X, He G, Qing H, et al. Hypoxia facilitates Alzheimer's disease pathogenesis by up-

regulating BACE1 gene expression. Proceedings of the National Academy of Sciences of the United

States of America 2006;103:18727-32.

Page 148: Omonigho A. Michael Bubu

136

67. Benito-Leon J, Louis ED, Bermejo-Pareja F. Cognitive decline in short and long sleepers: a

prospective population-based study (NEDICES). J Psychiatr Res 2013;47:1998-2003.

68. Johar H, Kawan R, Emeny RT, Ladwig KH. Impaired Sleep Predicts Cognitive Decline in Old

People: Findings from the Prospective KORA Age Study. Sleep 2016;39:217-26.

69. Gagnon K, Baril AA, Gagnon JF, et al. Cognitive impairment in obstructive sleep apnea.

Pathologie-biologie 2014.

70. Kinugawa K, Nguyen-Michel VH, Mariani J. [Obstructive sleep apnea syndrome: a cause of

cognitive disorders in the elderly?]. La Revue de medecine interne / fondee ... par la Societe nationale

francaise de medecine interne 2014;35:664-9.

71. Atkins D, Best D, Briss PA, et al. Grading quality of evidence and strength of recommendations.

BMJ (Clinical research ed.) 2004;328:1490.

Page 149: Omonigho A. Michael Bubu

137

Table 2.1. Characteristics of included studies. Meta-analysis of sleep, cognitive decline and Alzheimer's disease/cognitive decline

Authors, year Study design

(study quality)

Sample N

(location)

Study population, mean age/age

range in years (follow-up time,

years)

Sleep assessment Cognitive/AD

measures

Impact of sleep problems/disorders

on AD

Benito-León et al.

201432

Prospective cohort

(high)

3,857

(Spain)

Community-dwelling elderly, 65

(median,12.5)

Daily total sleep duration

(self-reported)

Death

certificates,

ICD-9

Long sleepers (≥ 9h), aHR=1.63 [1.04-

2.56]* Short sleepers (≤ 5h), aHR=0.76

[0.33-1.74]

Blackwell et al.

200631

Cross sectional

(high)

2,242

(U.S.)

Community-dwelling women, 83.5

(N/A)

Poor sleep (wrist

actigraphy)

MMSE, Trails

B

aOR=1.48 [1.44–1.52]*ˠ

Blackwell et al.

201451

Prospective cohort

(high)

2822

(U.S.)

Community-dwelling men, 76, (3.4 ±

0.5 y)

Sleep quality (wrist

actigraphy, PSQI, ESS).

MMSE, Trails

B

aOR=1.44 [1.38, 1.51]*ˠ

Chang et al.

201333

Retrospective cohort

(high)

8484

(Taiwan)

Sleep apnea patients. 40+ (5) Sleep apnea (ICD-9) ICD-9 aHR=1.70 [1.26-2.31]*;

Females, aHR=2.38 [1.51–3.74]*;

Males, 50-59 years, aHR=6.08 [3.15-

11.70]*; 2.5 to 5 y of follow-up,

aHR=2.31 [2.07-2.58]*ˠ

Cohen-Zion et al.

200152

Prospective cohort

(Low)

46

(U.S.)

Community-dwelling, 80 (2) Wrist actigraphy MMSE EDS, aOR=2.26 [1.21-4.20]*;

SA, aOR=4.04 [2.40-6.68]*

Cricco et al.

200134

Prospective cohort

(high)

6,444

(Iceland)

Community-dwelling elderly, 72 (3) Insomnia (self-reported) SPMSQ Male non-depressed, aOR=1.33 [1.25-

1.42]*; Male depressed, aOR=1.89

[1.60-2.23]*; Female non-depressed,

aOR=1.07 [1.03-1.11]*;

Female depressed, aOR=1.23 [1.16-

1.31]*

Page 150: Omonigho A. Michael Bubu

138

Foley et al.

200153

Prospective cohort

(high)

2,346

(U.S.)

Community-dwelling Japanese-

American men, 76.6 (3)

Sleep disturbance (self-

reported)

CASI, DSM-

IIIR.

aOR=1.44 [1.34-1.54]*

Hahn et al.

201354

Prospective cohort

(high)

214

(U.S.)

Community-dwelling elderly, 83.4

(9)

Poor sleep (CPRS) DSM-IIIR

NINCDS-

ADRDA

aOR=1.78 [1.50-2.11]*ˠ

Jelicic et al.

200255

Prospective cohort

(medium)

838

(Netherlands)

Community-dwelling elderly, 63.3 (3) Poor sleep (self-reported) MMSE aOR=1.46 [1.38-1.54]*

Keage et al.

201239

Prospective cohort

(high)

2,012

(Australia,

U.K.)

Community-dwelling elderly, 64-94

(10)

EDS (self-report) MMSE aOR=2.09 [1.75-2.50]*ˠ

Lim et al.

2013a40

Prospective cohort

(medium)

698

(Canada)

Community-dwelling elderly, 83.3 (6) Sleep consolidation

(actigraphy)

NINCDS-

ADRDA

aHR=1.49 [1.39-1.60]*

Lim et al.

2013b36

Prospective cohort

(high)

737

(Canada)

Community-dwelling elderly, 81.6 (6) Sleep fragmentation

(actigraphy)

Cognitive

performance

aHR =1.22 [1.03-1.44]*;

Male, aHR=1.33 [0.90-1.96]*;

Female, aHR=1.29 [0.99-1.69]*

Merlino et al.

201044

Cross-sectional

(low)

750

(Italy)

Institutionalized and community-

dwelling elderly, 76.7 (NA)

Insomnia (self-report) DSM-IV-TR,

NINCS-

ARDRA,

NINDS-

AIREN

cOR=1.76 [1.02–3.11]*

Nebes et al.

200945

Cross-sectional

(medium)

157

(U.S.)

Community volunteers, 72.2 (NA) Sleep quality (PSQI) RBANS

TONI

aOR=2.21 [1.88-2.59]*

Page 151: Omonigho A. Michael Bubu

139

Ooms et al.

201461

Randomized controlled

trial (high)

26

(Netherlands)

Middle-aged men, 49.9 (1 night) Sleep deprivation (PSG) CSF Aβ42,

Aβ40, P-tau,

T-tau

aOR=5.06 [1.48-17.28]*

Oosterman et al.

200946

Cross-sectional

(medium)

144

(Netherlands)

Cardiology/internal medicine

outpatients, 69.5 (NA)

Sleep fragmentation

(actigraphy)

Cognitive

performance

aOR=1.84 [1.60-2.12]*

Osorio et al.

201156

Prospective cohort

(high)

346

(U.S.)

Brain aging study participants, 24-96

(7.7)

Insomnia (HAM-D) MMSE Insomnia, aOR=2.39 [1.03–5.55]*;

Insomnia w/o depression, aOR=3.32

[1.33–8.28]*

Osorio et al.

201437

Cross-sectional

(medium)

95

(U.S.)

Brain aging study participants, 67.7

(NA)

SDB (AHI ≥ 15). CSF P-tau, T-

tau, Aβ42

aOR=4.46 [2.35-8.48]*

Saint Martin et al.

201247

Cross-sectional

(medium)

272

(France)

Community-dwelling elderly, 74.8

(NA)

Sleep quality (PSQI)

MMSE,

GBSRT,

BVRT, Trail

A & B,

WAIS-III,

Stroop test,

AFCFT

aOR=1.65 [1.46-1.86]*

Schmidt et al.

201349

Case-control

(medium)

66

(Germany)

Consecutive AD patients and controls,

62.9 (NA)

Melanin-concentrating

hormone (MCH) and

hypocretin-1 (HCRT-1,

CERAD,

WMS-R, Trail

B, MRI, CSF

aOR=5.1 [1.98-13.12]*

Page 152: Omonigho A. Michael Bubu

140

orexin-A) (fluorescence

immunoassay (FIA))

Aβ42, Aβ40,

P-tau, T-tau

Spira et al.

201348

Cross-sectional

(medium)

70

(U.S.)

Normative aging study participants,

76.4 (NA)

WHIIRS β-amyloid Lower sleep quality, aOR=3.38 [2.05–

5.58]*; Insomnia, aOR=1.87 [1.19–

2.94]*

Sterniczuk et al.

201350

Retrospective cohort

(medium)

28,697

(Canada)

Community-dwelling middle-aged

and elderly, 64.6 (4.3)

Sleep disturbance Index

(SDI) (self-report)

AD (self-

reported),

aOR=1.18 [1.17-1.19]*

Tranah et al.

201157

Prospective cohort

(high)

1,282

(U.S.)

Community-dwelling women, 82.7

(4.9)

CAR (actigraphy). Modified

MMSE CVLT

IQCODE,

dementia

(self-reported)

aOR=1.66 [1.55-1.77]*

Tworoger et al.

200638

Prospective cohort

(medium)

1,844

(U.S.)

Community-dwelling women, 74.2 (2) Sleep duration (self-

reported)

Cognitive

performance

≤ 6 h sleep, aOR=2.35 [1.90-2.91]*;

Difficulty initiating sleep, aOR=2.21

[1.84-2.65]*;

≥ 8 h sleep, a0R=1.31 [0.76-2.29]

Virta et al.

201358

Prospective cohort

(high)

2,336

(Finland)

Elderly twins, 65+ (18-26) Poor sleep (self-reported) Linear

cognitive

score (self-

report)

≤ 7 h sleep, aOR=4.15 [3.12–5.54]*;

Hypnotic use, aOR=1.23 [0.73–2.08]

Page 153: Omonigho A. Michael Bubu

141

Walsh et al.

201459

Prospective cohort

(high)

287

(U.S.)

Community-dwelling women, 82.8 (5) CAR (actigraphy) Modified

MMSE

CVLT, Trails

B

aOR=3.43 [1.74–6.75]*

Yaffe et al.

201160

Prospective cohort

(high)

298

(U. S.)

Community-dwelling women, 82.3

(2.3)

SDB (AHI ≥ 15). DSM-IV Sleep-wake problems, aOR=1.09

[0.90–1.32]ˠ; SDB , aOR=1.87 [1.61–

2.18]*ˠ; Sleep

duration, aOR=0.56 [0.46-0.67]*ˠ

*: significant, ˠ: multiple outcome measure on same subject, so mean of the outcome computed. Abbreviations; a: adjusted, Aβ40/42: amyloid beta-40/42, AD: Alzheimer's disease, AFCFT: alphabetic fluency and category fluency

tasks, AHI ≥ 15: apnea hypopnea index of 15 or more events per hour of sleep, APOE: apolipoprotein epsilon4, BVRT: Benton visual retention test, c: crude, CAR: circadian activity rhythms, CASI: cognitive abilities screening

instrument, CERAD: consortium to establish a registry for Alzheimer’s disease, CPRS: comprehensive psychopathological rating scale, CSF: cerebrospinal fluid, CVLT: California verbal learning test , DSM-IIIR/IV-TR:

diagnostic and statistical manual of mental disorders, third edition/fourth edition, text revised, EDS: Excessive daytime sleepiness, ESS: Epworth sleepiness scale, GBSRT: Grober and Buschké selective reminding test, HAM-D:

Hamilton Depression Rating Scale , HR: hazard ratio, ICD-9/10: international classification of diseases ninth/tenth edition AD criteria, IQCODE: informant questionnaire on cognitive decline in the elderly, , MMSE: mini mental

state examination, MRI: magnetic resonance imaging, N: number of participants, NA: not applicable, N/A: not available, NINCDS-ADRDA: national institute of neurological and communicative disorders and stroke and the

Alzheimer's disease and related disorders association, NINDS-AIREN: national institute of neurological disorders and stroke and association internationale pour la recherché et l'enseignement en neurosciences vascular dementia

criteria, OR: odds ratio, PSG: polysomnography, PSQI: Pittsburgh sleep quality index, P-tau: phosphorylated tau, RBANS: repeatable battery for the assessment of neuropsychological status, RR: relative risk, SDB: sleep

disordered breathing, SPMSQ: short portable mental status questionnaire , TONI: test of nonverbal intelligence, Trails B: trail making b test, T-tau: total-tau, WAIS-III: Wechsler adult intelligence scale third version, WHIIRS:

women’s health initiative insomnia rating scale, WMS-R: Wechsler memory scale-revision

Page 154: Omonigho A. Michael Bubu

142

Table 2.2: Pooled relative risks from sub-group meta-analysis for the effect of sleep on Alzheimer's disease/cognitive decline

Subgroup N Relative risk (95% CI)

Sleep parameter

Sleep Efficiency 7 1.98 (1.11, 3.52)

Sleep Latency 4 1.33 (1.13, 1.57)

Wake After Sleep Onset 7 1.26 (1.19, 1.33)

Daytime vs. nighttime sleepiness

Daytime Sleepiness 7 1.49 (1.17, 1.90)

Nighttime problems 31 1.7 (1.48, 1.94)

Night & Daytime problems 6 1.45 (1.17, 1.78)

Gender

Female 7 1.56 (1.24, 1.96)

Male 13 1.49 (1.23, 1.79)

Both male & female 24 1.74 (1.49, 2.03)

Sleep duration

Long sleepers > 8 hrs/day 6 2.05 (1.24, 3.38)

Short sleepers ≤ 7 hrs/day 6 1.63 (1.13, 2.34)

Follow-up time

< 5 years 18 1.4 (1.23, 1.59)

≥ 5 years 18 1.76 1.46, 2.112

Classified sleep disorders

Insomnia 11 1.5 (1.25, 1.80)

Obstructive Sleep Apnea Syndrome 11 1.96 (1.59, 2.41)

Circadian-Rhythm Sleep Disorders 8 1.38 (1.09, 1.75)

Page 155: Omonigho A. Michael Bubu

143

Sleep Depth (Not classified) 18 1.62 (1.38, 1.91)

Continent

North America 25 1.53 (1.35, 1.74)

Europe 14 1.54 (1.27, 1.86)

Australia 1 2.09 (1.16, 3.78)

Asia 4 2.23 (1.60, 3.11)

Temporality

Prospective studies 36 1.57 (1.41, 1.77)

Non-prospective studies 8 1.70 (1.44, 2.02)

Study quality

High quality 31 1.58 (1.40, 1.79)

Medium quality 10 1.67 (1.32, 2.10)

Low quality 3 2.17 (1.38, 3.41)

Abbreviations N: Number of relative risk estimates, CI: confidence interval

Page 156: Omonigho A. Michael Bubu

144

Table 2.3: Results of meta-regression analysis examining potential effects of different factors on the natural logarithm of the odds ratio between sleep and Alzheimer's disease/cognitive decline

Meta-regression variables

Risk for Alzheimer's disease

Univariable analysis Multivariable analysis

Coef. p-Value Coef. p-Value

Proportion of females (in %) -0.0004 0.8647 0.0007 0.7224

Mean age (in years) -0.0022 0.7898 -0.0147 0.2287

Sample size (in number of people) -0.000 0.0413 -0.000 0.0175

Quality rating (in %) 0.1043 0.3046 0.0198 0.8721

Page 157: Omonigho A. Michael Bubu

145

Figure 2.1: Study retrieval and selection for effects of sleep on Alzheimer’s disease/cognitive decline meta-analysis.

Studies identified through database searching (n = 2341)

(PubMed=904, Embase=1026, Web of Science=305, Cochrane library=106)

Studies after duplicates removed (n = 1221)

Studies screened for titles (n = 1221)

Studies excluded (n = 180)

Full-text articles assessed for eligibility (n = 201)

Full-text articles excluded, with reasons (n = 130)

- Review article (n = 7) - No risk estimate (n = 17) - Sleep not assessed (n = 18) - Relationship not studied (n =

53) - Not peer reviewed (n = 10) - Alzheimer not examined (n

= 25)

Studies included in systematic review

(n = 72)

Studies included in meta-analysis (n = 27)

Article identified from reference search (n = 1)

Studies screened for abstracts

(n = 1041)

Studies excluded (n = 840)

Duplicates removed (n = 1120)

Studies excluded from analysis, with reasons (n = 45)

- Study design (n = 16) - Duplicate studies (n =

12) - Insufficient

information for a meta-analysis (n = 17)

Page 158: Omonigho A. Michael Bubu

146

Figure 2.2 Study retrieval and selection for effects of sleep on cognitive decline and/or Alzheimer’s disease meta-analysis

Abbreviations: EDS, excessive daytime sleepiness; RR, relative risk; SA, sleep apnea; SDB, sleep disordered breathing; TST, total sleep time; WASO, wake after sleep onset; w/D, with depression; w/o D, without depression; w/o, without

Page 159: Omonigho A. Michael Bubu

147

Figure 2.3: Forest plot presenting sub-group meta-analysis based on risk estimates for the effect of sleep on Cognitive decline, Pre-clinical and Symptomatic Alzheimer’s disease

Abbreviations: EDS, excessive daytime sleepiness; RR, relative risk; SA, sleep apnea; SDB, sleep disordered breathing; TST, total sleep time; WASO, wake after sleep onset; w/D, with depression; w/o D, without depression; w/o, without

Page 160: Omonigho A. Michael Bubu

148

Figure 2.4*: Trim and fill funnel plot for effects of sleep on Alzheimer’s disease/cognitive decline meta- analysis

*Filled dots represent published studies. Empty dots represent unpublished and/or imputed studies

Observed Outcome

Sta

nd

ard

Err

or

0.7

92

0.5

94

0.3

96

0.1

98

0.0

00

-1.00 0.00 1.00 2.00

Page 161: Omonigho A. Michael Bubu

149

Figure 2.5 Forest plot presenting sub-group meta-analysis based on risk estimates for the effect of different sleep problems and disorders on cognitive decline and/or Alzheimer’s disease

Abbreviations: EDS, excessive daytime sleepiness; RR, relative risk; SA, sleep apnea; SDB, sleep disordered breathing; TST, total sleep time; WASO, wake after sleep onset; w/D, with depression; w/o D, without depression; w/o, without

Page 162: Omonigho A. Michael Bubu

150

Figure 2.6: Forest plot presenting sub-group meta-analysis based on risk estimates for the effect of self-report versus actigraphic data of poor sleep on cognitive decline and/or Alzheimer’s disease

Abbreviations: EDS, excessive daytime sleepiness; RR, relative risk; SA, sleep apnea; SDB, sleep disordered breathing; TST, total sleep time; WASO, wake after sleep onset; w/D, with depression; w/o D, without depression; w/o, without

Page 163: Omonigho A. Michael Bubu

151

APPENDIX A

SUPPLEMENTAL TABLES AND FIGURES: SLEEP AND AD META-ANALYSIS

Table 1A: Main findings from included studies and comments regarding computed conversions of the different indices to a common index: odds ratios and regarding

computing a summary effect size of the impact of sleep problems on AD

Authors, year, reference

number, (country) Study Main Findings Comments for conversion

Impact of sleep problems/disorders on

Alzheimer's disease

Benito-León et al. 2014,32 (Spain)

Long sleepers (≥ 9h) aHR=1.63 [1.04-2.56] Short sleepers (≤ 5h) aHR=0.76 [0.33-1.74]

No conversions and/or mean computation done

Long sleepers (≥ 9h), aHR=1.63 [1.04-2.56]* Short sleepers (≤ 5h), aHR=0.76 [0.33-1.74]

Blackwell et al. 2006,31 (United States)

Sleep efficiency (MMSE) aOR=1.61 [1.20–2.16], Sleep efficiency (Trails B) aOR=1.96 [1.43–2.67], Sleep latency (MMSE) aOR=1.23 [1.13–1.33]; Sleep latency (Trails B) aOR=1.13 [1.04–1.24], WASO (MMSE) aOR=1.15 [1.06–1.23]; Sleep latency (Trails B) aOR=1.24 [1.15–1.34]. Napping (MMSE) aOR=1.42 [1.05–1.93]; Napping (Trails B) aOR=1.74 [1.26–2.40].

We accounted for multiple outcome measures on the same subjects by computing the mean of the effect sizes for each study and used the average effect size as the unit of analysis

aOR=1.44 [1.07–1.94]*ˠ

Blackwell et al. 2014,51 (United States)

Sleep efficiency (Trails B) aOR=1.53 [1.07, 2.18]; WASO aOR=1.47 [1.09, 1.98]; LWEP aOR=1.38 [1.02, 1.86]. 3MS:LWEP aOR=1.36 [1.09, 1.71]

We accounted for multiple exposure measures on the same subjects by computing the mean of the effect sizes for each study and used the average effect size as the unit of analysis

aOR=1.48 [1.17, 1.88]*ˠ;

Chang et al. 2013,33 Taiwan

SA aHR=1.70 [1.26-2.31] Females aHR=2.38 [1.51–3.74] Males 50-59 years aHR= 6.08 [1.96-18.90], Females ≥ 70 years aHR=3.20 [1.71–6.00]. First 2.5 years of follow-up aHR: =2.04 [1.35-3.07]. 5 years of follow-up aHR=1.70 [1.26-2.31]

No conversions and/or mean computation done for separate populations and measures. Follow-up time measures were on the same subjects as such we computed the mean of the effect sizes and used the average effect size as the unit of analysis

aHR=1.70 [1.25-2.31]*; Females, aHR=2.38 [1.51–3.74]*; Males, 50-59 years, aHR=6.08 [1.96-18.90]*; 2.5 to 5 y of follow-up, aHR=2.31 [1.45-3.69]*ˠ

Page 164: Omonigho A. Michael Bubu

152

Cohen-Zion et al. 2001,52 United States

Increased RDI (t=-2.17, 42 df, r=-0.32, P=.036) Increased EDS (t=-3.32, 42 df, r=-0.46, P=.002) EDS (t39 = -2.45, r=-0.37, P= .019)-Adjusted model RDI (t39=- 0.66, r=-0.11, P=.515)-Adjusted model

Converted from r to d (d = d2r/√(1-r^2 ) ) and from d to the log(OR) (Log(OR) = dp/√3 and mean computation done for separate populations

SA, aOR=2.26 [0.75-6.80]; EDS, aOR=4.04 [1.48-11.06]*

Cricco et al. 2001,34 Iceland

Non-depressed men chronic insomnia aOR=1.49 [1.03–2.14] Non-depressed men incident insomnia aOR=1.16 [0.82–1.65] Depressed men incident insomnia aOR=1.60 [0.87-2.95] Depressed men chronic insomnia aOR=2.18 [1.30-3.67] Non-depressed women chronic insomnia aOR=1.18 [0.92–1.52] Non-depressed women incident insomnia aOR=0.95 [0.71–1.26] Depressed women incident insomnia aOR=1.10 [0.75-1.61] Depressed women chronic insomnia aOR=1.36 [1.01-1.84]

We accounted for multiple exposure measures on the same subjects by computing the mean of the effect sizes for each study and used the average effect size as the unit of analysis for each population

Male non-depressed, aOR=1.33 [0.93-1.90]ˠ; Male depressed, aOR=1.89 [1.07-3.34]*ˠ; Female non-depressed, aOR=1.07 [1.03-1.11]*ˠ; Female depressed, aOR=1.23 [0.87-1.73]ˠ;

Foley et al. 2001,53 United States

Excessive daytime sleepiness DSM-IIIR aOR=2.19 [1.37–3.50] Excessive daytime sleepiness CASI aOR=1.44 [1.01–2.08]. Insomnia DSM-IIIR aOR=0.99 [0.70-1.41] Insomnia CASI aOR=1.14 [0.91-1.43]

We accounted for multiple exposure measures on the same subjects by computing the mean of the effect sizes for each exposure category and used the average effect size as the unit of analysis

EDS: aOR=1.82 [1.19-2.79]* Insomnia: aOR=1.07 [0.81-1.42]

Hahn et al. 2013,54 United States

Reduced sleep (Dementia)a aHR=1.75 [1.04-2.93] Reduced sleep (AD)a aHR=2.01 [1.12-3.61] Reduced sleep (Dementia)b aHR=1.50 [0.87-2.59] Reduced sleep (AD)b aHR=1.69 [0.91-3.14] Reduced sleep (Dementia)c aHR=1.73 [1.02-2.91] Reduced sleep (AD)c aHR=2.02 [1.11-3.65] Reduced sleep (Dementia)d aHR=1.68 [0.98-2.86] Reduced sleep (AD)d aHR=1.86 [1.00-3.70]

We accounted for multiple outcome measures on the same subjects by computing the mean of the effect sizes for each study and used the average effect size as the unit of analysis

aOR=1.78 [0.86-3.70]ˠ

Jelicic et al. 2002,55 The Netherlands

Overall sleep problems continuous scores β=-0.05 (-0.09,-0.00) Overall sleep problems dichotomized scores β=-0.38 (-0.71,-0.05)

Converted from β to OR aOR=1.46 [1.05-2.03]*

Page 165: Omonigho A. Michael Bubu

153

Ju et al. 2013, United States

Sleep efficiency OR=5.60 [0.97- 32.5] No conversions and/or mean computation done

OR=5.60 [0.97- 32.5]

Keage et al. 2012,39 Australia, & United Kingdom

Napping (2years) aOR=0.38 [0.22–0.67]. Napping (10years) aOR=0.48 [0.29–0.78]. Sleep duration (<= 6.5 h 10years) aOR=2.02 [1.17–3.48] Sleep duration (>= 9 h 10years) aOR=1.27 [0.64-2.48] Daytime sleepiness (10 years) aOR=2.43 [1.24–4.75].

We accounted for multiple exposure measures on the same subjects by computing the mean of the effect sizes for each exposure category and used the average effect size as the unit of analysis

Sleep Quality: aOR=1.10 [0.58-2.07]ˠ <= 6.5h sleep: aOR=2.02 [1.17-3.48]*ˠ; >= 9h sleep: aOR=1.27 [0.65-2.48]ˠ;

Lim et al. 2013a,40 Canada Better Sleep consolidation aHR 0.67 [0.46-0.97] We converted the risk estimate for better sleep consolidation to a risk estimate for poor sleep consolidation

aHR=1.49 [1.02-2.17]*

Lim et al. 2013b,36 Canada Sleep fragmentation aHR =1.22 [1.03-1.44] Male aHR=1.33 [0.90-1.96] Female aHR=1.29 [0.99-1.69]

No conversions and/or mean computation done

aHR =1.22 [1.03-1.44]*; Male, aHR=1.33 [0.90-1.96]; Female, aHR=1.29 [0.98-1.69]

Merlino et al. 2010,44 Italy Excessive daytime sleepiness OR=1.76 (1.02–3.11) No conversions and/or mean computation done

cOR=1.76 [1.02–3.11]*

Nebes et al. 2009,45 United States

Sleep latency correlated the RBANS (r = −.241, p = .002) Sleep latency correlated the TONI (r = −.258, p = .001). Sleep efficiency correlated with the RBANS (r = .185, p = .020), Sleep efficiency correlated with the TONI (r = .197, p = .013), Sleep efficiency correlated with the N-Back (r = .211, p = .008).

Converted from r to d (d = d2r/√(1-r^2 ) ) and from d to the log(OR) (Log(OR) = dp/√3 and computed the mean of the effect sizes for each study and used the average effect size as the unit of analysis

aOR=2.21 [1.26-3.87]*

Oosterman et al. 2009,46 Netherlands

Fragmentation of the sleep-wake rhythm (Mental: r =-0.16, Memory: - 0.19, andExecutive: - 0.16 respectively)

Converted from r to d (d = d2r/√(1-r^2 ) ) and from d to the log(OR) (Log(OR) = dp/√3 and computed the mean of the effect sizes for each study and used the average effect size as the unit of analysis

aOR=1.84 [1.09-3.11]*

Osorio et al. 2011,56 United States

Insomnia aOR=2.39 [1.03–5.55]. Insomnia without depression aOR=3.32 [1.33–8.28].

No conversions and/or mean computation done

Insomnia, aOR=2.39 [1.03–5.55]*; Insomnia w/o depression, aOR=3.32 [1.33–8.28]*

Page 166: Omonigho A. Michael Bubu

154

Osorio et al. 2014,37 United States

In ApoE3+ subjects, significant differences were found between sleep groups for p-tau (F[df2] = 4.3, p =0.017), and t-tau (F[df2] = 3.3, p = 0.043). Additionally, among ApoE3+ subjects, the apnea and/or hypopnea with 4% O2-desaturation index was positively correlated with p-tau (r = 0.30, p = 0.023), t-tau (r = 0.31, p = 0.021), and Aβ-42 (r = 0.31, p = 0.021). In ApoE2+ subjects, the apnea and/or hypopnea with 4% O2-desaturation index was correlated with lower levels of CSF Aβ-42 (r = −0.71, p = 0.004), similarly to ApoE4+ subjects where there was also a trend toward lower CSF Aβ-42 levels.

Converted from r to d (d = d2r/√(1-r^2 ) ) and from d to the log(OR) (Log(OR) = dp/√3 and computed the mean of the effect sizes for each study population and used the average effect size as the unit of analysis

ApoE3+ subjects: aOR=3.20 [1.18-8.66]* ApoE2+ subjects: aOR= 8.17 [1.96-34.07]*

Saint Martin et al. 2012,47 France

PSQI correlated with the MMSE (r = -0.14, p = .03) Sleep Quality correlated with Delayed free recall (r = -0.14, p < .05)

Converted from r to d (d = d2r/√(1-r^2 ) ) and from d to the log(OR) (Log(OR) = dp/√3 and computed the mean of the effect sizes for each study and used the average effect size as the unit of analysis

aOR=1.65 [1.02-2.68]*

Schmidt et al. 2013,49 Germany

A significant main effect of diagnosis (F(1,62) = 8.490, p<0.01) on MCH levels was found between AD (93.76 ± 13.47 pg/mL) and HS (84.65 ± 11.40 pg/mL). MCH correlated with T-tau (r = 0.47; p<0.01) and P-tau (r = 0.404; p,0.05) in the AD but not in the HS. CSF-MCH correlated negatively with MMSE scores in the AD (r = -0.362, p<0.05) and was increased in more severely affected patients (MMSE ≤ 20) compared to HS (p<0.001) and Behavioral and Psychological Symptoms of Dementia (BPSD) positive patients compared to HS (p<0.05). In CSF-HCRT- 1, a significant main effect of sex (F(1,31) = 4.400, p<0.05) with elevated levels in females (90.93 ± 17.37 pg/mL vs. 82.73 ± 15.39 pg/mL) was found whereas diagnosis and the sex*diagnosis interaction were not significant.

Converted from r to d (d = d2r/√(1-r^2 ) ) and from d to the log(OR) (Log(OR) = dp/√3 and computed the mean of the effect sizes for each study and used the average effect size as the unit of analysis

aOR=5.1 [1.31-19.89]*

Spira et al. 2013,48 United States

Shorter sleep duration Mean cortical DVR (B = 0.08 [95% CI, 0.03-0.14]; r =.38; P = .005) Mean precuneus DVR (B = 0.11 [0.03-0.18]; r =.36; P = .007). Lower sleep quality Mean cortical DVR (B = 0.04 [95% CI, -0.01-0.09]; r =.19; P = .13) Mean Precuneus DVR (B = 0.08 [0.01-0.15]; r=.29; P = .03). WHIIRS, Women’s Health Initiative Insomnia Rating Scale Mean cortical DVR (B = 0.01 [95% CI, -0.004-0.02]; r =.16; P=.23) Mean Precuneus DVR(B = 0.01 [95% CI, -0.004-0.02] r=.18; P=.16).

Converted from β to OR Converted from r to d (d = d2r/√(1-r^2 ) ) and from d to the log(OR) (Log(OR) = dp/√3 and accounted for multiple exposure measures on the same subjects by computing the mean of the effect sizes for each study and used the average effect size as the unit of analysis

Sleep duration, aOR= 4.25 [1.52-11.88]*ˠ; Lower sleep quality, aOR=2.51 [0.96–6.56]ˠ; Insomnia, aOR=1.87 [0.73–4.80]ˠ

Page 167: Omonigho A. Michael Bubu

155

Sterniczuk et al. 2013,50 Canada

SDI aOR=1.23 [1.11-1.36] SDI aOR=1.12 [1.00-1.25]

We accounted for multiple outcome measures on the same subjects by computing the mean of the effect sizes for each study and used the average effect size as the unit of analysis

aOR=1.18 [1.06-1.31]*ˠ;

Tranah et al. 2011,57 United States

Decreased activity rhythms aOR=1.57 [1.09–2.25] Rhythm robustness aOR=1.57 [1.10–2.26] Peak Activity timing aOR=1.83 [1.29–2.61]

We accounted for multiple outcome measures on the same subjects by computing the mean of the effect sizes for each study and used the average effect size as the unit of analysis

aOR=1.66 [1.16-1.31]*ˠ;

Tworoger et al 2006,38 United States

<=5hrs of sleep aOR=2.19 (1.10-4.39) 6hrs of sleep aOR=1.01 (0.61-1.67) 8hrs of sleep aOR=1.16 (0.74-1.82) >=9hrs of sleep aOR=1.46 (0.77-2.76) TICS aOR=2.51 (1.38, 4.54) Women who regularly had difficulty falling or staying asleep TICS (aOR: 1.85, 95% CI: 1.04, 3.30) global score (aOR: 2.57, 95% CI: 1.39, 4.74) TICS (Telephone Interview of Cognitive Status)

We accounted for multiple exposure measures on the same subjects by computing the mean of the effect sizes for each exposure category and used the average effect size as the unit of analysis

≤ 6 h sleep, aOR=1.68 [0.92-3.08]ˠ; Difficulty initiating sleep, aOR=2.21 [1.22-4.01]*ˠ; ≥ 8 h sleep, a0R=1.30 [0.71-2.39]ˠ;

Virta et al 2013,58 Finland,

Sleep Length (N=2328) Sleep Length (< 7 h/day) (β = -0.84 (-1.51,-0.17), P = 0.014 Sleep Length (< 7 h/day) (β = -0.79 (-1.44,-0.14), P = 0.019 and Sleep Length (> 8 h/day) (β = -1.66 (-2.37,-0.95) P < 0.001 Sleep Length (> 8 h/day) (β = -1.61 (-2.33,-0.91) P < 0.001. Poor sleep quality (β = -1.00 (-1.77,-0.23) P = 0.011. Poor sleep quality (β = -0.89 (-1.69,-0.09) P = 0.028 The use of hypnotics ≥ 60 days per year (β = -1.92 (-3.11,-0.73), P = 0.002). The use of hypnotics ≥ 60 days per year (β = -1.58 (-2.75,-0.41), P = 0.008). Model 1 adjusted for age, sex, education, ApoE status, and follow-up. Model 2 adjusted for life satisfaction, obesity, hypertension, physical inactivity, heavy drinking, and binge drinking in addition to the factors adjusted for in Model 1

We accounted for multiple exposure measures on the same subjects by computing the mean of the effect sizes for each exposure category and used the average effect size as the unit of analysis

≤ 7 h sleep, aOR=2.26 [1.17–4.38]*ˠ; >=8 h sleep, aOR=5.13 [2.51-10.49]*ˠ; Sleep quality, aOR=3.72 [2.45-5.65]*ˠ; Hypnotic use, aOR=5.85 [1.80–19.03]*ˠ

Page 168: Omonigho A. Michael Bubu

156

Walsh et al 2014,59 United States

Women lowest vs. highest quartile CAR amplitude (group difference (d) = 30.42 sec, d = -1.01 words respectively, P < 0.05). Categorical fluency mesor (d = -0.86 words, P < 0.05). Women Categorical fluency later acrophase categorical fluency (d = -0.69 words, P < 0.05). Controlling for baseline Mini-Mental State Examination and sleep factors had negligible effects on the results.

Converted from d to the log(OR) aOR=3.43 [1.08–10.85]*

Yaffe et al. 2011,60 United States

SDB (Women) aOR=1.85 [1.11-3.08]. AHI (≥15 events/hour) aOR=1.71 [1.04 − 2.83] >7% of sleep time in apnea/hypopnea aOR=2.04 [1.10 − 3.78] respectively). Sleep Fragmentation Mild aOR=0.54 [0.29-0.98] Sleep Fragmentation High aOR=0.58 [0.32-1.07] WASO Mild aOR=1.17 [0.63-2.19] WASO High aOR=1.79 [0.97-3.29] Sleep Duration Mild aOR=0.58 [0.31-1.09] Sleep Duration High aOR=0.83 [0.46-1.51]

We accounted for multiple exposure measures on the same subjects by computing the mean of the effect sizes for each exposure category and used the average effect size as the unit of analysis

Sleep Fragmentation, aOR=0.56 [0.31-1.02]ˠ WASO, aOR=1.48 [0.80–2.74]ˠ; SDB , aOR=1.87 [1.08–23.23]*ˠ; TST, aOR=0.71 [0.37-1.35]ˠ

*: significant, ˠ: multiple exposure/outcome measure on same subject, so mean of the outcome computed. Abbreviations; a: adjusted, AD: Alzheimer's disease, APOE: apolipoprotein epsilon4, c: crude, CAR: circadian activity rhythms, CASI: cognitive abilities screening instrument, MMSE: mini mental state examination, OR: odds ratio, PSG: polysomnography, PSQI: Pittsburgh sleep quality index, RR: relative risk, SDB: sleep disordered breathing, Trails B: trail making b test, TST: total sleep time, WASO: wake after sleep onset,

Page 169: Omonigho A. Michael Bubu

157

Table 2A: Formula used for converting among effect sizes and for calculating the variance of the

effect sizes

Converting Among Effect

Sizes*

Formula Variance Formula**

Converting from d to the log odds ratio

Log(OR) = dπ

√� , where π is

the mathematical constant (approximately 3.14159)

The variance of Log(OR) would then be

��(��) = �π

�3

Converting from r to d d = d��

√���� The variance of d computed in this way (converted from r) is

� = 4�(1 − �� )�

Converting from d to r r = �

√���� , where a is a

correction factor for cases where n1 ≠ n2,

a = (��� ��)�

����

The correction factor (a) depends on the ratio of n1 to n2, rather than the absolute values of these numbers. Therefore, if n1 and n2 are not known precisely, use n1= n2, which will yield a=4.

The variance of r computed in this way (converted from d) is

� = �� �(�� + �)�

Converting from the log odds ratio to d

d = Log(OR)* √�π

� = ��(��) ∗ √�π

converting from β to OR exp(β) N/A

*Effect sizes from the reviewed studies included odds ratios (OR), hazard ratios (HR), Pearson’s correlation coefficient (r), beta estimates (β) and standardized mean differences (d). To proceed with the meta-analysis, we converted the different indices to a common index: odds ratios (meta-analyses were computed using Log(OR) but were converted back to OR for presentation). ** Reference #30 in manuscript: Borenstein Michael HLV, Higgins J. P. T., Rothstein H.R. Introduction to Meta-Analysis. : John Wiley & Sons, Ltd.; © 2009

Page 170: Omonigho A. Michael Bubu

158

Table 3A: Formula used for computing the variance of a composite or a difference

Computing the variance of a composite or

a difference*

Formula**

The variance of the sum of two correlated variables

If we know that the variance of Y1 is V1 and the variance of Y2 is V2, then Var (!� + !�) = � + � + 2�#�#� Where r is the correlation coefficient that describes the extent to which Y1 and Y2 co-vary. If Y1 and Y2 are inextricably linked (so that a change in one determines completely the change in the other), then r =1, and the variance of the sum is roughly twice the sum of the variances. At the other extreme, if Y1 and Y2 are unrelated, then r=0 and the variance is just the sum of the individual variances.

The variance of the mean of two correlated variables

Var $�� (!� + !�)% = &�

�'� Var (!� + !�) =

�( )� + � + 2�#�#� *

The variance of the mean of several correlated variables

var& �+ ∑ !-+-.� '= & �

+'� var)∑ !-+-.� �*=

& �+'� /∑ -�

+-.� + ∑ /�-0 1- #02- 30 2

The variance of the difference between two correlated variables

If we know that the variance of Y1 is V1 and the variance of Y2 is V2, then Var (!� − !�) = � + � − 2�#�#�

*Since our aim was to compute a summary effect size of the impact of sleep problems on cognitive decline and Alzheimer’s disease, we accounted for multiple exposure or outcome measures on the same subjects by computing the mean of the effect sizes for each study and used the average effect size as the unit of analysis. This influenced the calculation of the variance of the sum and mean effects of the correlated variables. For our analysis, we worked with a plausible range of correlations ranging from 0.5-1.0 and performed sensitivity analyses to find the most appropriate standard error range. ** Reference #30 in manuscript: Borenstein Michael HLV, Higgins J. P. T., Rothstein H.R. Introduction to Meta-Analysis. : John Wiley & Sons, Ltd.; © 2009

Page 171: Omonigho A. Michael Bubu

159

Figure 1A Forest plot presenting sub-group meta-analysis based on continent in which study was published for the effect of sleep problems and disorders on cognitive decline and/or Alzheimer’s disease

Page 172: Omonigho A. Michael Bubu

160

Figure 2A: Forest plot presenting sub-group meta-analysis based on day versus night time sleepiness for the effect of sleep problems and disorders on cognitive decline and/or Alzheimer’s disease

Page 173: Omonigho A. Michael Bubu

161

Figure 3A Forest plot presenting sub-group meta-analysis based on study temporality for the effect of sleep problems and disorders on cognitive decline and/or Alzheimer Disease

Page 174: Omonigho A. Michael Bubu

162

Figure 4A Forest plot presenting meta-analysis risk estimates based on omitting one study for the effect of sleep problems and disorders on cognitive decline and/or Alzheimer Disease

Page 175: Omonigho A. Michael Bubu

163

SECTION 3

OBSTRUCTIVE SLEEP APNEA IS ASSOCIATED WITH LONGITUDINAL INCREASES IN BRAIN

FLORBETAPIR PET IMAGING, CSF TAU, PTAU, AND DECREASE IN CSF AΒ42 BURDEN, IN

ELDERLY COGNITIVE NORMAL AND MCI INDIVIDUALS.

KEY POINTS

Question: Does Obstructive Sleep Apnea (OSA) affect longitudinal changes in brain amyloid deposition

and cerebrospinal fluid (CSF) Aβ42, t-tau and p-tau181 in cognitive normal (NL), mild cognitive

impairment (MCI) and Alzheimer’s Disease (AD) older adults.

Findings: OSA accelerates increases in brain amyloid deposition, CSF TAU and PTAU and decreases in

CSF biomarkers burden, over time, both in elderly NL and MCI individuals.

Meaning: Clinical interventions aimed at OSA, such as treatment with CPAP or dental appliances, in NL

and MCI patients, could possibly slow the progression of cognitive impairment to AD.

ABSTRACT

Importance: Recent studies demonstrate that OSA is associated with AD biomarkers. To evaluate

evidence for a causal association between OSA and AD, demonstrating AD-specific neuropathology

changes resulting from increased disease burden in OSA patients is vital.

Objective: To determine the effect of OSA on longitudinal changes in brain amyloid deposition and CSF

Aβ42, t-tau and p-tau181 in NL, MCI and AD older adults.

Design: Longitudinal study. Mean follow up time was 2.52 ±0.51 years.

Page 176: Omonigho A. Michael Bubu

164

Setting: Data used for this study were obtained from the Alzheimer’s disease Neuroimaging Initiative

(ADNI) database (adni.loni.usc.edu) on December 7, 2016.

Participants: 516 NL, 798 MCI and 325 AD subjects.

Exposure: Participant self-reported physician diagnosis of OSA.

Main Outcomes and Measures: Rate of change in brain Aβ42, CSF TAU, PTAU, and CSF Aβ42

burden over time. Multi-level mixed effects linear regression models with randomly varying intercepts

and slopes were constructed to test whether the rate of change in biomarker data differed between subjects

with and without OSA. The final models were adjusted for age, sex, BMI, CPAP use, APOE e4 status,

and history of chronic medical conditions.

Results: The self-reported prevalence of OSA in the 3 study cohorts was as follows: NL (6%), MCI

(13%), AD (7%). Across all groups, mean ages of OSA+ and OSA- were 72.3 ±7.1 and 73.9 ±7.3

respectively. Females were 49% in the NL group, 40% in the MCI group, and 37% in the AD group. In

NL and MCI groups, the annual rate of AD biomarker burden change over time, indicated that OSA+

subjects experienced faster increase in brain Aβ-42 (B = - .06, 95% CI, -.09, -.04 for both), CSF TAU (B

= -2.89, 95% CI, -3.51 to -2.29 and B = -1.89, 95% CI, -2.91, -.87, respectively), CSF PTAU (B = -1.21,

95% CI, -1.71, -.74 and, B = -1.48, 95% CI, -2.05, -.94, respectively), and a faster decrease in CSF Aβ-42

(B = 3.93, 95% CI, 3.56, 4.31 and B = 2.69, 95% CI, 2.02, 3.36, respectively); compared to OSA-

subjects, over the follow-up period.

Conclusion and Relevance: OSA appears to accelerate increases in brain amyloid deposition, CSF TAU

and PTAU and decreases in CSF biomarkers burden, over time, both in elderly Cognitive Normal and

MCI individuals. Clinical interventions aimed at OSA, such as treatment with CPAP or dental appliances,

in cognitive normal and MCI patients, could possibly slow the progression of cognitive impairment to AD

Page 177: Omonigho A. Michael Bubu

165

INTRODUCTION

Obstructive Sleep Apnea (OSA) and Alzheimer’s disease (AD) are both common chronic disease

conditions in older adults. OSA affects between 19% and 57% of individuals aged over 65.1-4 By 2050,

the incidence of AD in the U.S is expected to approximate a million people each year, with a total

estimated prevalence of 11 to 16 million people.5 Both OSA and AD cause significant morbidity and

mortality to those afflicted,6-8 and have very high socio-economic burden worldwide.5,8-16 It is therefore

pertinent that preventive and/or treatment efforts targeting both disease conditions are consolidated to

create a better quality of life in older adults.

OSA is characterized by intermittent partial or complete breathing cessation during sleep,

occurring from narrowing of various upper airway sites.1 OSA related hypoxemia and sleep fragmentation

have been implicated as possible links between OSA and AD, with recent studies demonstrating

associations between OSA and AD biomarkers in cognitive normal (NL) and MCI older adults.17-21

Clearly distinguishing whether OSA individuals with normal cognition or MCI are at heightened risk to

develop AD is critical to our ultimate mission of preventing AD.

AD biomarkers serve as in-vivo indicators of particular AD pathologies.22-24 They can be used in

the clinical setting to predict future clinical course of AD. 25,26 PET amyloid imaging of significant Aβ

load and significant levels of cerebrospinal fluid (CSF) Aβ42 are robust predictors of the development of

AD in at risk populations’ i.e. cognitive normal to amnestic mild cognitive impairment (aMCI) and MCI

to AD.25,26 Aβ deposition commences many years prior to AD symptoms’ onset27,28 while MRI brain

imaging show abnormalities later in the disease trajectory.29 This suggests a time-dependent risk of

developing AD from amnestic MCI. Significant increases of CSF P-tau have been demonstrated in AD

patients compared with controls.30 P-tau in CSF is a measurable core marker of AD.31 Since cross-

sectional studies demonstrate that OSA is associated with AD biomarkers, we further examined OSA’s

effect on longitudinal changes of these biomarkers, especially since they have been shown to predict time-

to-progression from MCI to AD.32 Demonstrating associations between OSA and AD-specific

Page 178: Omonigho A. Michael Bubu

166

neuropathology changes, progression or worsening consolidates the evidence for a potential causal

relationship between OSA and AD.

METHODS

Data used in the preparation of this article were obtained from the Alzheimer’s Disease

Neuroimaging Initiative (ADNI) database (adni.loni.usc.edu). The ADNI was launched in 2003 as a

public-private partnership, led by Principal Investigator Michael W. Weiner, MD. The primary goal of

ADNI has been to test whether serial magnetic resonance imaging (MRI), positron emission tomography

(PET), other biological markers, and clinical and neuropsychological assessment can be combined to

measure the progression of mild cognitive impairment (MCI) and early Alzheimer’s disease (AD). Thus

far, ADNI has recruited over 2,000 adults aged 55 to 90 years, consisting of cognitively normal (NL)

older individuals, people with early or late MCI, and people with early AD-dementia. Follow-up is at 6

month intervals with duration ranging from 2 to 3 years as specified in the protocols for ADNI-1, ADNI-

2, and ADNI-GO.

Study Participants

Participant data used for this study were based on medical history from baseline and follow-up

visits obtained from downloaded ADNI data on December 7, 2016. Study participants included 1,639

subjects (516 Cognitive Normal (NL), 798 MCI and 325 AD). Subjects missing important study covariates

were excluded in each of the study groups. Subjects having co-morbid sleep disorders, body mass index

(BMI) change greater than 5 between any follow-up visits, were further excluded, since disturbed sleep,

and BMI are known independent risk factor for cognitive decline and AD, and being overweight is

associated with SDB and AD.33 Other exclusions included subjects who indicated they have had previous

OSA surgery, and those who had a reversible diagnosis (i.e. they had an MCI or AD diagnosis at any time

point but an NL diagnosis or MCI diagnosis at their last visit, thereby allowing us to control for unspecified

diagnoses.

Page 179: Omonigho A. Michael Bubu

167

OSA diagnosis

Presence or absence of OSA was self-reported (variable name: MHDESC). Patients with reported

“sleep apnea” or “OSA” were labeled OSA+ and the remaining participants were considered OSA−. To

ensure that patients were allocated into the correct groups, three physicians (R.O, S.A and O.B.) reviewed

medical history descriptions in the ADNI download, for OSA+ and OSA-.

NL, MCI and AD Diagnosis

ADNI criteria for subject classification is described elsewhere.34 In summary, NL subjects had no

memory complaints while MCI and AD subjects had memory complaints. NL and MCI scored between

24 – 30 on the Mini-Mental State Examination (MMSE) while AD subjects scored between 20 – 26. NL,

MCI and AD subjects had a Clinical Dementia Rating (CDR) score of zero, 0.5 with a mandatory

requirement of the memory box score being 0.5 or greater, and 0.5 or 1, respectively. In addition, MCI

subjects had to have largely intact general cognition and functional performance, and could not qualify for

dementia diagnosis.35 The diagnosis of AD was made using established clinical criteria.36 Patient

diagnosis was recorded at 6-month intervals for 24 months. Subjects were classified as converters if they

converted to AD between 12 months and 24 months and as stable if they did not convert by 24 months.

Florbetapir-PET Imaging Acquisition and Interpretation

ADNI Florbetapir summary data are updated regularly, and uploaded to LONI by the University

of California at Berkeley group.37,38 More information on the methods can be found at:

https://adni.loni.usc.edu/wp-content/uploads/2010/05/ADNI2_PET_Tech_Manual_0142011.pdf,

http://adni.loni.usc.edu/updated-florbetapir-av-45-pet-analysis-results/.

Cerebrospinal Fluid Methods (CSF)

CSF bio-specimen data collection are explained in detail at http://adni.loni.usc.edu/data-

samples/biospecimen-data/ and (http://www.adni-info.org/index.php).

Page 180: Omonigho A. Michael Bubu

168

Covariates/potential confounders

Covariates/potential confounders in our study include age, sex, educational status, smoking

status, body mass index (BMI), APOE4 status, hypertension, diabetes and history of cardiovascular

disease.

Data analysis

Given that ADNI data is unbalanced with unequal numbers of measurement for each study

participant, we used multi-level mixed effects linear regression models with normal errors39-41 for our

longitudinal analyses to examine the relationship between OSA and CSF P-tau, and Aβ-42 volumes, as

they provide a flexible and valuable tool for analyzing such unbalanced longitudinal data. Our method

allowed for incorporation of all the available information in the data, and possibly reduced or even

eliminated any bias resulting from an analysis confined to the complete cases.42

Specifically, to determine the specific type of modeling, we examined profile plots for the

biomarker data levels over time, by OSA status (=1 if OSA, =0 if non-OSA) using both the original data

and within-subject residuals. We also examined trajectory plots obtained by subtracting the baseline

measurement from the original measurements (i.e. dij = Yij − Yi1), and examined plots of mean and

variance of the biomarker data at each time point by OSA status. Further, using the slopes as summary

statistics, we conducted different formal tests to compare the two OSA statuses. Based on the above, our

assumption was that the rate of change of biomarker data is approximately linear over time, so we

proposed a parametric model with time as a continuous variable. Using PROC MIXED in SAS software;

we fit the models with randomly varying intercepts and slopes and allowed them to depend on exposure

group (i.e., OSA status). Based on the findings from above, we fit a random coefficients model for the

relationship between OSA status and time using unstructured covariance model. For finding a

parsimonious covariance structure, we fit as many different covariance models and conducted a likelihood

ratio test of each nested model using information criteria for non-nested models. Finally, using the

preferred covariance structure obtained above, we used PROC MIXED to construct a test of whether the

Page 181: Omonigho A. Michael Bubu

169

rate of change in biomarker data differed between the two OSA groups. The final models were adjusted

for age, sex, BMI, CPAP use, APOE e4 status, and history of chronic medical conditions.

These analyses allowed us to examine whether there was significant variation between OSA and

non-OSA subjects in mean AD biomarker level at baseline (intercept), as well as whether significant

variation in the change in AD biomarker level over time (slope) occurred. Furthermore, the covariance

between the baseline AD biomarker level (intercept) and AD biomarker change over time indicated

whether OSA+ or OSA- subjects had experienced a faster increase or decrease in AD biomarker level

over time (significant slope). It also allowed for assessment of significant differences in the rate of change

in AD biomarker level between OSA+ and OSA- patient groups over time. Statistical analyses was

performed using SAS software (version 9.4; SAS Institute Inc., Cary, NC). For sensitivity analyses, we

attempted using the dichotomized brain Aβ level outcome (Aβ+ vs. Aβ-) and examined the rate of change

in AD biomarker level in these populations (SDB+/ Aβ+ vs. SDB-/ Aβ+; SDB+/ Aβ- vs. SDB-/ Aβ-; and

SDB+/ Aβ+ vs. SDB+/ Aβ-; SDB-/ Aβ - vs. SDB-/ Aβ+). However, power issues limited our analyses;

moreover, convergence issues did not permit any estimation with these categorical variables.

RESULTS

Demographic and Clinical Characteristics

Table 3.1 shows the demographic and clinical characteristics of study participants. Overall, the

mean ages of OSA+ and OSA- were 72.3 ±7.1 and 73.9 ±7.3 respectively. Forty-nine percent were female

in the NL group, while in the MCI and AD groups, females consisted of 40% and 37% of the participants

respectively. In the NL group, 6% were OSA+, while 13% of the MCI group and 7% of the AD group

were OSA+. Participants differed markedly in APOE4 status with 28%, 51% and 66% being APOE

positive in the NL, MCI and AD groups respectively. Participants were similar in age, educational status,

BMI, and chronic medical conditions across groups (i.e. NL, MCI and AD), however, within group

Page 182: Omonigho A. Michael Bubu

170

differences (i.e. OSA+ vs. OSA-) existed on these characteristics. Mean follow up time for all participants

was 2.52 ±0.51 years.

Baseline AD Biomarker levels by clinical group and OSA status

Table 3.1 and Figure 3.1 show baseline CSF Aβ-42, TAU, PTAU and Brain Florbetapir PET

imaging levels by NL, MCI and AD group and OSA status without controlling for confounders. There

were significant differences in CSF Aβ-42 levels for both MCI (P =0.04), and AD (P <0.01) groups

respectively, with OSA+ participants having significantly higher levels at baseline. No significant

difference was seen in NL group by OSA status. Significant difference in TAU levels was seen for the

MCI patients (P =0.02), with OSA+ individuals having significantly lower levels. No significant

difference was seen in both the NL and AD groups. For PTAU levels, no significant difference was seen

across all groups. For Brain Florbetapir PET imaging levels, at baseline, there was significant differences

between OSA groups for both NL and MCI participants (P =0.02 for all), with OSA+ participants having

significantly lower brain Florbetapir levels. No difference was seen in AD group. For the MCI group, the

inclusion of an interaction term OSA*APOE4 was also significant (P =0.01).

Rate of Change in AD Biomarker by OSA status

Table 3.2 reports the covariance parameter estimates and between subject (i.e. OSA+ vs. OSA-)

variation in AD biomarker levels over time.

Brain Aβ-42 levels

In NL participants, there was significant variation by OSA status in the annual change in brain

Florbetapir PET volumes over time (slope) (mean SUVR; B = 0.06, p < .0001). The covariance between

the baseline brain Florbetapir level and the annual brain Florbetapir volume change over time was -.06,

indicating that OSA+ subjects experienced faster increase in brain Aβ-42 deposition over time (p < .0001)

when compared to OSA- subjects. In MCI participants, there was also significant variation by OSA status

in the annual change in brain Florbetapir volumes over time (slope) (mean SUVR; B = 0.08, p < .0001).

Page 183: Omonigho A. Michael Bubu

171

The covariance between the baseline brain Aβ-42 level and the annual brain Aβ-42 volume change over

time was -.006, indicating that OSA+ subjects experienced faster increase in brain Aβ-42 deposition over

time (p < .0001) when compared to OSA- subjects. For the MCI group, the inclusion of an interaction

term OSA*APOE4 was also significant (P =0.01) with APOE4+/OSA+ individuals showing a faster

increase in brain Aβ-42 compared to APOE4-/OSA- patients. No significant variation in the change in

brain Florbetapir PET volumes by OSA status over time was seen for the AD group.

CSF Aβ-42, TAU and PTAU levels

For the NL and MCI groups, there was significant variation in the annual change in CSF Aβ-42

by OSA status (mean SUVR; B = -2.708, mean SUVR; B = -2.264, p < .0001 for all respectively) over

time. Significant differences in the annual change in TAU and PTAU levels over time (slope) were also

seen in both groups. Covariance parameters between the baseline CSF Aβ-42, TAU and PTAU volume

change over time indicated that OSA+ subjects experienced a faster decrease in CSF Aβ-42 and increases

in TAU and PTAU volumes over time (p < .0001 for all) compared to OSA- participants, in both the CN

and MCI groups (Table 3.2). No significant variations in the change in CSF Aβ-42, TAU and PTAU over

time was seen for the AD group.

DISCUSSION

Major findings from this study include the following:

i. At baseline, OSA+ patients had significantly higher CSF Aβ-42 in the MCI group; and lower

brain Florbetapir PET in NL and MCI groups; and lower TAU levels in the MCI group,

compared to OSA- patients.

ii. Longitudinally, there was significant differences in the annual rate of change in brain Florbetapir

PET, CSF Aβ-42, TAU and PTAU levels over the follow up period for the NL and MCI group

iii. OSA+ subjects experienced significantly faster increase in brain Aβ-42, TAU and PTAU levels

and a faster decrease in CSF Aβ-42 over the follow-up period in both the NL and MCI groups.

Page 184: Omonigho A. Michael Bubu

172

iv. For the MCI group, APOE4+/OSA+ individuals showed a faster increase in brain Aβ-42

compared to APOE4-/OSA- patients

Baseline AD Biomarker levels by clinical group and OSA status

At baseline, the data showed that OSA+ patients had significantly higher CSF Aβ-42 and lower

brain Aβ-42 in NL and MCI groups, and lower TAU levels in the MCI group, compared to OSA- patients.

Theoretically, this may be attributed to slow wave sleep (SWS) occurring during non-rapid eye movement

(NREM) sleep, a known Aβ modulator.43-45 Sleep is involved in the clearance of Aβ,45 and fragmented

SWS limits brain Aβ clearance, thereby leaving higher levels that reflect in the CSF.46,47 At cross-section,

disrupted SWS has been shown to be associated with higher CSF Aβ-42 in middle-aged48,49 and older

adults.50 It is also important to note that CSF tau protein pathology and CSF Aβ-42 burden may not be

specific to AD in these participants.51 The effect of normal aging in NL subjects on CSF turnover and

brain Aβ-42 clearance is still not fully understood.52,53 Furthermore, cross-sectional data show little

evidence for an association between age and CSF Aβ-42.54,55 It is also noteworthy that the functional

significance of high CSF Aβ42 levels is not fully known.

Rate of Change in AD Biomarker by OSA status

The major objective of this study was to examine the effect of OSA on longitudinal changes in

brain amyloid deposition, and AD CSF biomarkers. There were significant differences in the annual rate

of change in brain Florbetapir PET, CSF Aβ-42, TAU and PTAU levels over the follow-up period for the

NL and MCI group, with OSA+ subjects experiencing significantly faster increase in brain Florbetapir

PET, TAU and PTAU levels and a faster decrease in CSF Aβ-42. The direction of these longitudinal

changes are in cognizance with emanating evidence-linking disturbed sleep, OSA with AD pathogenesis.

Self-reported sleep duration and quality has been shown to be associated with higher brain Aβ-42

burden.19 MCI individuals with higher apnea-hypopnea index and oxygen desaturation index

demonstrated higher brain Aβ-42 burden globally and regionally in the precuneus.20 A recent study by

Yun et al56 examined whether OSA increased brain amyloid burden relative to controls in middle-aged

Page 185: Omonigho A. Michael Bubu

173

participants from the Korean Genome and Epidemiology Study. After adjusting for potential confounders,

OSA patients had a higher brain amyloid burden in the right posterior cingulate gyrus and right temporal

cortex, relative to controls, suggesting that OSA may have been responsible for these findings. Bu et al.17

examined whether hypoxia indices in OSA was associated with serum amyloid (Aβ40, Aβ42), total tau

and phosphorylated tau 181 (P-tau 181) comparing their levels with controls. Results from this study

demonstrated significantly higher levels of serum Aβ40, Aβ42, total Aβ and P-tau 181 in OSA patients

compared to control. Hypoxia indices including apnea-hypopnea index, the oxygen desaturation index,

and the mean and lowest oxyhemoglobin saturations correlated positively with Aβ40, Aβ42, and total Aβ

in OSA patients, suggesting that hypoxia possibly effects AD pathogenesis. In a population of MCI

individuals, Liguori et al57 compared CSF β-amyloid42, tau proteins, and lactate levels in OSA versus

CPAP treated OSA and controls. Findings from the study demonstrated lower CSF β-amyloid42, higher

lactate levels and t-tau/ Aβ42 ratio compared to controls and CPAP treated OSA patients. Controls and

CPAP treated OSA MCI patients had similar AD biomarker levels. These findings suggest that OSA may

effect early AD biomarker changes that may be susceptible to CPAP treatment.

In our study, among MCI patients, APOE4+/OSA+ individuals showed a faster increase in brain

Aβ-42 compared to APOE4-/OSA- patients, suggesting that the APOE4 allele may be a moderator of the

association between OSA and AD. In a population of cognitive normal (NL) older adults, Osorio et al18

demonstrated an association between intermittent hypoxia and increases in CSF T-Tau, P-Tau and Aβ42

in ApoE3+, suggesting that hypoxia may be responsible for changes in the AD biomarkers in this

population. In ApoE4+ subjects, OSA severity tended towards lower CSF Aβ42 levels. There was no

difference in the rate of biomarker change in the AD group by OSA status. This may be because in AD

patients, risk profile is possibly linear with brain atrophy until it reaches a threshold thereby exhibiting a

ceiling effect at higher levels of brain Aβ.32 OSA’s effect as it relates to AD biomarker changes may

therefore be attenuated or have reached its maximum.

Page 186: Omonigho A. Michael Bubu

174

Strengths and Limitations

Our study possesses several strengths including a well-defined cohort, and objective assessment

of CSF P-Tau, Hippocampal atrophy and β-amyloid burden, which allowed for a high degree of certainty

regarding measurement of our outcome.58 Furthermore, our statistical analytic methods were robust with

respect to unbalanced number of observations per subject over time. However, we were limited by our

measurement of OSA since this was self-reported. It is known that self-reported sleep measures can be

impacted by diminished cognition59 and in certain situations might not be correlated with objective

measurements using polysomnography or wrist actigraphy.60 Moreover any positive findings from our

study will need to be replicated using objective measures of sleep function. The ADNI racial demographic

is largely white; as such this limits the generalizability of our findings to other racial populations.

CONCLUSIONS

OSA appears to accelerate increases in brain amyloid deposition, CSF TAU and PTAU and

decreases in CSF biomarkers burden, over time, both in elderly Cognitive Normal and MCI individuals.

Sleep fragmentation and/or intermittent hypoxia from OSA are likely candidate mechanisms. Thus,

clinical interventions aimed at OSA, such as treatment with CPAP or dental appliances, in cognitive

normal and MCI patients, could possibly mitigate or slow the progression of cognitive impairment to AD.

Further research examining mechanisms underlying these observed effects are needed.

References

1. Aloia MS, Arnedt JT, Davis JD, Riggs RL, Byrd D. Neuropsychological sequelae of obstructive

sleep apnea-hypopnea syndrome: a critical review. Journal of the International

Neuropsychological Society : JINS. 2004;10(5):772-785.

2. Castronovo V, Canessa N, Strambi LF, et al. Brain activation changes before and after PAP

treatment in obstructive sleep apnea. Sleep. 2009;32(9):1161-1172.

Page 187: Omonigho A. Michael Bubu

175

3. Ju G, Yoon IY, Lee SD, Kim TH, Choe JY, Kim KW. Effects of sleep apnea syndrome on

delayed memory and executive function in elderly adults. Journal of the American Geriatrics

Society. 2012;60(6):1099-1103.

4. Young T, Peppard PE, Gottlieb DJ. Epidemiology of obstructive sleep apnea: a population health

perspective. American journal of respiratory and critical care medicine. 2002;165(9):1217-1239.

5. 2015 Alzheimer's disease facts and figures. Alzheimer's & dementia : the journal of the

Alzheimer's Association. 2015;11(3):332-384.

6. Hebert LE, Beckett LA, Scherr PA, Evans DA. Annual incidence of Alzheimer disease in the

United States projected to the years 2000 through 2050. Alzheimer Dis Assoc Disord.

2001;15(4):169-173.

7. Hebert LE, Weuve J, Scherr PA, Evans DA. Alzheimer disease in the United States (2010-2050)

estimated using the 2010 census. Neurology. 2013;80(19):1778-1783.

8. Heinzer R, Marti-Soler H, Haba-Rubio J. Prevalence of sleep apnoea syndrome in the middle to

old age general population. The Lancet Respiratory medicine. 2016;4(2):e5-6.

9. Chiatti C, Furneri G, Rimland JM, et al. The economic impact of moderate stage Alzheimer's

disease in Italy: evidence from the UP-TECH randomized trial. International psychogeriatrics /

IPA. 2015;27(9):1563-1572.

10. Choo WY, Low WY, Karina R, Poi PJ, Ebenezer E, Prince MJ. Social support and burden among

caregivers of patients with dementia in Malaysia. Asia-Pacific journal of public health.

2003;15(1):23-29.

11. Frahm-Falkenberg S, Ibsen R, Kjellberg J, Jennum P. Health, social and economic consequences

of dementias: a comparative national cohort study. Eur J Neurol. 2016;23(9):1400-1407.

12. Gerves C, Bellanger MM, Ankri J. Economic analysis of the intangible impacts of informal care

for people with Alzheimer's disease and other mental disorders. Value in health : the journal of

the International Society for Pharmacoeconomics and Outcomes Research. 2013;16(5):745-754.

Page 188: Omonigho A. Michael Bubu

176

13. Gibson GJ. Obstructive sleep apnoea syndrome: underestimated and undertreated. British medical

bulletin. 2004;72:49-65.

14. Gray A, Fenn P. Alzheimer's disease: the burden of the illness in England. Health trends.

1993;25(1):31-37.

15. Hossain JL, Shapiro CM. The prevalence, cost implications, and management of sleep disorders:

an overview. Sleep Breath. 2002;6(2):85-102.

16. Lopez-Bastida J, Serrano-Aguilar P, Perestelo-Perez L, Oliva-Moreno J. Social-economic costs

and quality of life of Alzheimer disease in the Canary Islands, Spain. Neurology.

2006;67(12):2186-2191.

17. Bu XL, Liu YH, Wang QH, et al. Serum amyloid-beta levels are increased in patients with

obstructive sleep apnea syndrome. Scientific reports. 2015;5:13917.

18. Osorio RS, Ayappa I, Mantua J, et al. The interaction between sleep-disordered breathing and

apolipoprotein E genotype on cerebrospinal fluid biomarkers for Alzheimer's disease in

cognitively normal elderly individuals. Neurobiol Aging. 2014;35(6):1318-1324.

19. Spira AP, Gamaldo AA, An Y, et al. Self-reported Sleep and beta-Amyloid Deposition in

Community-Dwelling Older Adults. JAMA Neurol. 2013.

20. Spira AP, Yager C, Brandt J, et al. Objectively Measured Sleep and beta-amyloid Burden in

Older Adults: A Pilot Study. SAGE open medicine. 2014;2.

21. Yun CH, Lee HY, Lee SK, et al. Amyloid Burden in Obstructive Sleep Apnea. Journal of

Alzheimer's disease : JAD. 2017.

22. Hampel H, Burger K, Teipel SJ, Bokde AL, Zetterberg H, Blennow K. Core candidate

neurochemical and imaging biomarkers of Alzheimer's disease. Alzheimers Dement.

2008;4(1):38-48.

23. Hampel H, Teipel SJ, Fuchsberger T, et al. Value of CSF beta-amyloid1-42 and tau as predictors

of Alzheimer's disease in patients with mild cognitive impairment. Mol Psychiatry.

2004;9(7):705-710.

Page 189: Omonigho A. Michael Bubu

177

24. Shaw LM, Vanderstichele H, Knapik-Czajka M, et al. Cerebrospinal fluid biomarker signature in

Alzheimer's disease neuroimaging initiative subjects. Annals of neurology. 2009;65(4):403-413.

25. Brys M, Pirraglia E, Rich K, et al. Prediction and longitudinal study of CSF biomarkers in mild

cognitive impairment. Neurobiol Aging. 2009;30(5):682-690.

26. Waragai M, Okamura N, Furukawa K, et al. Comparison study of amyloid PET and voxel-based

morphometry analysis in mild cognitive impairment and Alzheimer's disease. J Neurol Sci.

2009;285(1-2):100-108.

27. Bourgeat P, Chetelat G, Villemagne VL, et al. Beta-amyloid burden in the temporal neocortex is

related to hippocampal atrophy in elderly subjects without dementia. Neurology. 2010;74(2):121-

127.

28. Bouwman FH, Schoonenboom NS, Verwey NA, et al. CSF biomarker levels in early and late

onset Alzheimer's disease. Neurobiol Aging. 2009;30(12):1895-1901.

29. Carlson NE, Moore MM, Dame A, et al. Trajectories of brain loss in aging and the development

of cognitive impairment. Neurology. 2008;70(11):828-833.

30. Buerger K HH. Evaluation of phosporylated tau protein as a core biomarker of Alzheimer’s

disease. . In: Iqbal I, Winblad B, editors Proceedings of the 9th International Conference on

Alzheimer’s Disease and Related Disorders Philadelphia, Pennsylvania: Alzheimer’s

Association; 2004.p. 46–58.

31. Frank RA, Galasko D, Hampel H, et al. Biological markers for therapeutic trials in Alzheimer's

disease. Proceedings of the biological markers working group; NIA initiative on neuroimaging in

Alzheimer's disease. Neurobiol Aging. 2003;24(4):521-536.

32. Jack CR, Jr., Wiste HJ, Vemuri P, et al. Brain beta-amyloid measures and magnetic resonance

imaging atrophy both predict time-to-progression from mild cognitive impairment to Alzheimer's

disease. Brain : a journal of neurology. 2010;133(11):3336-3348.

33. Fitzpatrick AL, Kuller LH, Lopez OL, et al. Midlife and late-life obesity and the risk of dementia:

cardiovascular health study. Arch Neurol. 2009;66(3):336-342.

Page 190: Omonigho A. Michael Bubu

178

34. Petersen RC, Aisen PS, Beckett LA, et al. Alzheimer's Disease Neuroimaging Initiative (ADNI):

clinical characterization. Neurology. 2010;74(3):201-209.

35. Petersen RC, Smith GE, Waring SC, Ivnik RJ, Tangalos EG, Kokmen E. Mild cognitive

impairment: clinical characterization and outcome. Arch Neurol. 1999;56(3):303-308.

36. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of

Alzheimer's disease: report of the NINCDS-ADRDA Work Group under the auspices of

Department of Health and Human Services Task Force on Alzheimer's Disease. Neurology.

1984;34(7):939-944.

37. Landau SM, Breault C, Joshi AD, et al. Amyloid-beta imaging with Pittsburgh compound B and

florbetapir: comparing radiotracers and quantification methods. Journal of nuclear medicine :

official publication, Society of Nuclear Medicine. 2013;54(1):70-77.

38. Landau SM, Lu M, Joshi AD, et al. Comparing positron emission tomography imaging and

cerebrospinal fluid measurements of beta-amyloid. Annals of neurology. 2013;74(6):826-836.

39. Harville DA. Maximum Likelihood Approaches to Variance Component Estimation and to

Related Problems. . Journal of the American Statistical Association. 1977;72((358):):p. 339-340.

40. Jennrich RI, Schluchter MD. Unbalanced repeated-measures models with structured covariance

matrices. Biometrics. 1986;42(4):805-820.

41. Laird NM, Ware JH. Random-effects models for longitudinal data. Biometrics. 1982;38(4):963-

974.

42. Ten Have TR, Kunselman AR, Pulkstenis EP, Landis JR. Mixed effects logistic regression

models for longitudinal binary response data with informative drop-out. Biometrics.

1998;54(1):367-383.

43. Cirrito JR, Yamada KA, Finn MB, et al. Synaptic activity regulates interstitial fluid amyloid-beta

levels in vivo. Neuron. 2005;48(6):913-922.

44. Nir Y, Staba RJ, Andrillon T, et al. Regional slow waves and spindles in human sleep. Neuron.

2011;70(1):153-169.

Page 191: Omonigho A. Michael Bubu

179

45. Xie L, Kang H, Xu Q, et al. Sleep drives metabolite clearance from the adult brain. Science (New

York, NY). 2013;342(6156):373-377.

46. Hosselet J, Ayappa I, Norman RG, Krieger AC, Rapoport DM. Classification of sleep-disordered

breathing. American journal of respiratory and critical care medicine. 2001;163(2):398-405.

47. Hosselet JJ, Norman RG, Ayappa I, Rapoport DM. Detection of flow limitation with a nasal

cannula/pressure transducer system. American journal of respiratory and critical care medicine.

1998;157(5 Pt 1):1461-1467.

48. Ju YE, Finn MB, Sutphen CL, et al. Obstructive sleep apnea decreases central nervous system-

derived proteins in the cerebrospinal fluid. Annals of neurology. 2016;80(1):154-159.

49. Ju YS, Ooms SJ, Sutphen C, et al. Slow wave sleep disruption increases cerebrospinal fluid

amyloid-beta levels. Brain : a journal of neurology. 2017;140(8):2104-2111.

50. Varga AW, Wohlleber ME, Gimenez S, et al. Reduced Slow-Wave Sleep Is Associated with High

Cerebrospinal Fluid Abeta42 Levels in Cognitively Normal Elderly. Sleep. 2016;39(11):2041-

2048.

51. de Leon MJ, DeSanti S, Zinkowski R, et al. MRI and CSF studies in the early diagnosis of

Alzheimer's disease. J Intern Med. 2004;256(3):205-223.

52. Bading JR, Yamada S, Mackic JB, et al. Brain clearance of Alzheimer's amyloid-beta40 in the

squirrel monkey: a SPECT study in a primate model of cerebral amyloid angiopathy. J Drug

Target. 2002;10(4):359-368.

53. Silverberg GD, Levinthal E, Sullivan EV, et al. Assessment of low-flow CSF drainage as a

treatment for AD: results of a randomized pilot study. Neurology. 2002;59(8):1139-1145.

54. Andreasen N, Minthon L, Davidsson P, et al. Evaluation of CSF-tau and CSF-Abeta42 as

diagnostic markers for Alzheimer disease in clinical practice. Arch Neurol. 2001;58(3):373-379.

55. Hulstaert F, Blennow K, Ivanoiu A, et al. Improved discrimination of AD patients using beta-

amyloid(1-42) and tau levels in CSF. Neurology. 1999;52(8):1555-1562.

Page 192: Omonigho A. Michael Bubu

180

56. Yun CH, Lee HY, Lee SK, et al. Amyloid Burden in Obstructive Sleep Apnea. Journal of

Alzheimer's disease : JAD. 2017;59(1):21-29.

57. Liguori C, Mercuri NB, Izzi F, et al. Obstructive Sleep Apnea is Associated With Early but

Possibly Modifiable Alzheimer's Disease Biomarkers Changes. Sleep. 2017;40(5).

58. Johnson KA, Fox NC, Sperling RA, Klunk WE. Brain imaging in Alzheimer disease. Cold Spring

Harb Perspect Med. 2012;2(4):a006213.

59. Van Den Berg JF, Van Rooij FJ, Vos H, et al. Disagreement between subjective and actigraphic

measures of sleep duration in a population-based study of elderly persons. J Sleep Res.

2008;17(3):295-302.

60. Unruh ML, Redline S, An MW, et al. Subjective and objective sleep quality and aging in the

sleep heart health study. J Am Geriatr Soc. 2008;56(7):1218-1227.

Probably need to add Figure Legends here.

Page 193: Omonigho A. Michael Bubu

181

Table 3.1: Descriptive Characteristics of Participants by Obstructive Sleep Apnea Status at Baseline

Cognitive Normal

Characteristics All OSA- OSA+

Number of participants (%) 516 (100) 487 (94) 29 (6) Female gender, number (%) 253 (49) 241 (49) 9 (32) Age, years, median (interquartile range) 74 (71, 78) 71 (70, 78) 71 (70, 76)

APOE positive, number (%) 145 (28) 140 (29) 5 (17) Education, years, median (interquartile range) 16 (14, 18) 16 (14, 18) 16 (15, 18) BMI (kg/m2) 27.2 ± 4.8 27.1 ± 4.7 29.8 ± 5.9 Hypertension, number (%) 251 (49) 235 (48) 16 (55) Diabetes, number (%) 45 (9) 40 (8) 5 (17) Thyroid Disease, number (%) 112 (22) 109 (22) 3 (10)

Respiratory Disease, number (%) 123 (24) 100 (21) 23 (79)

CSF-ABETA pg/ml median (interquartile range) 210 (155,

241) 209 (155,

241) 226 (199, 259)

TAU pg/ml median (interquartile range) 59 (45, 84) 59 (45 83) 56 (48, 84) PTAU pg/ml median (interquartile range) 27 (20, 40) 27 (20, 42) 27 (20, 32) Aβ, median (interquartile range) 1.1 (1.0, 1.2) 1.1 (1.0, 1.2) 1.0 (1.0, 1.0)

Mild Cognitive Impairment

Characteristics All OSA- OSA+

Number of participants (%) 798 (100) 695 (87) 103 (13) Female gender, number (%) 319 (40) 297 (43) 25 (25) Age, years, median (interquartile range) 74 (68, 79) 71 (70, 78) 71 (70, 76) APOE positive, number (%) 410 (51) 368 (53) 42 (41)

Education, years, median (interquartile range) 16 (14, 18) 16 (14, 18) 16 (14, 18) BMI (kg/m2) 26.9 ± 4.6 26.5 ± 4.4 29.4 ± 5.3 Hypertension, number (%) 395 (49) 337 (48) 58 (56) Diabetes, number (%) 75 (9) 57 (8) 18 (17) Thyroid Disease, number (%) 156 (20) 133 (19) 23 (22) Respiratory Disease, number (%) 195 (24) 133 (19) 62 (60)

CSF-ABETA pg/ml median (interquartile range) 153 (130,

209) 150 (128,

206) 169 (139, 214)

TAU pg/ml median (interquartile range) 80 (54, 116) 81 (54, 122) 67 (51, 97) PTAU pg/ml median (interquartile range) 36 (23, 51) 37 (23, 52) 32 (21, 44)

Aβ, median (interquartile range) 1.2 (1.0, 1.4) 1.2 (1.0, 1.4) 1.1 (1.0, 1.3)

Alzheimer's Disease

Characteristics All OSA- OSA+

Number of participants (%) 325 (100) 303 (93) 22 (7) Female gender, number (%) 119 (37) 113 (37) 6 (27)

Age, years, median (interquartile range) 76 (71, 80) 76 (71, 80) 71 (64, 76)

Page 194: Omonigho A. Michael Bubu

182

APOE positive, number (%) 216 (66) 198 (65) 18 (82) Education, years, median (interquartile range) 16 (14, 18) 16 (14, 18) 16 (14, 18) BMI (kg/m2) 25.9 ± 4.6 25.6 ± 4.3 29.0 ± 6.8 Hypertension, number (%) 165 (51) 149 (49) 16 (73) Diabetes, number (%) 32 (10) 30 (10) 2 (9)

Thyroid Disease, number (%) 65 (20) 61 (20) 4 (18) Respiratory Disease, number (%) 67 (21) 52 (17) 15 (68)

CSF-ABETA pg/ml median (interquartile range) 132 (116,

151) 115 (96,

182) 115 (79, 151)

TAU pg/ml median (interquartile range) 80 (54, 116) 67 (51, 97) 81 (54, 122) PTAU pg/ml median (interquartile range) 42 (33, 61) 41 (33, 61) 54 (35, 66)

Aβ, median (interquartile range) 1.4 (1.3, 1.5) 1.4 (1.3, 1.5) 1.4 (1.3, 1.6) Abbreviation: Aβ: amyloid beta, APOE: Apolipoprotein epsilon, BMI: body mass index, CSF: cerebrospinal fluid, TAU: tau protein, PTAU: phosphorylated tau

Table 3.2 Covariance Parameter estimates and Between Subject variation in AD Biomarker Deposition

Parameters Estimate 95% CI P-value

OSA+ vs. OSA- (Cognitive Normal Patients)

β-Amyloid Burden over time (slope) 0.06 .02, .11 <.0001

β-Amyloid Burden over time (covariance) -0.06 -.09, -.04 <.0001

CSF Aβ-42 volume over time (slope) -2.71 -3.11, -2.35 <.0001

CSF Aβ-42 volume over time (covariance) 3.93 3.56 - 4.31 <.0001

CSF TAU volume over time (slope) 3.68 3.31 - 4.07 <.0001

CSF TAU volume over time (covariance) -2.89 -3.51, -2.29 <.0001

CSF PTAU volume over time (slope) 1.22 1.02 - 1.42 <.0001

CSF PTAU volume over time (covariance) -1.21 -1.71, - .74 <.0001

OSA+ vs. OSA- (Mild Cognitive Impairment Patients)

β-Amyloid Burden over time (slope) 0.08 .05, .12 <.0001

β-Amyloid Burden over time (covariance) -0.06 -.09, -.04 <.0001

CSF Aβ-42 volume over time (slope) -2.62 -3.23, -2.03 <.0001

CSF Aβ-42 volume over time (covariance) 2.69 2.02, 3.36 <.0001

CSF TAU volume over time (slope) 2.21 1.58, 2.86 <.0001

CSF TAU volume over time (covariance) -1.89 -2.91, -.87 <.0001

CSF PTAU volume over time (slope) 1.74 1.22, 2.27 <.0001

CSF PTAU volume over time (covariance) -1.48 -2.05, -.94 <.0001

OSA+ vs. OSA- (Alzheimer's disease Patients)

β-Amyloid Burden over time (slope) 0.07 -1.19, 1.33 0.33

β-Amyloid Burden over time (covariance) -0.29 -2.07, 1.49 0.31

Page 195: Omonigho A. Michael Bubu

183

CSF Aβ-42 volume over time (slope) -1.11 -3,31, 1.09 0.53

CSF Aβ-42 volume over time (covariance) -1.14 -3.38, 1.63 0.56

CSF TAU volume over time (slope) 0.26 -1.02, 1,28 0.47

CSF TAU volume over time (covariance) -0.15 -1.94, 1.64 0.47

CSF PTAU volume over time (slope) 0.94 0.23, 1.65 0.11

CSF PTAU volume over time (covariance) -0.16 -1.66, 1.34 0.11 Abbreviation: Aβ: amyloid beta, APOE: Apolipoprotein epsilon, BMI: body mass index, CSF: cerebrospinal fluid, TAU: tau protein, PTAU: phosphorylated tau

Models were adjusted for age, sex, BMI, CPAP use, APOE e4 status, and history of chronic medical

conditions.

Page 196: Omonigho A. Michael Bubu

184

Figure 3.1: Baseline Alzheimer Disease Biomarker burden and Brain

Aβ-42 levels in Cognitive Normal subjects by OSA status

Page 197: Omonigho A. Michael Bubu

185

Figure 3.2: Baseline Alzheimer Disease Biomarker burden and Brain

Aβ-42 levels in Mild Cognitive Impairment subjects by OSA status

Page 198: Omonigho A. Michael Bubu

186

Figure 3.3: Baseline Alzheimer Disease Biomarker burden and Brain

Aβ-42 levels in Alzheimer disease subjects by OSA status

Page 199: Omonigho A. Michael Bubu

187

SECTION 4

Obstructive Sleep Apnea: A Distinct Physiological Phenotypic Risk Factor in older adults with Cognitive

decline and Alzheimer’s disease

ABSTRACT

Introduction: Studies suggest that Obstructive Sleep Apnea (OSA) in the elderly may result in varying

functional outcomes relative to OSA in the middle-aged. Therefore, understanding and appreciating the

heterogeneity of OSA and its outcomes in distinct age groups especially at it relates to cognition,

subsequent cognitive decline and Alzheimer disease (AD), is critical in mitigating the deleterious effects

of OSA.

Methods: In this review, we integrated over 3 decades of research examining OSA and cognition; OSA

and subsequent cognitive decline; and OSA and AD, with particular focus in appreciating the

heterogeneity of OSA and its outcomes in distinct age groups. A systematic literature search of

bibliographic databases including PubMed/Medline, Embase, Psych INFO and Cochrane library for

clinical trials, was conducted to identify all eligible studies (published from their onset up until August

31, 2017) that examined associations between OSA and cognitive function, OSA and subsequent

cognitive decline, and OSA and AD

Results: Thirty-four studies examining the association between OSA and cognition, 7 studies examining

the association between OSA and subsequent cognitive decline, and 15 studies examining the association

of OSA and AD or AD pathology were identified by the literature search after applying specific inclusion

and exclusion criteria. The data suggests that OSA is associated with greater cognitive deficits in middle-

Page 200: Omonigho A. Michael Bubu

188

age adults than in older adults and the elderly; greater risk of subsequent cognitive decline in middle-aged

adults than in older adults and elderly; and in older adults with MCI rather than in healthy older adults,

OSA may be the critical correlate of cognition.

Conclusion: OSA may be age-dependent in older adults (60 – 70 years old) and the elderly (70 years and

above) and is associated with neurodegenerative diseases particularly, cognitive decline and AD. In the

middle-aged (30 – 60 years old), the data suggests that OSA may be age-related and presents with a

distinct phenotype, with cardiovascular, and possibly metabolic effects. Intermittent hypoxia and sleep

fragmentation are two main processes by which OSA induces neurodegenerative changes.

INTRODUCTION

Cognitive decline in older adults and Alzheimer’s disease (AD) both have debilitating effects on

individuals affected, with significant socioeconomic implications.1-4 It is well known that advanced age is

the most notable risk factor for cognitive decline and AD. With projected substantial increase in the

ageing population, it is pertinent that efforts directed at comprehending risk factors of cognitive decline

and AD be consolidated. There is increasing evidence in recent years linking cognitive decline and AD to

various sleep problems and disorders.5-8 Notably, the link between cognitive impairment and Obstructive

Sleep Apnea (OSA) is well-established,9-12 and recent evidence suggest that a longitudinal risk between

OSA, cognitive decline and AD exist.13-15 It is therefore vital to clearly understand the distinct and unique

relationship OSA has with cognitive decline and AD in the elderly as this will help inform prevention and

treatment strategies of both OSA and AD.

Age-related and Age-dependent OSA co-morbidities

Obstructive sleep apnea (OSA) is characterized by intermittent hypoxemia, sleep fragmentation

and intrathoracic pressure changes.16,17 OSA is an issue of public health significance, because it is highly

prevalent,18-21 results in serious morbidity and significant mortality, 22-26 and has a high socio-economic

Page 201: Omonigho A. Michael Bubu

189

impact.27,28 Studies suggest that OSA in the elderly may result in varying functional outcomes relative to

OSA in the middle-aged. Therefore, understanding and appreciating the heterogeneity of OSA and its

outcomes in distinct age groups is critical in mitigating the deleterious effects of OSA.

Bliwise et al.29 in their study using the Bay Area Sleep Cohort described two peak incidences of

OSA, one being age-dependent and the other being age-related. The concept describes OSA as an age-

dependent condition in the elderly, relative to OSA being an age-related condition in the middle-aged.

OSA affects 3–7% of the middle-aged population and becomes more prevalent with increasing age.20,30-37

In middle-aged individuals, Bliwise noted that OSA is thought to show an age-related occurrence

conferring a specific period of susceptibility,38,39 in association with disparate morbidities. Age-related

diseases and disorders are not particularly related to the process of aging, occurring at a distinctive

age/age-group and then lessening in frequency with increasing age (e.g. multiple sclerosis and

amyotrophic lateral sclerosis).29

Conversely, in the elderly, the prevalence of OSA ranges from 30-80%,32,40-46 contingent on the

definition of OSA. The significance of these high rates in the elderly is ambiguous; while certain studies

suggest reduced mortality with increasing age,47 others suggest otherwise.48 However, the pathogenesis of

OSA somewhat appears to involve normal aging with exponential increases in mortality and morbidity

with advancing age.49,50 This physiologic susceptibility with increased incidence with chronological age

suggests that OSA is age-dependent in the elderly. Mechanisms underlying how aging increases the risk

of OSA are not completely understood. The effects of advancing age are generally considered to function

with other physiologic systems experiencing aging.51,52 Notable factors in OSA pathogenesis during

aging include an extremely collapsible airway, limited upper airway dilator muscle activity, diminished

respiratory arousal threshold, and a precarious ventilatory control system.35,53,54 Furthermore, age-

dependent changes in lung function as reported in cross-sectional studies,55-57 are associated with

increments in OSA frequency.

Page 202: Omonigho A. Michael Bubu

190

Much focus on OSA’s age-dependence in the elderly has been in cardiovascular outcomes

relative to cognitive outcomes. However, the various co-morbidities associated with increasing age in

OSA increases susceptibility to cognitive decline.58,59 Cardiovascular outcomes of OSA such as

hypertension, coronary heart disease, and congestive heart failure; 25,60,61 stroke; 62-64 multiple

inflammatory and metabolic effects including diabetes; 65-68 are associated with increased severity of

OSA, and may be more likely to occur in young and middle-aged (30 – 70 years), rather than older

populations.69-73 In young and middle-aged adults, substantive evidence exists establishing relationships

between OSA and cognitive function, including areas such as memory, motor, vigilance and executive

functions.74-76 Studies examining cardiovascular outcomes of OSA in the elderly show varying findings.

Some studies77-79 report increased mortality and incidence of stroke in older adults while other

studies47,48,80 report decreased mortality with age in OSA patients compared to the general population. In

the elderly, multiple studies demonstrate associations between OSA and cognitive decline.15,81-83 We

demonstrated that in the elderly, OSA patients had an earlier age of cognitive decline to MCI and to AD

than non-OSA controls,84 and showed that individuals with OSA were more than two times more likely to

have AD compared to non-OSA individuals in a recent meta-analysis.5 A meta-analysis of cross-sectional

studies also concluded OSA-AD associations, such that AD patients were at an increased risk of

presenting with OSA than cognitively normal individuals.85 Of particular concern is that longitudinal

studies also provide evidence of an association between OSA and cognitive decline. A 5-year prospective

study of older women (mean ±SD age: 82.3 ±3.2 years) found elevated oxygen desaturation index (≥15

events/ hr) to be associated with increased risk of developing mild cognitive impairment (MCI) or

dementia.15 In general, the literature regarding OSA, cognitive function, cognitive decline and AD have a

relationship with advancing age, suggesting that age may be an effect modifier of these association and

that OSA may present as a distinct physiologic phenotypic risk factor in older adults with cognitive

decline and AD.

Page 203: Omonigho A. Michael Bubu

191

Review Objective

Recent narrative reviews on OSA, Cognitive Decline and AD described the cognitive profiles

found in association with OSA in children and adults (young, middle-aged and elderly adults);9,86

explored mechanisms, neurobiology and treatment for geriatric psychiatry audience; and discussed

probable explanatory mechanisms linking OSA, depression and cognitive dysfunction. 87-89 Other

narrative discussions focused on the probable explanatory mechanisms linking OSA to dementia.82

Previous systematic and meta-reviews focused on how OSA affects specific neurocognitive domains,

producing inconsistent74,90 and sometimes non-conclusive findings.12,91 The only meta-review focusing on

older adults reported a small association between OSA and cognitive dysfunction and suggested that some

individuals may be more at risk of adverse cognitive effects.92

In this review, we integrate over 3 decades of research examining OSA and cognition; OSA and

subsequent cognitive decline; and OSA and AD; with particular focus in appreciating the heterogeneity of

OSA and its outcomes in distinct age groups. We note that the data suggests that OSA may be age-

dependent in older adults (60 – 70 years old) and the elderly (70 years and above) and is associated with

neurodegenerative diseases particularly, cognitive decline and AD. In the middle-aged (30 – 60 years

old), the data suggests that OSA may be age-related and presents with a distinct phenotype, with

cardiovascular, and possibly metabolic effects. We also explore and describe possible mechanisms linking

OSA as a precipitator of AD pathogenesis especially in the elderly, by systematically reviewing clinical

and epidemiological evidence of such an association. Where findings are discrepant, we focus on

methodological differences among studies.

METHODS

Search strategy

A systematic literature search of bibliographic databases including PubMed/Medline, Embase,

Psych INFO and Cochrane library for clinical trials, was conducted to identify all eligible studies

Page 204: Omonigho A. Michael Bubu

192

(published from their onset up until August 31, 2017) that examined associations between OSA and

cognitive function, OSA and subsequent cognitive decline, and OSA and AD. Our search strategy utilized

the combination of terms characterizing cognitive function, cognitive impairment, AD or AD pathology

as the dependent variables, OSA as the independent variable and a third set of terms specifying specific

study types including clinical and epidemiological studies. For the dependent variables, search terms

included cognition, cognitive function, executive function, neuropsychological function, cognitive

decline, mild cognitive impairment, Alzheimer’s disease, AD biomarkers and amyloid beta. For the

independent variable, search terms included sleep apnea; sleep disordered breathing and obstructive sleep

apnea. All terms were combined with the Boolean Logic operator OR, within each set, and the three sets

were combined using AND. Furthermore, we performed a manual search of references of included

articles to identify relevant references not identified by the automated search. Indexing and management

of the references was done using EndNote X7 (Thompson Reuters, New York, NY). This review was

conducted adhering to the preferred reporting items for systematic reviews and meta-analyses (PRISMA)

statement by Moher et al.93

Selection criteria

Eligible studies for this review had to meet the following selection criteria: 1) be original research

investigations examining associations between OSA and cognition, OSA and cognitive decline, and OSA

and AD; 2) conducted in humans; 3) include both healthy controls and OSA patients and conducted

between group comparisons. Studies without controls that conducted within group comparisons based on

OSA severity were also considered; 4) conducted in adults (young, middle-aged and older); 5) studies

examining cognition or cognitive decline as an outcome must have utilized objective neuropsychological

cognitive tests; 6) studies examining AD or AD pathology as an outcome must have utilized objective

measures; and 7) diagnosis of OSA must have been based on polysomnography. Excluded studies include

case reports, case series, narrative or systematic reviews, abstracts or editorials. Studies conducted in

OSA patients that did not include relevant cognitive parameters (i.e. executive, motor, verbal, attention,

Page 205: Omonigho A. Michael Bubu

193

memory) and those that examined the effects of CPAP but did not include an examination of OSA vs.

control at baseline, were also excluded.

Reviewing Procedure and Data Extraction

Bibliographic data base searches were conducted first in May 2017. Another search was

conducted in August 2017 to identify manuscripts published between May and August 2017. Independent

examination of all titles and abstracts of identified eligible studies by the search strategy was performed

by two authors (OB and XX) using EndNote X7. Selected articles at this stage were further assessed by

retrieving and reading the manuscript’s full text. Identified articles for the review were assessed for

possible inclusion by the same two authors. Where there were discordant decisions regarding inclusion, a

resolution was reached by two other authors (XX and XX). Two authors (OB and XX) performed data

extraction for each reference. Extracted fields included authors, year of publication, study design, study

population, age, exposure and outcome assessment, statistical analytic methods used, covariates, and the

main findings of the study. Two other authors (XX and XX) resolved discrepancy in the information

extracted. Reviewers were not blinded to the funding, authors, or institutions. Figure 1 shows a summary

of the study selection and retrieval process.

Assessment of Study Quality

We assessed the quality of included studies in this review using an adaptation from the modified

version of the Newcastle-Ottawa scale (NOS) for quality assessment of the observational studies,94 with

addition of new items relevant to this review. Following adaptations from previous reviews, parameters

used for the quality assessment included well-specified hypothesis, study design type, appropriately

described sample, sample size, assessment and definition of OSA, cognitive impairment or AD, statistical

analytic methods used, and approach used to adjust for potential confounders. We utilized a star rating

system with increasing number representing increasing quality. Tables 1-3.

Page 206: Omonigho A. Michael Bubu

194

RESULTS

OSA and Cognition (Cross-sectional studies)

Main findings

Table 4.1 summarizes the findings from 34 studies that examined the association of OSA and

cognitive function at a single time point. Most studies that were conducted in the middle-aged adults

consistently linked OSA and deficits in attention,95-99 memory,100-102 executive function,99,103,104 motor

function,99 reaction time,105 psychomotor vigilance106 and information processing speed.107 Explanations

and plausible mechanisms responsible for these findings in the middle-aged include daytime sleepiness or

drowsiness from fragmented sleep because of frequent apneic episodes,108-110 and hypoxia.111,112

Specifically, deficits of attention and memory may be due to fragmented sleep and excessive daytime

sleepiness,102,113 while motor function, executive function, reaction time and vigilance may be related to

the severity of hypoxemia.114-116 For example, studies where middle-aged adults with OSA who

complained of excessive daytime somnolence were compared to healthy controls, consistently showed

scores in memory and attention that were lower than normal.99,100,102 Furthermore, correlation analysis

revealed that daytime somnolence correlated with attention while nocturnal hypoxemia correlated with

executive function and visual-constructive abilities.99

Studies in the middle-aged that examined the effect of CPAP treatment on cognitive impairment

after both a short and long-term treatment period revealed conflicting results. Studies included in our

review administered CPAP and assessed for its effect after 3-months,96,97 6 months117 and 12 months.98

After 3-months of CPAP treatment, Canessa et al.96 observed significant improvements involving

memory, attention, and executive functioning in OSA patients, while Saunamaki et al.117 did not show

improvements in OSA patients’ visuospatial organizational skills or their mental set-shifting performance,

after a 6 months CPAP treatment. Variations in methods of assessing cognitive decline, and the possible

presence of confounding variables, such as rapid eye movement (REM) and slow-wave sleep (SWS)

occurring after first night of CPAP use could be responsible for the discrepant findings.

Page 207: Omonigho A. Michael Bubu

195

Castronova et al.96 employed imaging techniques using diffused tensor imaging (DTI) to examine

changes in white matter integrity and cognition following CPAP treatment in severe OSA patients. Pre-

treatment OSA patients showed impairments in most cognitive areas, and diffuse reduction of white

matter integrity. Limited changes of white matter were seen after 3 months of CPAP. However, the

authors report an almost complete reversal of white matter abnormalities over the course of 12 months

CPAP treatment. In addition, significant improvements involving memory, attention, and executive-

functioning paralleled white matter changes after treatment. Other studies using imaging techniques

showed regions of diminished grey matter volume in the left hippocampus, frontal structures of the brain

and within more lateral temporal areas in patients with OSA after CPAP treatment.97,118,119 These findings

suggest that cognitive impairment seen in middle-aged patients with moderate to severe OSA is

associated with damage to brain tissue in areas involved in several cognitive tasks. In addition, nocturnal

intermittent hypoxia and fragmented sleep could be culprits in cognitive and brain structural deficits in

OSA.

Studies that restricted their populations to older adults or whose mean age occurred in that range

(i.e., ages 61 – 70) generally show weaker results with cognition where associations were identified.

Otherwise the plurality of findings in studies where potential confounders were accounted for were null

findings. First, consider evidence for hypoxia indices and executive, memory and attention. Boland et

al.120 in their study examined the effect of Respiratory Disturbance Index (RDI) values on attention,

executive function and memory in 1700 subjects free of clinically diagnosed SDB who underwent at-

home polysomnography (PSG) as part of the Sleep Heart Health Study (SHHS). This study controlled for

age, education, occupation, field center, diabetes, hypertension, body-mass index, use of CNS

medications, and alcohol drinking status, and found no association between the RDI and any of the three

cognitive function measures assessed. Foley et al.121 assessed the association between sleep-disordered

breathing and cognitive functioning in an elderly cohort of Japanese-American men and found no

association between sleep-disordered breathing and memory function, concentration, and attention.

Phillips et al.122 and Sforza et al.123 also had null findings between AHI findings and global cognition,

Page 208: Omonigho A. Michael Bubu

196

attention, executive function, memory, language and motor functions. Dlugaj et al.124 examined the

association between Mild Cognitive Impairment (MCI) and MCI sub-types and SDB severity and found

no association. Except for Sforza and Dlugaj, the other studies made use of a healthy population and this

may have contributed to this finding.

On the contrary, other studies conducted in older adults showed associations between OSA and

delayed verbal memory,125,126 non-verbal IQ memory delayed recall,127 executive function, and

attention128,129 and decreased vigilance.130 As earlier specified most of these studies were only able to

demonstrate relatively weak associations. For example, Yesavage et al.,129 Terpening et al.,131 and Berry

et al.,127 demonstrated effects between hypoxia measures and cognitive measures that were minimally

significant. Perhaps OSA in older adults with MCI rather than in healthy older adults is the critical

correlate of cognition in older adults. Kim et al.132 specifically examined the effect of SDB in older adults

with MCI on language test performance. Individuals with MCI have an increased risk of developing AD.

Although the study found no differences between MCI and control groups on AHI and hypoxia measures,

higher AHI was associated with language test performance among individuals with MCI. These findings

possibly reflect frontal-subcortical pathology in patients with MCI suggesting that susceptibility to a

specific brain damage associated with OSA could increase the risk for AD.

Examination of studies conducted in the elderly (i.e. ages > 70 years) are also contradictory;

however, more recent studies tend to show associations between hypoxemia and executive function or

global cognition. Blackwell et al130 examined SDB and cognition in elderly men and adjusted for age,

race, education, BMI, lifestyle, comorbidities and medication use. Findings from the study showed

significant associations between SDB and cognition. Spira et al.133 conducted a similar study in elderly

women and determined whether the apolipoprotein E (APOE) e4 allele modifies the association. Results

showed that increased AHI was associated with decreased cognition and APOE4 increased this risk by

five times.

Page 209: Omonigho A. Michael Bubu

197

OSA and Cognitive Decline (Longitudinal studies)

Main findings

Table 4.2 summarizes the only seven prospective epidemiological studies that have attempted to

answer this critical question. Six of the studies were in older adults and the elderly while one study

included a mixture of middle-aged, older adults and the elderly.

In older adults and elderly, OSA at baseline may be a weak predictor of cognitive decline. Only

two studies that included older adults and the elderly reported significant cognitive associations with OSA

that were mostly modest. Blackwell et al. conducted a population-based study in a setting that included

six centers in the United States. They included community-dwelling older men without cognitive

impairment at baseline, and followed them for approximately 3 years. Results from the study showed a

modest association between nocturnal hypoxemia and subsequent decline in cognition. Martin et al

utilized a subset of individuals enrolled in the PROgnostic indicator OF cardiovascular and

cerebrovascular events, study (PROOF). They assessed whether cognitive function changes occurred in

untreated elderly SBD patients and without dementia. After an 8-year follow-up, SDB showed a

minimally significant association with decline in the attentional domain and this was more evident in the

patients with an AHI > 30. There were no significant changes in the executive and memory functions over

the follow-up period. In this study chronic hypoxemia accounted for between 4–7% of variance in the

decline in attention. In contrast, Cohen-Zion et al in their study utilizing community-dwelling elderly

individuals with high risk for SDB found no association of hypoxia indices with cognitive decline after

controlling for oximetry, sleep and subjective report. However, a significant association existed between

increases in daytime sleepiness and declining cognitive function. In addition, Lutsey et al utilizing a

subset of individuals from The Atherosclerosis Risk in Communities (ARIC) study, who participated in

the Sleep Heart Health Study (SHHS), examined the effect of OSA and cognitive decline over a 15-year

follow-up. Overall, this study found no evidence that OSA severity, degree of nocturnal hypoxemia, sleep

fragmentation, or habitual sleep duration were associated with subsequent cognitive decline. The

relationship between midlife OSA and later life cognition was also null.

Page 210: Omonigho A. Michael Bubu

198

Studies examining the relationship between OSA and Dementia outcomes tend to be more

consistent in their findings. Yaffe et al in their prospective study of SDB and cognition in elderly women

without dementia at baseline who were a substudy of the Study of Osteoporotic Fractures (SOF) followed

for approximately 5 years, found that elderly women had an 85% increased risk of developing

MCI/Dementia. In another study, Yaffe and her colleagues conducted a retrospective cohort study using

medical record data, and examined the relationship between a diagnosis of sleep disturbance and

dementia in elderly veterans. Sleep disturbance was significantly associated with an increased risk of

dementia and specifically, OSA demonstrated significant associations with increased risk of AD, Vascular

dementia and other dementias combined (frontotemporal dementia, senile dementia, or dementia not

otherwise specified). Furthermore, Chang et al in their retrospective matched-control cohort study that

utilized data from Taiwan’s Health Insurance Database estimated dementia risk in OSA versus non-OSA

patients in individuals 40 years and older, followed for 5 years. Results from the study showed a 70%

increased risk of developing dementia among OSA compared to non-OSA individuals. This study also

demonstrated gender-dependent, age-dependent and time-dependent associations of OSA and dementia.

OSA females were more like to develop dementia; OSA males aged 50-59 (middle-aged) had an

increased risk for developing dementia, while OSA females aged ≥ 70 years (elderly) had a greater risk of

developing dementia, and OSA patients were more likely to develop dementia in the first 2.5 years of

follow-up.

Summary, critique and future research directions

In middle-aged adults, cross-sectional studies mostly demonstrate that OSA is often associated

with cognitive decline. Longitudinal studies demonstrating that OSA can precede cognitive decline in

middle-aged adults are rare, however, the one study that examined this relationship, suggests so.

Nocturnal intermittent hypoxia and fragmented sleep could be culprits in cognitive and brain structural

deficits in middle-aged OSA patients. Studies utilizing brain-imaging techniques suggest that cognitive

impairment seen in middle-aged patients with moderate to severe OSA is associated with damage to brain

tissue in areas involved in several cognitive tasks including grey matter volume in the left hippocampus,

Page 211: Omonigho A. Michael Bubu

199

frontal structures of the brain and lateral temporal areas. CPAP treatment seems to be beneficial however

over a long-term as one study reported an almost complete reversal of white matter abnormalities over the

course of 12 months CPAP treatment.

In older adults and elderly, cross-sectional studies have not consistently demonstrated OSA-

Cognition associations and studies that identified such only revealed relatively weak associations. This

distinction and pattern in OSA-Cognition associations in middle-aged, older adults and elderly provides

the foundation for envisioning age-related modification of OSA-cognition relationships. Most of the

studies that examined this relationship in healthy community older adults revealed null associations. On

the contrary, studies examining this association in older adult OSA patients with MCI consistently

showed significant associations of OSA with cognition, suggesting that perhaps OSA in older adults with

MCI rather than in healthy older adults is the critical correlate of cognition in older adults. Longitudinal

studies in older adults and elderly, suggest that OSA at baseline may be a weak predictor of cognitive

decline. However, longitudinal studies examining the relationship between OSA and Dementia outcomes

produced consistent significant findings. This evidence that OSA can possibly precede MCI/AD and that

perhaps OSA in older adults with MCI rather than in healthy older adults is the critical correlate of

cognitive decline in older adults, may indicate that the presence and deposition of the AD biomarker, beta

amyloid, may be a moderator in these relationships. We consider this theme in the remaining sections.

Methodological differences existed among studies reviewed and can be rightly viewed as

limitations in the field of OSA and AD research. Issues related to the single assessment of OSA in

longitudinal studies, absent or incomplete CPAP intervention information during follow-up, and the

possibility that the etiological timeframe relevant for the association between OSA and AD could be

outside the examined period, variability in ways in which cognition was assessed, and issues relating to

selection bias, are all opportunities for future improvements. Many studies utilized clinic patients. It is

clear that the likelihood of clinic attendance in such participants is associated with disturbed sleep.

Therefore, when analyses are conducted on only such participants, biased results are likely the outcome.

In addition, not all studies accounted for the possible role of depression and its symptoms, which may be

Page 212: Omonigho A. Michael Bubu

200

important mediator or confounder of the association between disturbed sleep and cognition.134,135

Therefore, new research in the field should endeavor to separate causality relating to OSA, depression and

cognition.

Despite methodological strengths such as use of PSG sleep measures, and certain long follow-

ups, these were not all present in all studies, therefore limiting the strength of causal inferences that can

be made. Lastly, future studies should examine whether these associations are causal, focusing on the

mechanisms responsible for the somewhat different OSA effects seen at different ages or in different

populations.

OSA and Alzheimer’s disease (Cross-sectional studies)

Main findings

Studies examining any association between OSA and a diagnosis of AD were conducted in older

adults and the elderly since most AD diagnoses occur after age 65 (Tables 4.3 and 4.4). The scarcity of

epidemiological studies examining OSA and AD co-morbid associations was noteworthy. The five

studies included were conducted some 2 to 3 decades ago and had varying findings (Table 4.3). Two

studies136,137 demonstrated a significant association for OSA in AD patients relative to controls while

three of the studies had null associations. However, a recent meta-analysis of these studies concluded that

the aggregate odds ratio for OSA in AD vs. healthy control was 5.05 and homogeneous.85 Given the

cross-sectional nature of these analyses, the data cannot be interpreted as suggesting direction of causality

or temporality between OSA and AD. However, it does suggest the possibility of a reciprocal relationship

between these two disorders. OSA possibly facilitates progressive central nervous alterations and

excessive daytime sleepiness in patients and this further increases the risk of cognitive decline or AD

progression in older adults.86

Page 213: Omonigho A. Michael Bubu

201

OSA and AD Pathology

Main findings

Cross-sectional studies

Observational studies of OSA and AD pathology are vital in establishing links between OSA and

AD. If the relationship between OSA and AD is causal then demonstrating AD-specific neuropathology

resulting from increased disease burden in OSA patients can be vital. Cross-sectional studies examining

this association were conducted mostly in older adults and the elderly. We identified two studies

conducted in middle-aged participants (Table 4.4). A recent study by Yun et al138 examined whether OSA

increased brain amyloid burden relative to controls in middle-aged participants from the Korean Genome

and Epidemiology Study. After adjusting for potential confounders, OSA patients had a higher brain

amyloid burden in the right posterior cingulate gyrus and right temporal cortex, relative to controls,

suggesting that OSA may have been responsible for these findings. In this study, examination of regional

differences in cortical thickness were not significant. Bu et al.139 examined whether hypoxia indices in

OSA was associated with serum amyloid (Aβ40, Aβ42), total tau and phosphorylated tau 181 (P-tau 181)

comparing their levels with controls. Results from this study demonstrated significantly higher levels of

serum Aβ40, Aβ42, total Aβ and P-tau 181 in OSA patients compared to control. Hypoxia indices

including apnea-hypopnea index, the oxygen desaturation index, and the mean and lowest oxyhemoglobin

saturations correlated positively with Aβ40, Aβ42, and total Aβ in OSA patients, suggesting that hypoxia

possibly effects AD pathogenesis.

In older adults, all cross-sectional studies examined demonstrated associations between OSA and

AD pathology (Table 3). Osorio et al140 examined whether an association existed between SDB severity,

cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarkers, and the APOE alleles in cognitively

normal older adults. Findings from the study demonstrated an association between SDB and CSF

Page 214: Omonigho A. Michael Bubu

202

AD- biomarkers. They also found an association between intermittent hypoxia and increases in CSF T-

Tau, P-Tau and Aβ42 in ApoE3+, suggesting that hypoxia may be responsible for changes in the AD

biomarkers in this population of cognitive normal older adults. In ApoE4+ subjects, the study did not

detect significant SDB group differences for Aβ42, T-Tau or P-Tau, though SDB severity tended towards

lower CSF Aβ42 levels. However, participants were at varying stages of preclinical disease amongst the

ApoE4+ subjects. A recent study by Liguori et al141 compared CSF β-amyloid42, tau proteins, and lactate

levels in OSA versus CPAP treated OSA and controls. All participants had MCI. Findings from the study

demonstrated lower CSF β-amyloid42, higher lactate levels and t-tau/ Aβ42 ratio compared to controls

and CPAP treated OSA patients. Controls and CPAP treated OSA MCI patients had similar AD

biomarker levels. These findings suggest that OSA may effect early AD biomarker changes that may be

susceptible to CPAP treatment. Furthermore, in the elderly MCI patients, Spira et al showed that greater

SDB severity was associated with greater brain amyloid burden

Prospective/Longitudinal studies

Lutsey et al142 examined whether diagnosed OSA in the middle-aged was associated with adverse

morphological brain changes 15 years later. They utilized participants from the Atherosclerosis Risk in

Communities Study (ARIC). After accounting for body mass index in a series of multivariate models,

OSA at mid-life was not associated with cerebral markers of dementia or White Matter Hyper intensity

brain volume or regional brain volume changes. This study’s strength is its length of follow-up, since

dementia is known to have a long preclinical duration and middle-aged individuals may represent the

etiological relevant phase, similar to other dementia risk factors including diabetes,143-145 hypertension,146-

149 and smoking.145,150 Also, use of community based participants rather than small sample sized clinic

participants is also a plus. However, various methodological concerns could have been responsible for

their findings. Selection bias issues could have occurred from loss to follow-up as a third of participants

did not attend follow-up for neurocognitive assessments. The study had relatively few diagnosed severe

OSA patients, necessitating lumping of moderate and severe OSA patients together, which could have

Page 215: Omonigho A. Michael Bubu

203

attenuated any association that could potentially have occurred in severe OSA patients. The study also

did not account for CPAP use or treatment of OSA during the follow-up period. In contrast, Osorio et al14

using Alzheimer’s Disease Neuroimaging Initiative (ADNI) data examined whether SDB was associated

with an earlier onset age at MCI or AD. The study also determined whether CPAP treatment delayed MCI

or AD onset. Findings showed that in all population subsets examined, SDB patients had an earlier onset

age to MCI or AD-dementia. CPAP use also delayed the age of MCI onset. This study utilized a well-

defined longitudinal cohort; however, study limitations include the self-reported SDB and CPAP status.

Randomized Controlled Trials

The most effective treatment for OSA is continuous positive airway pressure (CPAP). With

CPAP treatment, OSA patients without dementia show an increase in slow wave sleep (SWS) and rapid

eye movement (REM) sleep.151-154 However, what is the effect of CPAP on sleep parameters and

cognition in AD patients with OSA? All four RCTs identified in this review included elderly participants

(mean age > 70) and reported significant improvements in deep sleep,155 mood,156 cognition,156,157 and

daytime sleepiness158 in OSA patients with AD (Table 4.4). For example, in a randomized placebo-

controlled trial, Cooke et al examined the effect of CPAP on sleep parameters in AD patients with OSA.

The researchers administered 3 weeks of CPAP treatment and compared the outcome with 3 weeks

placebo CPAP in patients with AD and OSA. Results from the study showed significant improvements in

SWS after one night with the improved effect extending for three weeks. Chong et al also examined the

effect of CPAP treatment on daytime sleepiness and found a significant reduction in patients with mild-

moderate AD with SDB after CPAP treatment. Examining the effects of CPAP on cognition in AD

patients, Ancoli-Israel et al157 compared CPAP treatment for three weeks versus placebo CPAP for three

weeks and found significant cognitive improvements. In addition, post-hoc analyses showed

improvements in episodic verbal learning and memory, executive functioning such as cognitive

flexibility, and mental processing speed.

Page 216: Omonigho A. Michael Bubu

204

A limitation identified in these RCTs include the issue of power to detect meaningful changes

across treatment arms. Some studies were underpowered and as such could not make definitive statements

specific to improvements within certain cognitive constructs. Other limitations include examination of

sleep parameters post-hoc while the study was powered for changes in cognition, inability to make causal

inferences due to non-random group assignment (continued use versus discontinuation of CPAP), limited

validation of ESS in elderly patients with AD, and issues with generalizability. Despite these limitations,

there is sufficient evidence to conclude that CPAP treatment may be effective in improving cognition in

OSA patients with AD.

Summary, critique and future research directions

In older adults and the elderly, cross-sectional studies suggest the possibility of a reciprocal relationship

between OSA and AD. The aggregate odds ratio in older adults, for OSA in AD vs. healthy control was

5.05 and homogeneous as demonstrated in a recent meta-analysis. OSA patients have an increased risk of

cognitive decline as shown in the above in the OSA and Cognition section, and cognitive decline

increases the risk for AD. Methodological concerns relating to these studies include the fact that

participants were mostly probable AD, because of the clinical and cognitive presentations of the enrolled

patients and the lack of reported neuroimaging or biomarkers data. Participants were also significantly

more educated which could have been responsible for the null findings seen in some studies. Higher

educational level and cognitive reserve are known protective risk factors for OSA induced cognitive

decline.86,159 Other studies failed to control for demographic and other possible confounders of the

association between OSA and AD. For example, Smallwood et al.160 failed to account for gender

differences in their sample. Women were significantly higher in their sample, which could have been

responsible for their null findings since studies suggest that OSA is more prevalent in men and increases

with age.161

In middle-aged, older adults and elderly individuals, cross-sectional data consistently suggests

that there is an association between OSA and preclinical-AD in the cognitively normal. Studies

consistently demonstrated OSA associations with AD biomarkers including higher levels of serum Aβ40,

Page 217: Omonigho A. Michael Bubu

205

Aβ42, total Aβ and P-tau 181 in OSA patients compared to control, with APOE possibly acting as an

effect modifier. Although determination of the causal relationships and directionality of these associations

is limited in these studies, it is possible that OSA increases the risk for preclinical AD or vice versa.

Prospective studies examining whether OSA accelerates amyloid deposition and effects regional

brain morphological changes that contribute to the development or progression of Alzheimer’s disease are

sparse in all age groups. The two prospective studies examined had contradictory findings. Both studies

also had their strengths and weaknesses (see section on OSA and AD Pathology/Prospective studies).

Therefore, future research is needed in the field to address the temporal nature of OSA-AD relationship

and the possible mechanisms underlying the relationship.

Evidence from RCTs provide an insight into the possibility of causal associations between OSA

and AD and are somewhat compelling. All RCTs were conducted in the elderly, and showed that CPAP

treatment not only improved sleep parameters in AD patients with OSA, such as improved SWS, and

excessive daytime sleepiness, it also increased cognitive function including areas such as episodic verbal

learning and memory, executive functioning such as cognitive flexibility, and mental processing speed.

Such compelling is the evidence such that effects were seen with a single night of CPAP treatment. These

findings clearly provides evidence that AD patients (particularly mild to moderate AD) with OSA can

benefit from CPAP treatment. It is also important to note that studies have shown that management of AD

with donepezil improved various hypoxia and sleep duration measures, suggesting that the cholinesterase

inhibitor improves subjective measures of OSA independently, 162,163 therefore highlighting the potential

reciprocal relationship between sleep and AD.

Future RCTs need to include dose-response studies stratified not only to the mild, moderate, and

severe categories of OSA, but also to include categories addressing duration of disease, extent of

intermittent hypoxemia extent, fragmented sleep severity, and presence of comorbidities. Issues with

design and sample size of double-blinded, placebo controlled clinical trials addressing the effect of CPAP

exist and need to be improved. Furthermore, as Pan et al164 noted in their review, non-inferiority trials

utilizing sleep apnea dental devices or other non-PAP therapeutics will also be beneficial.

Page 218: Omonigho A. Michael Bubu

206

DISCUSSION

Altogether, over three decades of research has investigated OSA-cognition, OSA-cognitive decline

and OSA-AD associations in the middle-aged, older adults and elderly. The data suggests the following:

- OSA is associated with greater cognitive deficits in middle-age adults than in older adults and the

elderly;

- OSA is associated with greater risk of subsequent cognitive decline in middle-aged adults than in

older adults and elderly;

- OSA in older adults with MCI rather than in healthy older adults may be the critical correlate of

cognition in older adults, therefore suggesting that presence or absence of amyloid burden might

act as a moderator in these relationships. This could be responsible for findings showing

increased amyloid deposition in older MCI patients with higher AHI but not in cognitive normal

controls165

- There is a link between OSA and AD biomarkers of neurodegeneration (e.g. Aβ40, Aβ42, total

Aβ and P-tau 181) and subsequent cognitive decline, in the middle-aged, older-adults and even in

elderly cognitive normal individuals.

We believe that this distinct pattern observed in OSA-Cognition and OSA-AD biomarker associations in

middle-aged, older adults and elderly provides the foundation for envisioning age-related modification of

OSA-cognition relationships.

A pertinent question arises from the findings: why are OSA-cognitive associations particularly

pronounced in the middle-aged and less so in older adults or the elderly? Several studies suggest that the

link between sleep and cognition weakens with increasing age because the aging brain is unable to

adequately and efficiently facilitate specific sleep supported cognitive processes.166-171 If this is true, then

it could have been responsible for the null or weaker results with cognition where associations were

identified in older adults and the elderly in this review. It also implies that improving duration and quality

of sleep in older adults and the elderly may not improve cognitive dysfunction because of diminished

neural plasticity, increased neuronal loss and atrophy, neuroendocrine changes, and nocturnal hypoxia

Page 219: Omonigho A. Michael Bubu

207

that occurs as one ages.172 These neurobiological changes seen in older adults and the elderly may also

compromise memory consolidation processes despite possible preservation of slow wave sleep amplitude

and density. Scullin and Bliwise in their review make the case that functional weakening of the brain in

their support of sleep-specific cognitive processes as we age, that hippocampal—neocortical

consolidation will not occur regardless of slow wave sleep quantity, if the hippocampus, neocortex, or

hippocampal—neocortical connections are greatly impaired in aging.170,173

Another pertinent question from the above is what mechanisms underlie the subsequent cognitive

decline in the middle-aged, even when no OSA-cognition associations are seen in older adults and the

elderly? Scullin and Bliwise highlight the possibility of disturbed sleep causing changes in sleep

modulated cognitive functions across the lifespan such that there is weakening of and/or compensation

attempts incited at sleep-cognition links. They note that studies demonstrate increases in insulin, cortisol,

blood pressure, proinflammatory cytokines and sympathetic tones, during acute or chronic sleep

restriction states,174,175 all of which possibly hasten cognitive aging.176 In addition, experimental studies

show that restriction of sleep in young animals can increase beta amyloid burden,177,178 and cause protein

misfolding,179 therefore compromising consolidation of memory.180,181 The prolonged accumulation over

time of beta amyloid and protein misfolding in the young and middle-aged further results in irreversible

neurodegeneration,182 thereby suggesting that sleep disturbances could possibly underlie subsequent

cognitive decline, even when no sleep-cognition associations are expressed in older adults and the

elderly.172

In middle-aged OSA individuals with cognitive deficits and particularly in OSA elderly AD

patients, randomized placebo controlled clinical trials provide promising findings of improvement of

cognitive deficits that range from attention and vigilance to memory and executive function. Although

CPAP improved hypoxia and other sleep measures relative to pre-CPAP indices, treated OSA patients

demonstrate comparable level of performance in certain areas such as working memory storage functions,

but still show deficits in other areas such as working memory.157,183 Furthermore, intricate

neuropsychological measures of attention, psychomotor speed and executive function tend to improve

Page 220: Omonigho A. Michael Bubu

208

only minimally since intermittent hypoxia upregulates death of neurons and hypertrophy of astrocytes in

the hippocampus and parietal cortex.184-186 The literature suggests that deprived sleep negatively affects

the prefrontal cortex (PFC) which largely controls executive function.187,188 Maillet and Rajah189 in their

review found that task performance matched between age groups yielded negative associations between

regional PFC and activity in older adults, however, the reverse was the case between volume and right

lateral PFC, when older adults performed worse than younger adults did. In addition, several studies

showed that in healthy older adults, PFC activity is positively related to grey matter volume in the medial

temporal lobe, but the reverse was observed in MCI and AD patients. In older adults, prefrontal sleep

spindles that facilitates hippocampal episodic learning are markedly reduced, and the increase in disturbed

sleep further compromises cognition.190,191 Therefore, OSA worsens metabolic injury192 that is particularly

resplendent in middle-aged, and exacerbates neuronal injury and facilitates memory and cognitive

impairment,193-197 that is particularly resplendent in older adults and the elderly.

Recent exciting trends in the literature point to a link between OSA and AD biomarkers of

neurodegeneration and subsequent cognitive decline, in the middle-aged, older-adults and even in elderly

cognitive normal individuals. This finding across our entire examined age spectrum poses a thought

provoking question as to why this is so. The data suggests that intermittent hypoxia and sleep

fragmentation are two main processes by which OSA induces neurodegenerative changes. Chronic

intermittent hypoxia, hypercapnia and hypertension in OSA can induce neuronal damage, including

axons,198 white matter,199 and reduced Diffusion tensor imaging based mean diffusivity in multiple brain

regions.200 Multiple studies also show grey matter loss in OSA patients compared to controls.119,201,202 As

a result of the pathophysiological effect of hypoxia precipitating hypertension,203-206 hypoperfusion,207-209

impaired glucose metabolism,210-212 and adverse cardiovascular,213,214 and metabolic consequences,215,216

beta amyloid deposition,217-220 and tau hyperphosphyrylation,212,221-224 ultimately these effects lead to

cognitive decline and progress to AD. In addition, inflammation,210,221-224 and oxidative stress,225,226

upregulate neurocognitive impairment in OSA and sustained OSA-cognitive dysfunction overlay with

that seen in AD-associated cognitive decline.82

Page 221: Omonigho A. Michael Bubu

209

CONCLUSION

OSA may be age-dependent in older adults (60 – 70 years old) and the elderly (70 years and

above) and is associated with neurodegenerative diseases particularly, cognitive decline and AD. In the

middle-aged (30 – 60 years old), the data suggests that OSA may be age-related and presents with a

distinct phenotype, with cardiovascular, and possibly metabolic effects. Intermittent hypoxia and sleep

fragmentation are two main processes by which OSA induces neurodegenerative changes.

References

1. Harada CN, Natelson Love MC, Triebel KL. Normal cognitive aging. Clinics in geriatric

medicine. 2013;29(4):737-752.

2. Hebert LE, Beckett LA, Scherr PA, Evans DA. Annual incidence of Alzheimer disease in the

United States projected to the years 2000 through 2050. Alzheimer disease and associated

disorders. 2001;15(4):169-173.

3. Hebert LE, Weuve J, Scherr PA, Evans DA. Alzheimer disease in the United States (2010-2050)

estimated using the 2010 census. Neurology. 2013;80(19):1778-1783.

4. Thies W, Bleiler L. 2013 Alzheimer's disease facts and figures. Alzheimer's & dementia : the

journal of the Alzheimer's Association. 2013;9(2):208-245.

5. Bubu OM, Brannick M, Mortimer J, et al. Sleep, Cognitive impairment, and Alzheimer's disease:

A Systematic Review and Meta-Analysis. Sleep. 2017;40(1).

6. McKinnon A, Terpening Z, Hickie IB, et al. Prevalence and predictors of poor sleep quality in

mild cognitive impairment. Journal of geriatric psychiatry and neurology. 2014;27(3):204-211.

7. Naismith SL, Hickie IB, Terpening Z, et al. Circadian misalignment and sleep disruption in mild

cognitive impairment. Journal of Alzheimer's disease : JAD. 2014;38(4):857-866.

Page 222: Omonigho A. Michael Bubu

210

8. Naismith SL, Rogers NL, Lewis SJ, et al. Sleep disturbance in mild cognitive impairment:

differential effects of current and remitted depression. Acta neuropsychiatrica. 2011;23(4):167-

172.

9. Gagnon K, Baril AA, Gagnon JF, et al. Cognitive impairment in obstructive sleep apnea.

Pathologie-biologie. 2014;62(5):233-240.

10. Naismith S, Winter V, Gotsopoulos H, Hickie I, Cistulli P. Neurobehavioral functioning in

obstructive sleep apnea: differential effects of sleep quality, hypoxemia and subjective sleepiness.

Journal of clinical and experimental neuropsychology. 2004;26(1):43-54.

11. Stranks EK, Crowe SF. The Cognitive Effects of Obstructive Sleep Apnea: An Updated Meta-

analysis. Archives of clinical neuropsychology : the official journal of the National Academy of

Neuropsychologists. 2016;31(2):186-193.

12. Vaessen TJ, Overeem S, Sitskoorn MM. Cognitive complaints in obstructive sleep apnea. Sleep

medicine reviews. 2015;19:51-58.

13. Chang WP, Liu ME, Chang WC, et al. Sleep apnea and the risk of dementia: a population-based

5-year follow-up study in taiwan. PLoS One. 2013;8(10):e78655.

14. Osorio RS, Gumb T, Pirraglia E, et al. Sleep-disordered breathing advances cognitive decline in

the elderly. Neurology. 2015;84(19):1964-1971.

15. Yaffe K, Laffan AM, Harrison SL, et al. Sleep-disordered breathing, hypoxia, and risk of mild

cognitive impairment and dementia in older women. JAMA : the journal of the American Medical

Association. 2011;306(6):613-619.

16. Epstein LJ, Kristo D, Strollo PJ, Jr., et al. Clinical guideline for the evaluation, management and

long-term care of obstructive sleep apnea in adults. Journal of clinical sleep medicine : JCSM :

official publication of the American Academy of Sleep Medicine. 2009;5(3):263-276.

17. Sateia MJ. International classification of sleep disorders-third edition: highlights and

modifications. Chest. 2014;146(5):1387-1394.

Page 223: Omonigho A. Michael Bubu

211

18. Arnardottir ES, Bjornsdottir E, Olafsdottir KA, Benediktsdottir B, Gislason T. Obstructive sleep

apnoea in the general population: highly prevalent but minimal symptoms. The European

respiratory journal. 2016;47(1):194-202.

19. Heinzer R, Vat S, Marques-Vidal P, et al. Prevalence of sleep-disordered breathing in the general

population: the HypnoLaus study. The Lancet Respiratory medicine. 2015;3(4):310-318.

20. Peppard PE, Young T, Barnet JH, Palta M, Hagen EW, Hla KM. Increased prevalence of sleep-

disordered breathing in adults. American journal of epidemiology. 2013;177(9):1006-1014.

21. Senaratna CV, Perret JL, Lodge CJ, et al. Prevalence of obstructive sleep apnea in the general

population: A systematic review. Sleep medicine reviews. 2016.

22. Bolona E, Hahn PY, Afessa B. Intensive care unit and hospital mortality in patients with

obstructive sleep apnea. Journal of critical care. 2015;30(1):178-180.

23. Gami AS, Olson EJ, Shen WK, et al. Obstructive sleep apnea and the risk of sudden cardiac

death: a longitudinal study of 10,701 adults. Journal of the American College of Cardiology.

2013;62(7):610-616.

24. Ge X, Han F, Huang Y, et al. Is obstructive sleep apnea associated with cardiovascular and all-

cause mortality? PLoS One. 2013;8(7):e69432.

25. Wang X, Ouyang Y, Wang Z, Zhao G, Liu L, Bi Y. Obstructive sleep apnea and risk of

cardiovascular disease and all-cause mortality: a meta-analysis of prospective cohort studies.

International journal of cardiology. 2013;169(3):207-214.

26. Won CH, Chun HJ, Chandra SM, Sarinas PS, Chitkara RK, Heidenreich PA. Severe obstructive

sleep apnea increases mortality in patients with ischemic heart disease and myocardial injury.

Sleep & breathing = Schlaf & Atmung. 2013;17(1):85-91.

27. Sullivan. F. Hidden health crisis costing America billions. Underdiagnosing and undertreating

obstructive sleep apnea draining healthcare system. . Darien, IL: American Academy of Sleep

Medicine, 2016.Available from: http://www.aasmnet.org/sleep-apnea-economic-impact.aspx.

Page 224: Omonigho A. Michael Bubu

212

28. Watson NF. Health Care Savings: The Economic Value of Diagnostic and Therapeutic Care for

Obstructive Sleep Apnea. Journal of clinical sleep medicine : JCSM : official publication of the

American Academy of Sleep Medicine. 2016;12(8):1075-1077.

29. Bliwise DL. EPIDEMIOLOGY OF AGE-DEPENDENCE IN SLEEP DISORDERED

BREATHING (SDB) IN OLD AGE: THE BAY AREA SLEEP COHORT (BASC). Sleep

medicine clinics. 2009;4(1):57-64.

30. Ancoli-Israel S, Klauber MR, Butters N, Parker L, Kripke DF. Dementia in institutionalized

elderly: relation to sleep apnea. Journal of the American Geriatrics Society. 1991;39(3):258-263.

31. Ancoli-Israel S, Klauber MR, Stepnowsky C, Estline E, Chinn A, Fell R. Sleep-disordered

breathing in African-American elderly. American journal of respiratory and critical care

medicine. 1995;152(6 Pt 1):1946-1949.

32. Ancoli-Israel S, Kripke DF, Klauber MR, Mason WJ, Fell R, Kaplan O. Sleep-disordered

breathing in community-dwelling elderly. Sleep. 1991;14(6):486-495.

33. Ancoli-Israel S, Kripke DF, Klauber MR, et al. Natural history of sleep disordered breathing in

community dwelling elderly. Sleep. 1993;16(8 Suppl):S25-29.

34. Duran J, Esnaola S, Rubio R, Iztueta A. Obstructive sleep apnea-hypopnea and related clinical

features in a population-based sample of subjects aged 30 to 70 yr. American journal of

respiratory and critical care medicine. 2001;163(3 Pt 1):685-689.

35. Malhotra A, Huang Y, Fogel R, et al. Aging influences on pharyngeal anatomy and physiology:

the predisposition to pharyngeal collapse. The American journal of medicine.

2006;119(1):72.e79-14.

36. Mehra R, Stone KL, Blackwell T, et al. Prevalence and correlates of sleep-disordered breathing in

older men: osteoporotic fractures in men sleep study. Journal of the American Geriatrics Society.

2007;55(9):1356-1364.

Page 225: Omonigho A. Michael Bubu

213

37. Young T, Palta M, Dempsey J, Skatrud J, Weber S, Badr S. The occurrence of sleep-disordered

breathing among middle-aged adults. The New England journal of medicine. 1993;328(17):1230-

1235.

38. Bliwise DNaIK, MH.; Roth, T.; Dement, WC., editors. . Principles and Practice of Sleep

Medicine. 4. Philadelphia: Saunders/Elsevier; 2005. . 2005:p. 24-38.

39. Young T. Age dependence of sleep disordered breathing. In: Kuna, ST.; Suratt, PM.; Remmers,

JE.,editors. . Sleep and Respiration in Aging Adults Elsevier; New York: 1991. 1991:p. 161-170.

40. Ancoli-Israel S, Kripke DF, Mason W, Messin S. Sleep apnea and nocturnal myoclonus in a

senior population. Sleep. 1981;4(4):349-358.

41. Bixler EO, Vgontzas AN, Ten Have T, Tyson K, Kales A. Effects of age on sleep apnea in men:

I. Prevalence and severity. American journal of respiratory and critical care medicine.

1998;157(1):144-148.

42. Ocasio-Tascon ME, Alicea-Colon E, Torres-Palacios A, Rodriguez-Cintron W. The veteran

population: one at high risk for sleep-disordered breathing. Sleep & breathing = Schlaf &

Atmung. 2006;10(2):70-75.

43. Phoha RL, Dickel MJ, Mosko SS. Preliminary longitudinal assessment of sleep in the elderly.

Sleep. 1990;13(5):425-429.

44. Stoohs RA, Gingold J, Cohrs S, Harter R, Finlayson E, Guilleminault C. Sleep-disordered

breathing and systemic hypertension in the older male. Journal of the American Geriatrics

Society. 1996;44(11):1295-1300.

45. Tishler PV, Larkin EK, Schluchter MD, Redline S. Incidence of sleep-disordered breathing in an

urban adult population: the relative importance of risk factors in the development of sleep-

disordered breathing. JAMA : the journal of the American Medical Association.

2003;289(17):2230-2237.

Page 226: Omonigho A. Michael Bubu

214

46. Zamarron C, Gude F, Otero Y, Alvarez JM, Golpe A, Rodriguez JR. Prevalence of sleep

disordered breathing and sleep apnea in 50- to 70-year-old individuals. A survey. Respiration;

international review of thoracic diseases. 1999;66(4):317-322.

47. Lavie P, Lavie L, Herer P. All-cause mortality in males with sleep apnoea syndrome: declining

mortality rates with age. The European respiratory journal. 2005;25(3):514-520.

48. Punjabi NM, Caffo BS, Goodwin JL, et al. Sleep-disordered breathing and mortality: a

prospective cohort study. PLoS medicine. 2009;6(8):e1000132.

49. Janssens JP, Pautex S, Hilleret H, Michel JP. Sleep disordered breathing in the elderly. Aging

(Milan, Italy). 2000;12(6):417-429.

50. Edwards BA WA, Sands SA, Owens RL, Eckert DJ, White DP, Malhotra A. . Obstructive sleep

apnea in older adults is a distinctly different physiological phenotype. . Sleep. 2014(37):1227–

1236. .

51. Dement WM, LE.; Bliwise, DL. . Physiological markers of aging: human sleep pattern changes.

In: Reff, ME.; Schneider, EL., editors. Biological Markers of Aging Washington, DC: US

Department of Health and Human Services; . Apr. 1982:p. 177-187.(NIH Publication 182-2221).

52. Kannel WH, H. . Vital capacity as a biomarker of aging. In: Reff, ME.; Schneider, EL., editors.

Biological Markers of Aging Washington, DC: US Department of Health and Human Services;

Apr. 1982: p. 145-160.(NIH Publication 182-2221).

53. Eckert DJ WD, Jordan AS, Malhotra A, Wellman A. . Defining phenotypic causes of obstructive

sleep apnea. Identification of novel therapeutic targets. . American journal of respiratory and

critical care medicine. 2013(188):996–1004. .

54. Wellman A ED, Jordan AS, Edwards BA, Passaglia CL, Jackson AC, Gautam S, Owens RL,

Malhotra A, White DP. . A method for measuring and modeling the physiological traits causing

obstructive sleep apnea. . J Appl Physiol 2011(110):1627–1637. .

Page 227: Omonigho A. Michael Bubu

215

55. Dhar S, Shastri SR, Lenora RA. Aging and the respiratory system. The Medical clinics of North

America. 1976;60(6):1121-1139.

56. Morris JF, Koski A, Johnson LC. Spirometric standards for healthy nonsmoking adults. The

American review of respiratory disease. 1971;103(1):57-67.

57. Smith WD, Cunningham DA, Patterson DH, Rechnitzer PA, Koval JJ. Forced expiratory volume,

height, and demispan in Canadian men and women aged 55-86. Journal of gerontology.

1992;47(2):M40-44.

58. Biessels GJ, Staekenborg S, Brunner E, Brayne C, Scheltens P. Risk of dementia in diabetes

mellitus: a systematic review. Lancet neurology. 2006;5(1):64-74.

59. Justin BN, Turek M, Hakim AM. Heart disease as a risk factor for dementia. Clinical

epidemiology. 2013;5:135-145.

60. Hla KM, Skatrud JB, Finn L, Palta M, Young T. The effect of correction of sleep-disordered

breathing on BP in untreated hypertension. Chest. 2002;122(4):1125-1132.

61. Hla KM, Young T, Finn L, Peppard PE, Szklo-Coxe M, Stubbs M. Longitudinal association of

sleep-disordered breathing and nondipping of nocturnal blood pressure in the Wisconsin Sleep

Cohort Study. Sleep. 2008;31(6):795-800.

62. Barone DA, Krieger AC. Stroke and obstructive sleep apnea: a review. Current atherosclerosis

reports. 2013;15(7):334.

63. Li M, Hou WS, Zhang XW, Tang ZY. Obstructive sleep apnea and risk of stroke: a meta-analysis

of prospective studies. International journal of cardiology. 2014;172(2):466-469.

64. Lipford MC, Flemming KD, Calvin AD, et al. Associations between Cardioembolic Stroke and

Obstructive Sleep Apnea. Sleep. 2015;38(11):1699-1705.

65. Einhorn D, Stewart DA, Erman MK, Gordon N, Philis-Tsimikas A, Casal E. Prevalence of sleep

apnea in a population of adults with type 2 diabetes mellitus. Endocrine practice : official journal

of the American College of Endocrinology and the American Association of Clinical

Endocrinologists. 2007;13(4):355-362.

Page 228: Omonigho A. Michael Bubu

216

66. Kent BD, McNicholas WT, Ryan S. Insulin resistance, glucose intolerance and diabetes mellitus

in obstructive sleep apnoea. Journal of thoracic disease. 2015;7(8):1343-1357.

67. Meslier N, Gagnadoux F, Giraud P, et al. Impaired glucose-insulin metabolism in males with

obstructive sleep apnoea syndrome. The European respiratory journal. 2003;22(1):156-160.

68. Wang X, Bi Y, Zhang Q, Pan F. Obstructive sleep apnoea and the risk of type 2 diabetes: a meta-

analysis of prospective cohort studies. Respirology (Carlton, Vic). 2013;18(1):140-146.

69. Haas DC, Foster GL, Nieto FJ, et al. Age-dependent associations between sleep-disordered

breathing and hypertension: importance of discriminating between systolic/diastolic hypertension

and isolated systolic hypertension in the Sleep Heart Health Study. Circulation. 2005;111(5):614-

621.

70. Newman AB, Nieto FJ, Guidry U, et al. Relation of sleep-disordered breathing to cardiovascular

disease risk factors: the Sleep Heart Health Study. American journal of epidemiology.

2001;154(1):50-59.

71. Nieto FJ, Herrington DM, Redline S, Benjamin EJ, Robbins JA. Sleep apnea and markers of

vascular endothelial function in a large community sample of older adults. American journal of

respiratory and critical care medicine. 2004;169(3):354-360.

72. Nieto FJ, Young TB, Lind BK, et al. Association of sleep-disordered breathing, sleep apnea, and

hypertension in a large community-based study. Sleep Heart Health Study. JAMA : the journal of

the American Medical Association. 2000;283(14):1829-1836.

73. Shahar E, Whitney CW, Redline S, et al. Sleep-disordered breathing and cardiovascular disease:

cross-sectional results of the Sleep Heart Health Study. American journal of respiratory and

critical care medicine. 2001;163(1):19-25.

74. Beebe DW, Groesz L, Wells C, Nichols A, McGee K. The neuropsychological effects of

obstructive sleep apnea: a meta-analysis of norm-referenced and case-controlled data. Sleep.

2003;26(3):298-307.

Page 229: Omonigho A. Michael Bubu

217

75. Bucks RS, Olaithe M, Eastwood P. Neurocognitive function in obstructive sleep apnoea: a meta-

review. Respirology (Carlton, Vic). 2013;18(1):61-70.

76. Wallace A, Bucks RS. Memory and obstructive sleep apnea: a meta-analysis. Sleep.

2013;36(2):203-220.

77. Martinez-Garcia MA, Campos-Rodriguez F, Soler-Cataluna JJ, Catalan-Serra P, Roman-Sanchez

P, Montserrat JM. Increased incidence of nonfatal cardiovascular events in stroke patients with

sleep apnoea: effect of CPAP treatment. The European respiratory journal. 2012;39(4):906-912.

78. Martinez-Garcia MA, Soler-Cataluna JJ, Ejarque-Martinez L, et al. Continuous positive airway

pressure treatment reduces mortality in patients with ischemic stroke and obstructive sleep apnea:

a 5-year follow-up study. American journal of respiratory and critical care medicine.

2009;180(1):36-41.

79. Munoz R, Duran-Cantolla J, Martinez-Vila E, et al. Severe sleep apnea and risk of ischemic

stroke in the elderly. Stroke. 2006;37(9):2317-2321.

80. Gottlieb DJ, Yenokyan G, Newman AB, et al. Prospective study of obstructive sleep apnea and

incident coronary heart disease and heart failure: the sleep heart health study. Circulation.

2010;122(4):352-360.

81. Arli B, Bilen S, Titiz AP, et al. Comparison of Cognitive Functions Between Obstructive Sleep

Apnea Syndrome and Simple Snoring Patients: OSAS May Be a Modifiable Risk Factor for

Cognitive Decline. Applied neuropsychology Adult. 2015;22(4):282-286.

82. Daulatzai MA. Evidence of neurodegeneration in obstructive sleep apnea: Relationship between

obstructive sleep apnea and cognitive dysfunction in the elderly. Journal of neuroscience

research. 2015;93(12):1778-1794.

83. Lutsey PL, Bengtson LG, Punjabi NM, et al. Obstructive Sleep Apnea and 15-Year Cognitive

Decline: The Atherosclerosis Risk in Communities (ARIC) Study. Sleep. 2016;39(2):309-316.

84. Osorio RS, Pirraglia E, Aguera-Ortiz LF, et al. Greater risk of Alzheimer's disease in older adults

with insomnia. Journal of the American Geriatrics Society. 2011;59(3):559-562.

Page 230: Omonigho A. Michael Bubu

218

85. Emamian F, Khazaie H, Tahmasian M, et al. The Association Between Obstructive Sleep Apnea

and Alzheimer's Disease: A Meta-Analysis Perspective. Frontiers in aging neuroscience.

2016;8:78.

86. Rosenzweig I, Glasser M, Polsek D, Leschziner GD, Williams SC, Morrell MJ. Sleep apnoea and

the brain: a complex relationship. The Lancet Respiratory medicine. 2015;3(5):404-414.

87. Davies CR, Harrington JJ. Impact of Obstructive Sleep Apnea on Neurocognitive Function and

Impact of Continuous Positive Air Pressure. Sleep medicine clinics. 2016;11(3):287-298.

88. Kerner NA, Roose SP. Obstructive Sleep Apnea is Linked to Depression and Cognitive

Impairment: Evidence and Potential Mechanisms. The American journal of geriatric psychiatry :

official journal of the American Association for Geriatric Psychiatry. 2016;24(6):496-508.

89. Shastri A, Bangar S, Holmes J. Obstructive sleep apnoea and dementia: is there a link?

International journal of geriatric psychiatry. 2016;31(4):400-405.

90. Aloia MS, Arnedt JT, Davis JD, Riggs RL, Byrd D. Neuropsychological sequelae of obstructive

sleep apnea-hypopnea syndrome: a critical review. Journal of the International

Neuropsychological Society : JINS. 2004;10(5):772-785.

91. Kilpinen R, Saunamaki T, Jehkonen M. Information processing speed in obstructive sleep apnea

syndrome: a review. Acta neurologica Scandinavica. 2014;129(4):209-218.

92. Cross N, Lampit A, Pye J, Grunstein RR, Marshall N, Naismith SL. Is Obstructive Sleep Apnoea

Related to Neuropsychological Function in Healthy Older Adults? A Systematic Review and

Meta-Analysis. Neuropsychology review. 2017.

93. Moher D, Liberati A, Tetzlaff J, Altman DG, Grp P. Preferred Reporting Items for Systematic

Reviews and Meta-Analyses: The PRISMA Statement. Journal of Clinical Epidemiology.

2009;62(10):1006-1012.

94. Wells G SB, O’Connell D, Peterson J,Welch V, Losos M, et al. . The Newcastle-Ottawa scale

(NOS) for assessing the quality of nonrandomised studies in metaanalyses,.

http://wwwohrica/programs/clinical_epidemiology/oxfordasp/; 2011 [accessed 112514]. 2011.

Page 231: Omonigho A. Michael Bubu

219

95. Bawden FC, Oliveira CA, Caramelli P. Impact of obstructive sleep apnea on cognitive

performance. Arquivos de neuro-psiquiatria. 2011;69(4):585-589.

96. Canessa N, Castronovo V, Cappa SF, et al. Obstructive sleep apnea: brain structural changes and

neurocognitive function before and after treatment. American journal of respiratory and critical

care medicine. 2011;183(10):1419-1426.

97. Castronovo V, Canessa N, Strambi LF, et al. Brain activation changes before and after PAP

treatment in obstructive sleep apnea. Sleep. 2009;32(9):1161-1172.

98. Castronovo V, Scifo P, Castellano A, et al. White matter integrity in obstructive sleep apnea

before and after treatment. Sleep. 2014;37(9):1465-1475.

99. Ferini-Strambi L, Baietto C, Di Gioia MR, et al. Cognitive dysfunction in patients with

obstructive sleep apnea (OSA): partial reversibility after continuous positive airway pressure

(CPAP). Brain research bulletin. 2003;61(1):87-92.

100. Kloepfer C, Riemann D, Nofzinger EA, et al. Memory before and after sleep in patients with

moderate obstructive sleep apnea. Journal of clinical sleep medicine : JCSM : official publication

of the American Academy of Sleep Medicine. 2009;5(6):540-548.

101. Naegele B, Launois SH, Mazza S, Feuerstein C, Pepin JL, Levy P. Which memory processes are

affected in patients with obstructive sleep apnea? An evaluation of 3 types of memory. Sleep.

2006;29(4):533-544.

102. Naegele B, Thouvard V, Pepin JL, et al. Deficits of cognitive executive functions in patients with

sleep apnea syndrome. Sleep. 1995;18(1):43-52.

103. Hrubos-Strom H, Nordhus IH, Einvik G, et al. Obstructive sleep apnea, verbal memory, and

executive function in a community-based high-risk population identified by the Berlin

Questionnaire Akershus Sleep Apnea Project. Sleep & breathing = Schlaf & Atmung.

2012;16(1):223-231.

104. Saunamaki T, Himanen SL, Polo O, Jehkonen M. Executive dysfunction in patients with

obstructive sleep apnea syndrome. European neurology. 2009;62(4):237-242.

Page 232: Omonigho A. Michael Bubu

220

105. Alchanatis M, Zias N, Deligiorgis N, et al. Comparison of cognitive performance among different

age groups in patients with obstructive sleep apnea. Sleep & breathing = Schlaf & Atmung.

2008;12(1):17-24.

106. Kim H, Dinges DF, Young T. Sleep-disordered breathing and psychomotor vigilance in a

community-based sample. Sleep. 2007;30(10):1309-1316.

107. Verstraeten E, Cluydts R, Pevernagie D, Hoffmann G. Executive function in sleep apnea:

controlling for attentional capacity in assessing executive attention. Sleep. 2004;27(4):685-693.

108. Cheshire K, Engleman H, Deary I, Shapiro C, Douglas NJ. Factors impairing daytime

performance in patients with sleep apnea/hypopnea syndrome. Archives of internal medicine.

1992;152(3):538-541.

109. Telakivi T, Kajaste S, Partinen M, Koskenvuo M, Salmi T, Kaprio J. Cognitive function in

middle-aged snorers and controls: role of excessive daytime somnolence and sleep-related

hypoxic events. Sleep. 1988;11(5):454-462.

110. Valencia-Flores M, Bliwise DL, Guilleminault C, Cilveti R, Clerk A. Cognitive function in

patients with sleep apnea after acute nocturnal nasal continuous positive airway pressure (CPAP)

treatment: sleepiness and hypoxemia effects. Journal of clinical and experimental

neuropsychology. 1996;18(2):197-210.

111. Findley LJ, Barth JT, Powers DC, Wilhoit SC, Boyd DG, Suratt PM. Cognitive impairment in

patients with obstructive sleep apnea and associated hypoxemia. Chest. 1986;90(5):686-690.

112. Greenberg GD, Watson RK, Deptula D. Neuropsychological dysfunction in sleep apnea. Sleep.

1987;10(3):254-262.

113. Roehrs T, Merrion M, Pedrosi B, Stepanski E, Zorick F, Roth T. Neuropsychological function in

obstructive sleep apnea syndrome (OSAS) compared to chronic obstructive pulmonary disease

(COPD). Sleep. 1995;18(5):382-388.

114. Bedard MA, Montplaisir J, Richer F, Malo J. Nocturnal hypoxemia as a determinant of vigilance

impairment in sleep apnea syndrome. Chest. 1991;100(2):367-370.

Page 233: Omonigho A. Michael Bubu

221

115. Bedard MA, Montplaisir J, Richer F, Rouleau I, Malo J. Obstructive sleep apnea syndrome:

pathogenesis of neuropsychological deficits. Journal of clinical and experimental

neuropsychology. 1991;13(6):950-964.

116. Bliwise DL. Sleep apnea and cognitive function: where do we stand now? Sleep. 1993;16(8

Suppl):S72-73.

117. Saunamaki T, Himanen SL, Polo O, Jehkonen M. Executive dysfunction and learning effect after

continuous positive airway pressure treatment in patients with obstructive sleep apnea syndrome.

European neurology. 2010;63(4):215-220.

118. Salorio CF, White DA, Piccirillo J, Duntley SP, Uhles ML. Learning, memory, and executive

control in individuals with obstructive sleep apnea syndrome. Journal of clinical and

experimental neuropsychology. 2002;24(1):93-100.

119. Torelli F, Moscufo N, Garreffa G, et al. Cognitive profile and brain morphological changes in

obstructive sleep apnea. Neuroimage. 2011;54(2):787-793.

120. Boland LL, Shahar E, Iber C, Knopman DS, Kuo TF, Nieto FJ. Measures of cognitive function in

persons with varying degrees of sleep-disordered breathing: the Sleep Heart Health Study.

Journal of sleep research. 2002;11(3):265-272.

121. Foley DJ, Masaki K, White L, Larkin EK, Monjan A, Redline S. Sleep-disordered breathing and

cognitive impairment in elderly Japanese-American men. Sleep. 2003;26(5):596-599.

122. Phillips BA, Berry DT, Schmitt FA, Magan LK, Gerhardstein DC, Cook YR. Sleep-disordered

breathing in the healthy elderly. Clinically significant? Chest. 1992;101(2):345-349.

123. Sforza E, Roche F, Thomas-Anterion C, et al. Cognitive function and sleep related breathing

disorders in a healthy elderly population: the SYNAPSE study. Sleep. 2010;33(4):515-521.

124. Dlugaj M, Weinreich G, Weimar C, et al. Sleep-disordered breathing, sleep quality, and mild

cognitive impairment in the general population. Journal of Alzheimer's disease : JAD.

2014;41(2):479-497.

Page 234: Omonigho A. Michael Bubu

222

125. Aloia MS, Ilniczky N, Di Dio P, Perlis ML, Greenblatt DW, Giles DE. Neuropsychological

changes and treatment compliance in older adults with sleep apnea. Journal of psychosomatic

research. 2003;54(1):71-76.

126. Ju G, Yoon IY, Lee SD, Kim TH, Choe JY, Kim KW. Effects of sleep apnea syndrome on

delayed memory and executive function in elderly adults. Journal of the American Geriatrics

Society. 2012;60(6):1099-1103.

127. Berry DT, Phillips BA, Cook YR, et al. Geriatric sleep apnea syndrome: a preliminary

description. Journal of gerontology. 1990;45(5):M169-174.

128. Hayward L, Mant A, Eyland A, et al. Sleep disordered breathing and cognitive function in a

retirement village population. Age and ageing. 1992;21(2):121-128.

129. Yesavage J, Bliwise D, Guilleminault C, Carskadon M, Dement W. Preliminary communication:

intellectual deficit and sleep-related respiratory disturbance in the elderly. Sleep. 1985;8(1):30-33.

130. Blackwell T, Yaffe K, Ancoli-Israel S, et al. Associations between sleep architecture and sleep-

disordered breathing and cognition in older community-dwelling men: the Osteoporotic Fractures

in Men Sleep Study. Journal of the American Geriatrics Society. 2011;59(12):2217-2225.

131. Terpening Z, Lewis SJ, Yee BJ, Grunstein RR, Hickie IB, Naismith SL. Association between

Sleep-Disordered Breathing and Neuropsychological Performance in Older Adults with Mild

Cognitive Impairment. Journal of Alzheimer's disease : JAD. 2015;46(1):157-165.

132. Kim SJ, Lee JH, Lee DY, Jhoo JH, Woo JI. Neurocognitive dysfunction associated with sleep

quality and sleep apnea in patients with mild cognitive impairment. The American journal of

geriatric psychiatry : official journal of the American Association for Geriatric Psychiatry.

2011;19(4):374-381.

133. Spira AP, Blackwell T, Stone KL, et al. Sleep-disordered breathing and cognition in older

women. Journal of the American Geriatrics Society. 2008;56(1):45-50.

Page 235: Omonigho A. Michael Bubu

223

134. Hahn EA, Wang HX, Andel R, Fratiglioni L. A change in sleep pattern may predict Alzheimer

disease. The American journal of geriatric psychiatry : official journal of the American

Association for Geriatric Psychiatry. 2014;22(11):1262-1271.

135. Lichtenberg PA, Ross T, Millis SR, Manning CA. The relationship between depression and

cognition in older adults: a cross-validation study. The journals of gerontology Series B,

Psychological sciences and social sciences. 1995;50(1):P25-p32.

136. Hoch CC, Reynolds CF, 3rd, Kupfer DJ, Houck PR, Berman SR, Stack JA. Sleep-disordered

breathing in normal and pathologic aging. The Journal of clinical psychiatry. 1986;47(10):499-

503.

137. Reynolds CF, 3rd, Kupfer DJ, Taska LS, et al. Sleep apnea in Alzheimer's dementia: correlation

with mental deterioration. The Journal of clinical psychiatry. 1985;46(7):257-261.

138. Yun CH, Lee HY, Lee SK, et al. Amyloid Burden in Obstructive Sleep Apnea. Journal of

Alzheimer's disease : JAD. 2017.

139. Bu XL, Liu YH, Wang QH, et al. Serum amyloid-beta levels are increased in patients with

obstructive sleep apnea syndrome. Scientific reports. 2015;5:13917.

140. Osorio RS, Ayappa I, Mantua J, et al. The interaction between sleep-disordered breathing and

apolipoprotein E genotype on cerebrospinal fluid biomarkers for Alzheimer's disease in

cognitively normal elderly individuals. Neurobiol Aging. 2014;35(6):1318-1324.

141. Liguori C, Mercuri NB, Izzi F, et al. Obstructive Sleep Apnea is Associated With Early but

Possibly Modifiable Alzheimer's Disease Biomarkers Changes. Sleep. 2017;40(5).

142. Lutsey PL, Norby FL, Gottesman RF, et al. Sleep Apnea, Sleep Duration and Brain MRI Markers

of Cerebral Vascular Disease and Alzheimer's Disease: The Atherosclerosis Risk in Communities

Study (ARIC). PLoS One. 2016;11(7):e0158758.

143. Elias PK, Elias MF, D'Agostino RB, et al. NIDDM and blood pressure as risk factors for poor

cognitive performance. The Framingham Study. Diabetes care. 1997;20(9):1388-1395.

Page 236: Omonigho A. Michael Bubu

224

144. Gregg EW, Yaffe K, Cauley JA, et al. Is diabetes associated with cognitive impairment and

cognitive decline among older women? Study of Osteoporotic Fractures Research Group.

Archives of internal medicine. 2000;160(2):174-180.

145. Rawlings AM, Sharrett AR, Schneider AL, et al. Diabetes in midlife and cognitive change over

20 years: a cohort study. Annals of internal medicine. 2014;161(11):785-793.

146. Gottesman RF, Schneider AL, Albert M, et al. Midlife hypertension and 20-year cognitive

change: the atherosclerosis risk in communities neurocognitive study. JAMA neurology.

2014;71(10):1218-1227.

147. Kantarci K, Weigand SD, Przybelski SA, et al. Risk of dementia in MCI: combined effect of

cerebrovascular disease, volumetric MRI, and 1H MRS. Neurology. 2009;72(17):1519-1525.

148. Pathan SS, Gottesman RF, Mosley TH, Knopman DS, Sharrett AR, Alonso A. Association of

lung function with cognitive decline and dementia: the Atherosclerosis Risk in Communities

(ARIC) Study. Eur J Neurol. 2011;18(6):888-898.

149. Staessen JA, Richart T, Birkenhager WH. Less atherosclerosis and lower blood pressure for a

meaningful life perspective with more brain. Hypertension. 2007;49(3):389-400.

150. Alonso A, Mosley TH, Jr., Gottesman RF, Catellier D, Sharrett AR, Coresh J. Risk of dementia

hospitalisation associated with cardiovascular risk factors in midlife and older age: the

Atherosclerosis Risk in Communities (ARIC) study. Journal of neurology, neurosurgery, and

psychiatry. 2009;80(11):1194-1201.

151. Aldrich M, Eiser A, Lee M, Shipley JE. Effects of continuous positive airway pressure on phasic

events of REM sleep in patients with obstructive sleep apnea. Sleep. 1989;12(5):413-419.

152. Issa FG, Sullivan CE. The immediate effects of nasal continuous positive airway pressure

treatment on sleep pattern in patients with obstructive sleep apnea syndrome.

Electroencephalography and clinical neurophysiology. 1986;63(1):10-17.

Page 237: Omonigho A. Michael Bubu

225

153. Loredo JS, Ancoli-Israel S, Kim EJ, Lim WJ, Dimsdale JE. Effect of continuous positive airway

pressure versus supplemental oxygen on sleep quality in obstructive sleep apnea: a placebo-

CPAP-controlled study. Sleep. 2006;29(4):564-571.

154. Verma A, Radtke RA, VanLandingham KE, King JH, Husain AM. Slow wave sleep rebound and

REM rebound following the first night of treatment with CPAP for sleep apnea: correlation with

subjective improvement in sleep quality. Sleep medicine. 2001;2(3):215-223.

155. Cooke JR, Ancoli-Israel S, Liu L, et al. Continuous positive airway pressure deepens sleep in

patients with Alzheimer's disease and obstructive sleep apnea. Sleep medicine. 2009;10(10):1101-

1106.

156. Cooke JR, Ayalon L, Palmer BW, et al. Sustained use of CPAP slows deterioration of cognition,

sleep, and mood in patients with Alzheimer's disease and obstructive sleep apnea: a preliminary

study. Journal of clinical sleep medicine : JCSM : official publication of the American Academy

of Sleep Medicine. 2009;5(4):305-309.

157. Ancoli-Israel S, Palmer BW, Cooke JR, et al. Cognitive effects of treating obstructive sleep apnea

in Alzheimer's disease: a randomized controlled study. Journal of the American Geriatrics

Society. 2008;56(11):2076-2081.

158. Chong MS, Ayalon L, Marler M, et al. Continuous positive airway pressure reduces subjective

daytime sleepiness in patients with mild to moderate Alzheimer's disease with sleep disordered

breathing. Journal of the American Geriatrics Society. 2006;54(5):777-781.

159. Harris P, Fernandez Suarez, M., Surace, E. I., Chrem Mendez, P., Martin, M.E., Clarens, M.F.,et

al. Cognitive reserve and Ab1-42 in mild cognitive impairment (Argentina-Alzheimer’sdisease

neuroimaging initiative). Neuropsychiatr DisTreat 2015(11):2599–2604.

160. Smallwood RG, Vitiello MV, Giblin EC, Prinz PN. Sleep apnea: relationship to age, sex, and

Alzheimer's dementia. Sleep. 1983;6(1):16-22.

161. Franklin KA, Lindberg E. Obstructive sleep apnea is a common disorder in the population-a

review on the epidemiology of sleep apnea. Journal of thoracic disease. 2015;7(8):1311-1322.

Page 238: Omonigho A. Michael Bubu

226

162. Moraes W, Poyares D, Sukys-Claudino L, Guilleminault C, Tufik S. Donepezil improves

obstructive sleep apnea in Alzheimer disease: a double-blind, placebo-controlled study. Chest.

2008;133(3):677-683.

163. Sukys-Claudino L, Moraes W, Guilleminault C, Tufik S, Poyares D. Beneficial effect of

donepezil on obstructive sleep apnea: a double-blind, placebo-controlled clinical trial. Sleep

medicine. 2012;13(3):290-296.

164. Pan W, Kastin AJ. Can sleep apnea cause Alzheimer's disease? Neuroscience and biobehavioral

reviews. 2014;47:656-669.

165. Spira AP, Yager C, Brandt J, et al. Objectively Measured Sleep and beta-amyloid Burden in

Older Adults: A Pilot Study. SAGE open medicine. 2014;2.

166. Edinger JD, Glenn DM, Bastian LA, Marsh GR. Slow-wave sleep and waking cognitive

performance II: Findings among middle-aged adults with and without insomnia complaints.

Physiology & behavior. 2000;70(1-2):127-134.

167. Kronholm E. Sleep in cognitive life-time trajectory. Sleep medicine. 2012;13(7):777-778.

168. McCrae CS, Vatthauer KE, Dzierzewski JM, Marsiske M. Habitual Sleep, Reasoning, and

Processing Speed in Older Adults with Sleep Complaints. Cognitive therapy and research.

2012;36(2):156-164.

169. Pace-Schott EF, Spencer RM. Age-related changes in the cognitive function of sleep. Progress in

brain research. 2011;191:75-89.

170. Pace-Schott EF, Spencer RM. Age-related changes in consolidation of perceptual and muscle-

based learning of motor skills. Frontiers in aging neuroscience. 2013;5:83.

171. Scullin MK. Sleep, memory, and aging: the link between slow-wave sleep and episodic memory

changes from younger to older adults. Psychology and aging. 2013;28(1):105-114.

172. Scullin MK, Bliwise DL. Sleep, cognition, and normal aging: integrating a half century of

multidisciplinary research. Perspectives on psychological science : a journal of the Association

for Psychological Science. 2015;10(1):97-137.

Page 239: Omonigho A. Michael Bubu

227

173. Grady C. The cognitive neuroscience of ageing. Nature reviews Neuroscience. 2012;13(7):491-

505.

174. McEwen BS. Sleep deprivation as a neurobiologic and physiologic stressor: Allostasis and

allostatic load. Metabolism: clinical and experimental. 2006;55(10 Suppl 2):S20-23.

175. Vgontzas AN, Zoumakis E, Bixler EO, et al. Adverse effects of modest sleep restriction on

sleepiness, performance, and inflammatory cytokines. The Journal of clinical endocrinology and

metabolism. 2004;89(5):2119-2126.

176. McEwen BS, Sapolsky RM. Stress and cognitive function. Current opinion in neurobiology.

1995;5(2):205-216.

177. Kang JE, Lim MM, Bateman RJ, et al. Amyloid-beta dynamics are regulated by orexin and the

sleep-wake cycle. Science (New York, NY). 2009;326(5955):1005-1007.

178. Xie L, Kang H, Xu Q, et al. Sleep drives metabolite clearance from the adult brain. Science (New

York, NY). 2013;342(6156):373-377.

179. Naidoo N, Ferber M, Master M, Zhu Y, Pack AI. Aging impairs the unfolded protein response to

sleep deprivation and leads to proapoptotic signaling. The Journal of neuroscience : the official

journal of the Society for Neuroscience. 2008;28(26):6539-6548.

180. Borlikova GG, Trejo M, Mably AJ, et al. Alzheimer brain-derived amyloid beta-protein impairs

synaptic remodeling and memory consolidation. Neurobiol Aging. 2013;34(5):1315-1327.

181. Freir DB, Fedriani R, Scully D, et al. Abeta oligomers inhibit synapse remodelling necessary for

memory consolidation. Neurobiol Aging. 2011;32(12):2211-2218.

182. Hita-Yanez E, Atienza M, Gil-Neciga E, Cantero JL. Disturbed sleep patterns in elders with mild

cognitive impairment: the role of memory decline and ApoE epsilon4 genotype. Current

Alzheimer research. 2012;9(3):290-297.

183. Lau EY, Eskes GA, Morrison DL, Rajda M, Spurr KF. Executive function in patients with

obstructive sleep apnea treated with continuous positive airway pressure. Journal of the

International Neuropsychological Society : JINS. 2010;16(6):1077-1088.

Page 240: Omonigho A. Michael Bubu

228

184. Aviles-Reyes RX, Angelo MF, Villarreal A, Rios H, Lazarowski A, Ramos AJ. Intermittent

hypoxia during sleep induces reactive gliosis and limited neuronal death in rats: implications for

sleep apnea. Journal of neurochemistry. 2010;112(4):854-869.

185. Daulatzai MA. Death by a thousand cuts in Alzheimer's disease: hypoxia--the prodrome.

Neurotoxicity research. 2013;24(2):216-243.

186. Daulatzai MA. Neurotoxic saboteurs: straws that break the hippo's (hippocampus) back drive

cognitive impairment and Alzheimer's Disease. Neurotoxicity research. 2013;24(3):407-459.

187. Drummond SP, Brown GG, Stricker JL, Buxton RB, Wong EC, Gillin JC. Sleep deprivation-

induced reduction in cortical functional response to serial subtraction. Neuroreport.

1999;10(18):3745-3748.

188. Nilsson JP, Soderstrom M, Karlsson AU, et al. Less effective executive functioning after one

night's sleep deprivation. Journal of sleep research. 2005;14(1):1-6.

189. Maillet D, Rajah MN. Association between prefrontal activity and volume change in prefrontal

and medial temporal lobes in aging and dementia: a review. Ageing research reviews.

2013;12(2):479-489.

190. Fandakova Y, Lindenberger U, Shing YL. Deficits in process-specific prefrontal and

hippocampal activations contribute to adult age differences in episodic memory interference.

Cerebral cortex (New York, NY : 1991). 2014;24(7):1832-1844.

191. Mander BA, Rao V, Lu B, et al. Impaired prefrontal sleep spindle regulation of hippocampal-

dependent learning in older adults. Cerebral cortex (New York, NY : 1991). 2014;24(12):3301-

3309.

192. Drager LF, Jun JC, Polotsky VY. Metabolic consequences of intermittent hypoxia: relevance to

obstructive sleep apnea. Best practice & research Clinical endocrinology & metabolism.

2010;24(5):843-851.

Page 241: Omonigho A. Michael Bubu

229

193. Ayalon L, Ancoli-Israel S, Aka AA, McKenna BS, Drummond SP. Relationship between

obstructive sleep apnea severity and brain activation during a sustained attention task. Sleep.

2009;32(3):373-381.

194. Beebe DW, Gozal D. Obstructive sleep apnea and the prefrontal cortex: towards a comprehensive

model linking nocturnal upper airway obstruction to daytime cognitive and behavioral deficits.

Journal of sleep research. 2002;11(1):1-16.

195. Jackson ML, Howard ME, Barnes M. Cognition and daytime functioning in sleep-related

breathing disorders. Progress in brain research. 2011;190:53-68.

196. Mander BA, Rao V, Lu B, et al. Prefrontal atrophy, disrupted NREM slow waves and impaired

hippocampal-dependent memory in aging. Nature neuroscience. 2013;16(3):357-364.

197. Sforza E, Roche F. Sleep apnea syndrome and cognition. Frontiers in neurology. 2012;3:87.

198. Ludemann P, Dziewas R, Soros P, Happe S, Frese A. Axonal polyneuropathy in obstructive sleep

apnoea. Journal of neurology, neurosurgery, and psychiatry. 2001;70(5):685-687.

199. Kiernan TE, Capampangan DJ, Hickey MG, Pearce LA, Aguilar MI. Sleep apnea and white

matter disease in hypertensive patients: a case series. The neurologist. 2011;17(5):289-291.

200. Kumar R, Chavez AS, Macey PM, Woo MA, Yan-Go FL, Harper RM. Altered global and

regional brain mean diffusivity in patients with obstructive sleep apnea. Journal of neuroscience

research. 2012;90(10):2043-2052.

201. Macey KE, Macey PM, Woo MA, et al. Inspiratory loading elicits aberrant fMRI signal changes

in obstructive sleep apnea. Respiratory physiology & neurobiology. 2006;151(1):44-60.

202. Macey PM, Kumar R, Woo MA, Valladares EM, Yan-Go FL, Harper RM. Brain structural

changes in obstructive sleep apnea. Sleep. 2008;31(7):967-977.

203. Morrell MJ, Finn L, Kim H, Peppard PE, Badr MS, Young T. Sleep fragmentation, awake blood

pressure, and sleep-disordered breathing in a population-based study. American journal of

respiratory and critical care medicine. 2000;162(6):2091-2096.

Page 242: Omonigho A. Michael Bubu

230

204. Peppard PE, Young T, Palta M, Skatrud J. Prospective study of the association between sleep-

disordered breathing and hypertension. The New England journal of medicine.

2000;342(19):1378-1384.

205. Somers VK, White DP, Amin R, et al. Sleep apnea and cardiovascular disease: an American

Heart Association/american College Of Cardiology Foundation Scientific Statement from the

American Heart Association Council for High Blood Pressure Research Professional Education

Committee, Council on Clinical Cardiology, Stroke Council, and Council On Cardiovascular

Nursing. In collaboration with the National Heart, Lung, and Blood Institute National Center on

Sleep Disorders Research (National Institutes of Health). Circulation. 2008;118(10):1080-1111.

206. Young T, Shahar E, Nieto FJ, et al. Predictors of sleep-disordered breathing in community-

dwelling adults: the Sleep Heart Health Study. Archives of internal medicine. 2002;162(8):893-

900.

207. Corfield DR, Meadows GE. Control of cerebral blood flow during sleep and the effects of

hypoxia. Advances in experimental medicine and biology. 2006;588:65-73.

208. Foster GE, Hanly PJ, Ostrowski M, Poulin MJ. Effects of continuous positive airway pressure on

cerebral vascular response to hypoxia in patients with obstructive sleep apnea. American journal

of respiratory and critical care medicine. 2007;175(7):720-725.

209. Furtner M, Staudacher M, Frauscher B, et al. Cerebral vasoreactivity decreases overnight in

severe obstructive sleep apnea syndrome: a study of cerebral hemodynamics. Sleep medicine.

2009;10(8):875-881.

210. Pallayova M, Steele KE, Magnuson TH, et al. Sleep apnea predicts distinct alterations in glucose

homeostasis and biomarkers in obese adults with normal and impaired glucose metabolism.

Cardiovascular diabetology. 2010;9:83.

211. Spiegel K, Knutson K, Leproult R, Tasali E, Van Cauter E. Sleep loss: a novel risk factor for

insulin resistance and Type 2 diabetes. Journal of applied physiology (Bethesda, Md : 1985).

2005;99(5):2008-2019.

Page 243: Omonigho A. Michael Bubu

231

212. Zizi F, Jean-Louis G, Brown CD, Ogedegbe G, Boutin-Foster C, McFarlane SI. Sleep duration

and the risk of diabetes mellitus: epidemiologic evidence and pathophysiologic insights. Current

diabetes reports. 2010;10(1):43-47.

213. Dobrowolski P, Klisiewicz A, Florczak E, et al. Independent association of obstructive sleep

apnea with left ventricular geometry and systolic function in resistant hypertension: the RESIST-

POL study. Sleep medicine. 2014;15(11):1302-1308.

214. Narkiewicz K, Somers VK. The sympathetic nervous system and obstructive sleep apnea:

implications for hypertension. Journal of hypertension. 1997;15(12 Pt 2):1613-1619.

215. Lurie A. Metabolic disorders associated with obstructive sleep apnea in adults. Advances in

cardiology. 2011;46:67-138.

216. Seetho IW, Wilding JP. Sleep-disordered breathing, type 2 diabetes and the metabolic syndrome.

Chronic respiratory disease. 2014;11(4):257-275.

217. Guglielmotto M, Aragno M, Autelli R, et al. The up-regulation of BACE1 mediated by hypoxia

and ischemic injury: role of oxidative stress and HIF1alpha. Journal of neurochemistry.

2009;108(4):1045-1056.

218. Guglielmotto M, Tamagno E, Danni O. Oxidative stress and hypoxia contribute to Alzheimer's

disease pathogenesis: two sides of the same coin. TheScientificWorldJournal. 2009;9:781-791.

219. Ogunshola OO, Antoniou X. Contribution of hypoxia to Alzheimer's disease: is HIF-1alpha a

mediator of neurodegeneration? Cellular and molecular life sciences : CMLS. 2009;66(22):3555-

3563.

220. Zhang X, Zhou K, Wang R, et al. Hypoxia-inducible factor 1alpha (HIF-1alpha)-mediated

hypoxia increases BACE1 expression and beta-amyloid generation. The Journal of biological

chemistry. 2007;282(15):10873-10880.

221. Daulatzai MA. Dysfunctional nucleus tractus solitarius: its crucial role in promoting

neuropathogenetic cascade of Alzheimer's dementia--a novel hypothesis. Neurochemical

research. 2012;37(4):846-868.

Page 244: Omonigho A. Michael Bubu

232

222. Daulatzai MA. Quintessential risk factors: their role in promoting cognitive dysfunction and

Alzheimer's disease. Neurochemical research. 2012;37(12):2627-2658.

223. Daulatzai MA. "Boomerang Neuropathology" of Late-Onset Alzheimer's Disease is Shrouded in

Harmful "BDDS": Breathing, Diet, Drinking, and Sleep During Aging. Neurotoxicity research.

2015;28(1):55-93.

224. Daulatzai MA. Olfactory dysfunction: its early temporal relationship and neural correlates in the

pathogenesis of Alzheimer's disease. Journal of neural transmission (Vienna, Austria : 1996).

2015;122(10):1475-1497.

225. Schild L, Reiser G. Oxidative stress is involved in the permeabilization of the inner membrane of

brain mitochondria exposed to hypoxia/reoxygenation and low micromolar Ca2+. The FEBS

journal. 2005;272(14):3593-3601.

226. Zhu Y, Fenik P, Zhan G, et al. Selective loss of catecholaminergic wake active neurons in a

murine sleep apnea model. The Journal of neuroscience : the official journal of the Society for

Neuroscience. 2007;27(37):10060-10071.

Page 245: Omonigho A. Michael Bubu

233

Table 4.1: Literature Search Terms for the Systematic Review of Obstructive Sleep Apnea, Cognition and Alzheimer’s disease Obstructive Sleep Apnea

Related

Cognition and Alzheimer's

Disease Related

Alzheimer's disease

pathology related

Sleep apnea Alzheimer's disease Dementia Amyloid beta OR Obstructive Sleep Apnea OR Cognitive Decline OR Amyloid beta

deposition/generation OR Obstructive Sleep Apnea Syndrome

OR Cognitive Impairment OR Alzheimer's disease pathogenesis

OR Sleep Disordered Breathing OR Mild Cognitive Impairment (MeSH)

OR Amyloid Precursor Protein

OR OSA OR MCI OR Amyloid cascade hypothesis

OR OSAH OR aMCI OR Inflammation OR Obstructive Sleep Apnea Hypopnea Syndrome

OR Cognition OR P-Tau

OR Respiratory Disturbance Index

OR Alzheimer (MeSH) OR Phosphorylated Tau

OR Apnea Hypopnea Index OR Cognitive function OR Hippocampal atrophy OR Hypoxia OR Neuropsychological

function OR Apolipoprotein E

OR Sleep Apnea Syndromes OR Executive function OR APOE4 OR Sleep Apnea, Obstructive

OR CSF-Tau

OR Sleep Apnea, Central

OR CSF-PTau OR Anoxia

Page 246: Omonigho A. Michael Bubu

234

Table 4.2: Descriptive Study Characteristics and Main Findings: Obstructive Sleep Apnea, Cognition, Cognitive Decline and Alzheimer’s disease (Cross-sectional studies)

Authors,

Year

Published

Study Design,

Setting (Study

Quality)

Subjects

Cognitive Domains Adjusted

Variables Major Findings Controls OSA+

Gender SDB Severity N Age N Age

Middle Aged (30-60)

Alchantis et al., 2008

Cross-sectional, sleep clinic

41 49 (33-63) 58 49 (32-

65) N/A

AHI: range 31-137 OSAS;

Range=1-7 NC

Reaction time Age, AHI,

SaO2min/mean, T90, BMI

OSA individual’s ≥ 50 had decreased reaction times. This effect was not seen in those <50.

Bawden et al., 2011

Cross-sectional 20 27.6 (8.7) 17 41.4

(13.0) N/A

AHI: 5-15/>15-30/

>30

Global function, attention, memory, executive function

Age, education OSA individuals had significantly impaired cognitive performance

Canessa et al., 2011

Controlled clinical trial

15 42.15 (6.64)

17 44 (7.63) All men AHI: ≥ 30

Memory, executive functions, attention,

constructional abilities, abstract

reasoning, vigilance, ESS

Age, education

▪ At baseline OSA assoc. with

↓neurocognitive function

▪OSA group had improved

neurocognitive function after 3 months

of CPAP ▪OSA assoc. with structural deficits

Castronovo et al., 2009

Controlled clinical trial

14 42.15 (6.64)

14 43.93 (7.78)

All men AHI > 30 Working memory, Brain activation

(fMRI) Age, education

▪OSA associated with over recruitment of

brain regions in working-memory task

▪Decreases in activation after

treatment

Page 247: Omonigho A. Michael Bubu

235

Castronovo et al., 2014

Prospective clinical study

15 42.15 (6.64)

13 43.23 (7.63)

All men AHI ≥ 30

Global function, memory, attention, vigilance, abstract reasoning, visuo-

spatial, verbal

Age, education

▪After 12 months of CPAP treatment, almost a complete reversal of white

matter abnormalities was seen ▪Significant

improvements in neuropsychological

function

Ferini-Strambi et al.,

2003

Case-control, sleep clinic

23 55.8 (5.4) 23 56.6 (6.1) 40M6F AHI:

Controls: <5, OSA: ≥5-40

Processing speed, language, executive

function, motor learning

Age, education

▪ 15 days of CPAP treatment returned

visuospatial and motor skills to normal ▪CPAP

did not improve executive function/

constructional abilities

Hrubos-Strom et al.,

2012

Cross-sectional, community

N/A N/A 290 48.2

(11.2) 162M 129F

AHI:<15/ ≥15 Memory, Executive

function Age, sex, education

▪Average oxygen saturation was the

indicator of obstructive sleep apnea severity most strongly assoc.

with cognitive function

Kim et al., 2007

Cross-sectional, community

N/A N/A 611 Range: 35-74

346 M 265 W

AHI: median=2.8; range=0-121

Vigilance

BMI, caffeine/alcohol

consumption, smoking status,

stimulants before testing

▪SDB related with diminished vigilant

attention performance

Kloepfer et al., 2009

Cross-sectional 20 47.4(5.6) 15 46.4 (5.9) 22M 13F

Clinical diagnosis:

AHI >5 and questionnaire

Memory, global Age, sex, IQ

▪Moderate OSA assoc. with impairment of

procedural and verbal declarative memory

Mathieu et al., 2008

Cross-sectional, sleep clinic

Younger: 12

Older: 18

Younger: 38.9 (2.2)

Older: 62.5 (1.8)

Younger: 14

Older: 14

Younger: 37.7 (2.0)

Older: 62.3 (2.0)

26 M 2 W

AHI: 51(4) OSAS

younger; 43(4) OSAS

older

Attention, Executive Function, Memory

Age, education

▪No Group-by-Age interaction for

cognitive performance. ▪Main effects for both

Group and Age

Page 248: Omonigho A. Michael Bubu

236

Naegele et al., 1995

Cross-sectional 17 49 (3) 17 49 (3) All men RDI > 10

Attentional capacity, memory efficiency, long-term memory, frontal lobe function

Age, education, verbal IQ

▪Memory deficits were associated with the

number of apneas and hypopneas per hour of sleep ▪Typical frontal-

lobe related abnormalities were

associated with levels of nocturnal hypoxemia

Naegele et al., 2006

Cross-sectional 95 47.4 95 48 144M46F RDI ≥ 10 Memory Age, education

▪OSA associated with impaired episodic memory, overall

procedural memory performance, specific

working memory capabilities ▪No impairment of maintenance,

recognition, or forgetfulness

Nikodemova et al., 2013

Cross-sectional, community

AHI <5 AHI 5-14 AHI ≥

15 Total

1,081 M F

AHI: <5/5-14/≥ 15

Memory, Executive function

Age, sex, education, BMI

▪No significant association between SDB and cognitive

performance in APOE4- ▪AHI ≥15, APOE4+ associated

with ↓cognitive function

n: 1,148 age: 52.1

(9.9)

n: 399 age: 56.4

(9.6)

n: 298 age: 56.6

n: 1,845 age: 53.9

(10.1) age range:

30-81

Quan et al., 2006

Cross-sectional, community

74 57.4(9.2) 67 59.4 (9.2) 84M 57F

AHI: ≤5

Attention and vigilance, processing

speed, executive function, motor

learning, visuospatial ability, memory

Age, sex, education

▪No significant impact of OSAH and

neuropsychological function ▪↑hypoxemia associated with ↓motor and processing speed

Page 249: Omonigho A. Michael Bubu

237

Salorio et al., 2002

Cross sectional 24 44.2(8.5) 28 44.0 (7.9) 34M 18F

Hypoxic episodes: 5-

20/21-35/>35 episodes per

hour

Long-term Learning and Memory,

Executive control Sex, education, IQ

▪OSA individuals exhibited poorer recall, less efficient semantic clustering, poorer use

of semantic cues ▪Other than letter

fluency, deficits were not observed in general

executive control

Saunamaki, Himanen, et

al., 2009

Controlled clinical trial

15 44

Range: 30-63

15 50

Range: 37-59

All men AHI:≤5/> 10 WAIS-R Age, education

▪OSAS associated with mild visually based

cognitive dysfunction and reduced amount of

sleep in the right hemisphere even after

CPAP

Saunamaki et al., 2010

Controlled clinical trial

17 44Range:

30-63 20

50 Range: 37-65

All men AHI:≤5/> 10

WAIS-R; Short term memory, working memory, verbal

fluency, visuomotor tracking, visuospatial

organization

Age, education, IQ

▪OSAS showed poorer performance on

executive tests ▪OSAS did not show any improvement on

executive or visuospatial function even after long-term

CPAP treatment

Saunamaki, Jehkonen et

al., 2009

Controlled clinical trial

20 43

Range: 29-63

40 47

Range: 28-65

All men AHI:≤5/> 10

WAIS-R; Verbally-based cognitive

abilities, visually-based cognitive

abilities,

Age, education

▪OSAS had lower set-shifting and

analysis/synthesis performance than controls ▪Based on

normative data, most had normal, while others had more serious deficits

Page 250: Omonigho A. Michael Bubu

238

Sharma et al., 2010

Cross-sectional 25 45.6 (6.2) 50 43 (7.5) 63M 12F

AHI > 30

Alertness, working memory, response inhibition, problem solving, executive

function

Age, sex, education

▪OSA subjects had significantly impaired performance on tests on alertness, working

memory, response inhibition, problem solving, executive

function ▪Significance disappeared after

adjusting for delayed information processing

Older Adults (61-70)

Aloia et al., 2003

Cross-sectional, clinic

Noncompliant Compliant

N/A

RDI: 51(20) OSAS

compliant; 46(22) OSAS noncompliant

Attention, Executive Function,

Construction, Motor Speed, Memory,

Language

Age, education, sleep apnea

severity

▪↑RDI related to↓ verbal delayed recall

memory ▪↑sleep fragmentation

and hypoxemia associated with↓ verbal delayed recall memory

▪Cognitive benefits with CPAP compliance

6 64.8 (2.6) 6 64.8 (6.4)

Berry et al., 1990

Cross-sectional, sleep clinic

12 68.4 (2.2) 8 68.6(4.8) Men only AHI: 28(12)

OSAS; 3(3)NC

Global, IQ, Memory ▪OSAS: ↓nonverbal IQ and nonverbal memory

delayed recall

Boland et al., 2002

Cross-sectional, community

N/A N/A 1700 62 (range

52-75) 837M923W

RDI median range: .4-23

Attention, Executive Function, Memory

age, education, occupation, field center, diabetes,

hypertension, body-mass index,

CNS meds, alcohol use

▪No relationships

Dlugaj et al., 2014

Cross- sectional, population-based

AHI <15 AHI 15-29 AHI ≥

30 Total

919M 874F

AHI; <15/15-29/ ≥ 30

Memory, executive function

Age, sex, education

▪SDB not associated with MCI or MCI

subtypes (amnestic and non-amnestic)

n: 1323 age:

62.94 (7.23)

n: 327 age: 65.79

(7.41)

n: 143 age:

62.17 (7.31)

n: 1793 age: 63.79

(7.45)

Page 251: Omonigho A. Michael Bubu

239

Foley et al., 2003

Cross-sectional, community

N/A N/A 718 Range: 79-97

Men only 71% had

AHI≥5; 19% had AHI ≥ 30

Global, Attention, Executive Function,

Construction, Memory, Language

Age, education, marital status

▪No relationships

Ju et al., 2012 Case-control, sleep

clinic 21 68.7 (5.5) 42 68(4.4)

37M 26F

AHI: Controls :< 15. OSA:

<15.3*

General cognitive/intellectual

ability, executive function

Age, sex, education, BMI

▪Significant findings with delayed recall and

executive function

Kim et al., 2011

Cross-sectional, clinic

N/A N/A 30 67.4 (3.8) 42M 18W

AHI: 13(12) MCI;15(14)

NC

Executive function, Language, Memory,

Visuospatial construction

▪↑AHI associated with

language with MCI

Lutsey et al., 2016

Cross-sectional 521 60.7 (5.1) 445 62(4.9) 435M 531W

AHI (<5/5- 14.9/15-

29.9/≥30) Global

Age, sex, field center, education,

alcohol intake, smoking, physical activity, APOE4, BMI, CRP, CVD

comorbidities

▪OSA category and additional indices of

sleep were not associated with change

in any cognitive test

Phillips et al., 1992

Cross-sectional, community

N/A N/A 92 64.2 (8.6) 44M 48W

AHI: 3(4)

Global, IQ, Attention, Executive Function, Memory, Language,

Motor

None ▪No relationship

between AHI and cognition

Sforza et al., 2010

Cross-sectional, community

382

Combined exposure

and controls: 68(1.8)

445

Combined exposure

and controls: 68(1.8)

343M 484W

AHI: 20(15); 53% had AHI≥15

Global, Attention, Executive Function, Memory, Language

gender, BMI, diabetic status, hypertension,

education level, anxiety and

depression scores, ESS, blood

pressure, self-reported sleep

time

▪No relationships

Terpening et al., 2015

Case-control(convenience

sample), community

N/A N/A 40 63.5 (8.9) 19M21F Processing speed ▪Findings significant**

Yesavage et al., 1985

Cross-sectional, sleep clinic

N/A N/A 41 69.5 (6.5) Men only RDI: 26(30);

73% had RDI>5

Attention, Executive Function, Memory

Age, depression, education, typical sleepiness/fatigue

▪RDI associated with attention and executive

function

Elderly (>70)

Page 252: Omonigho A. Michael Bubu

240

Blackwell et al., 2011

Cross-sectional, population-based

N/A N/A 2,909 76 (6) Only men AHI: <5/5-

14/≥ 30 Global, Executive

Function

Age, race, clinic, BMI, IADL, CVD

comorbidities, antidepressant

use, benzodiazepine use, depressive

symptoms, education, alcohol

use, smoking, physical activity,

self-reported health

▪↓ time in REM, ↑ time in Stage 1 sleep, and ↑nocturnal hypoxemia

are associated with poorer cognition

Hayward et al., 1992

Cross-sectional, community

N/A N/A 96 78 (3.9) 21 M 75 W

RDI: 6(6) Attention, Executive Function, Memory, Language, Motor

Age, education

▪RDI not associated with memory, verbal,

or motor factors ▪RDI associated with

cerebral efficiency factor

Spira et al., 2008

Cross-sectional, community

391 82.7 (3.3) 57 83.6 (4.3) Women

only AHI: <30/≥

30 Global, Executive

Function

Age, Education, SSRI (BMI and

functional impairment were found to not have

a significant impact on results)

▪↑AHI&↑hypoxemia& ↑central apnea associated with

cognition ▪APOE4+ associated

with 5x risk of cognitive impairment

Abbreviations: a, adjusted; Aβ40/42, amyloid beta-40/42; AD, Alzheimer’s disease; AFCFT, alphabetic fluency and category fluency tasks; AHI ≥ 15, apnea hypopnea index of 15 or more events per hour of sleep; APOE, apolipoprotein epsilon4; BVRT, Benton visual retention test; c, crude; CAR, circadian activity rhythms; CASI, cognitive abilities screening instrument; CERAD, consortium to establish a registry for Alzheimer’s disease; CPRS, comprehensive psychopathological rating scale; CSF, cerebrospinal fluid; CVLT, California verbal learning test; DSM-IIIR/IV-TR, diagnostic and statistical manual of mental disorders; third edition/fourth edition, text revised; EDS, Excessive daytime sleepiness; ESS, Epworth sleepiness scale; GBSRT, Grober and Buschke selective reminding test; HAM-D, Hamilton Depression Rating Scale; HR, hazard ratio; ICD-9/10, international classification of diseases ninth/tenth edition AD criteria; IQCODE, informant questionnaire on cognitive decline in the elderly; MMSE, mini mental state examination; MRI, magnetic resonance imaging; N, number of participants; NA, not applicable; N/A, not available; NINCDS-ADRDA, national institute of neurological and communicative disorders and stroke and the Alzheimer’s disease and related disorders association; NINDS-AIREN, national institute of neurological disorders and stroke and association internationale pour la recherche et l’enseignement en neurosciences vascular dementia criteria; OR, odds ratio; PSG, polysomnography; PSQI, Pittsburgh sleep quality index; P-tau, phosphorylated tau; RBANS, repeatable battery for the assessment of neuropsychological status; RR, relative risk; SDB, sleep disordered breathing; SPMSQ, short portable mental status questionnaire; TONI, test of nonverbal intelligence; Trails B, trail making b test; TST, total sleep time; T-tau, total-tau; WAIS-III, Wechsler adult intelligence scale third version; WASO, wake after sleep onset; WHIIRS, women’s health initiative insomnia rating scale; WMS-R, Wechsler memory scale-revision; w/o, without.

Page 253: Omonigho A. Michael Bubu

241

Table 4.3: Descriptive Study Characteristics and Main Findings: Obstructive Sleep Apnea, Cognition, Cognitive Decline and Alzheimer’s disease (Longitudinal studies – older adults mainly)

Authors,

Year

Published

Study

design,

setting

(study

quality)

Subjects

Cognitive

Domains Adjusted Variables Major Findings Controls OSA+

Gender SDB

Severity N Age N Age

Blackwell et al., 2015

Longitudinal, community

based 1,504

75.8 (5.3)

1,132 76.4 (5.2)

Men only

ODI: <15, ≥15

AHI: <15, ≥15)

Global

Age, site, race, BMI, education, depressive

symptoms, CVD comorbidities, Parkinson's

disease, IADL, benzodiazepine use,

antidepressant use, self-reported health, physical activity, alcohol intake,

smoking

▪No significant association between AHI and cognitive decline in older community-

dwelling men ▪Modest association of

nocturnal hypoxemia with global cognitive decline

Chang et al., 2013

Longitudinal, community

based 7,070

55.5 (4.78)

1,414 55.5

(4.93)

M: 5,034

F: 3,450

Not specified; clinical

diagnosis (according to

the guidelines of the AASM)

Dementia diagnosis

Age, sex, CVD comorbidities, urbanization

level, income

▪ Hazard ratio of dementia is 1.44 times greater for patients

with SA than for the comparison group

▪SA is an age, time, and gender dependent risk factor for

dementia

Cohen-Zion et al., 2001

Longitudinal, Community

N/A N/A 46 80(3) 7M

39 W RDI: 10(9) Global Age, education

▪↑EDS associated with global cognitive decline over 3 years

follow-up ▪Hypoxemia and RDI not associated with cognition

Lutsey et al., 2016

Longitudinal 521 60.7 (5.1)

445 62(4.9) 435M 531W

AHI: <5/5- 14.9/ 15-29.9/ ≥30

Global

Age, sex, field center, education, alcohol intake, smoking, physical activity, APOE4, BMI, CRP, CVD

comorbidities

▪OSA category and additional indices of sleep were not

associated with change in any cognitive test

N/A N/A 599 67.0 (1) Global

Page 254: Omonigho A. Michael Bubu

242

Martin et al., 2015

Longitudinal, population

based

224M 335W

AHI: <15/ 15-30/ >30)

Age, sex, education, follow-up length, BMI, ESS, CVD

comorbidities, anxiety, depression

▪Abnormal breathing events associated slightly with decline

in attentional domain ▪No association with changes in executive and memory function

Yaffe et al., 2011

Longitudinal, Community

193 82.1(3.2) 105 82.6(3.1) Women

only AHI: ≥15

Global, Attention, Executive Function, Memory

Age, race, BMI, education, smoking, diabetes,

hypertension, antidepressant use, benzodiazepine use,

non-diazepam anxiolytics use

▪SDB: ↑hypoxemia had ↑risk of developing MCI or dementia over 5 year follow-up▪Sleep

fragmentation and duration not associated with cognition

Yaffe et al., 2015

Longitudinal ###### 68.5 13,480 66.9 Men Only

Not specified; clinical

diagnosis

Dementia (classified using

International

Classification of

Disease, Ninth Revision codes)

Age, CVD comorbidities, obesity, depression, income,

education

▪Those with a sleep disturbance had a 27% increased risk for

dementia (HR: 1.27)

Abbreviations: a, adjusted; Aβ40/42, amyloid beta-40/42; AD, Alzheimer’s disease; AFCFT, alphabetic fluency and category fluency tasks; AHI ≥ 15, apnea hypopnea index of 15 or more events per hour of sleep; APOE, apolipoprotein epsilon4; BVRT, Benton visual retention test; c, crude; CAR, circadian activity rhythms; CASI, cognitive abilities screening instrument; CERAD, consortium to establish a registry for Alzheimer’s disease; CPRS, comprehensive psychopathological rating scale; CSF, cerebrospinal fluid; CVLT, California verbal learning test; DSM-IIIR/IV-TR, diagnostic and statistical manual of mental disorders; third edition/fourth edition, text revised; EDS, Excessive daytime sleepiness; ESS, Epworth sleepiness scale; GBSRT, Grober and Buschke selective reminding test; HAM-D, Hamilton Depression Rating Scale; HR, hazard ratio; ICD-9/10, international classification of diseases ninth/tenth edition AD criteria; IQCODE, informant questionnaire on cognitive decline in the elderly; MMSE, mini mental state examination; MRI, magnetic resonance imaging; N, number of participants; NA, not applicable; N/A, not available; NINCDS-ADRDA, national institute of neurological and communicative disorders and stroke and the Alzheimer’s disease and related disorders association; NINDS-AIREN, national institute of neurological disorders and stroke and association internationale pour la recherche et l’enseignement en neurosciences vascular dementia criteria; OR, odds ratio; PSG, polysomnography; PSQI, Pittsburgh sleep quality index; P-tau, phosphorylated tau; RBANS, repeatable battery for the assessment of neuropsychological status; RR, relative risk; SDB, sleep disordered breathing; SPMSQ, short portable mental status questionnaire; TONI, test of nonverbal intelligence; Trails B, trail making b test; TST, total sleep time; T-tau, total-tau; WAIS-III, Wechsler adult intelligence scale third version; WASO, wake after sleep onset; WHIIRS, women’s health initiative insomnia rating scale; WMS-R, Wechsler memory scale-revision; w/o, without.

Page 255: Omonigho A. Michael Bubu

243

Table 4.4 Descriptive Study Characteristics and Main Findings: Obstructive Sleep Apnea, and Alzheimer’s disease/AD Pathology

Authors, Year

Published

Study

Design

Subjects

OSA

assessment

Alzheimer's disease

assessment Adjusted Variables Major findings

N

Age

(Mean ±

SD)

Gender

OSA and AD

Hoch et al., 1986

Cross-sectional

80 71.5 (8.1) 33 M 57 F

AHI DSM-III None Significant association

Hoch et al., 1989

Cross-sectional

27 74.5 (5.1) 7 M 20 F

AHI NICNDS-ADRDA,

DSM-III N/A

No association between OSA and dementia

Reynolds et al., 1985

Cross-sectional

61 69.7 (6.8) 19 M 42 F

AI, AHI

DSM 3, Hamilton rating, Folstein

score, and a modified Hachinski

Ischemia score

Gender Significant association between

sleep apnea and dementia in women

Reynolds et al.,1987

Cross-sectional

30 73.3 (9.1) 3 M 12 F

24 Chanel polygraphs

DSM 3, Hamilton rating, Folstein

score, and a modified Hachinski

Ischemia score

N/A No association between OSA

and dementia

Smallwood et al., 1983

Cross-sectional

55 Range: 23-

81 years 45 M 10 F

AHI DSM 3, neurological

examination Age, sex

No relationship between

dementia and apnea severity

OSA and AD pathology

Bu et al., 2015 Cross-

sectional 94

43.62 (9.78)

67M 27F

AHI, ODI, MSaO2, LSaO2

Amyloid beta levels Age, sex Significant association between

hypoxia and amyloid levels

Ligouri et al., 2017

Cross-sectional

50 66.96 (7.98)

33M 17F

AHI

CSI classified by subjective memory decline, cognitive

performance

Age, education Significant association between OSA and CSF AD biomarkers

Page 256: Omonigho A. Michael Bubu

244

Lutsey et al., 2017

Prospective 312 61.7 (5.0) 145M 167F

AHI, SHHS Sleep Habits

Questionnaire

Neurocognitive exam, brain MRI

age, sex, field center, education, physical

activity, ethanol intake, smoking status, leusire time physical activity,

and APOE e4 risk allele, BMI

No relationship between mid-life OSA and dementia

Osorio et al., 2014

Cross-sectional

95 67.6 37M58F AHI Neuropsychological

test battery

Age, BMI, CSF internal batch, time interval

between sleep study and lumbar puncture,ApoE4

status

Significant association between SDB and AD CSF biomarkers

Osorio et al., 2015

Prospective 2,285 Self reported Self-report; diagnosis by

clinician

APOE e4 status, sex, education, BMI,

depression, cardiovascular disease, hypertension, diabetes,

and age

Significant association between SDB and earlier age at

cognitive decline

Yun et al., 2017

Cross-sectional

38 56.7 (4.0) M: 18 F: 20

AHI Neuropsychological

test battery

Age, sex, education, APOE genotype, status

of sleep duration, hypertension, diabetes,

BMI, exercise, depressive mood,

smoking, and alcohol drinking

Significant association between OSA and amyloid deposition

Random Control Trials

Ancoli-Israel et al., 2008

RCT 52 78.2 (7.2) 39M 13F

Rechtschaffen and Kales

criteria

Nueropsychological test battery

None CPAP improved some cognitive functioning

Chong et al., 2006

RCT 39 78.0 (7.04) 29M 10F

RDI NINCDS-ADRDA

criteria None

CPAP reduces sleepiness in those with AD and OSA

Cooke et al., 2009a

RCT 52 77.8 (7.3) 39M 13F

Rechtschaffen and Kales

criteria

NINCDS-ADRDA criteria, MMSE

None After one night of tCPAP use: deeper sleep, affects for three

weeks

Cooke et al., 2009b

RCT 10 75.7 (5.9) 7M 3F

AHI, PSQI, ESS, FOSQ

Neuropsychological test battery

None Sustained CPAP use associated

with less cognitive decline

Page 257: Omonigho A. Michael Bubu

245

Spira et al., 2014

RCT 13 71.6 (7.8) 7M 6F

AHI, ODI Neuropsychological

tests, GDS, CDR None

SDB severity associated with amyloid deposition

Abbreviations: a, adjusted; Aβ40/42, amyloid beta-40/42; AD, Alzheimer’s disease; AFCFT, alphabetic fluency and category fluency tasks; AHI ≥ 15, apnea hypopnea index of 15 or more events per hour of sleep; APOE, apolipoprotein epsilon4; BVRT, Benton visual retention test; c, crude; CAR, circadian activity rhythms; CASI, cognitive abilities screening instrument; CERAD, consortium to establish a registry for Alzheimer’s disease; CPRS, comprehensive psychopathological rating scale; CSF, cerebrospinal fluid; CVLT, California verbal learning test; DSM-IIIR/IV-TR, diagnostic and statistical manual of mental disorders; third edition/fourth edition, text revised; EDS, Excessive daytime sleepiness; ESS, Epworth sleepiness scale; GBSRT, Grober and Buschke selective reminding test; HAM-D, Hamilton Depression Rating Scale; HR, hazard ratio; ICD-9/10, international classification of diseases ninth/tenth edition AD criteria; IQCODE, informant questionnaire on cognitive decline in the elderly; MMSE, mini mental state examination; MRI, magnetic resonance imaging; N, number of participants; NA, not applicable; N/A, not available; NINCDS-ADRDA, national institute of neurological and communicative disorders and stroke and the Alzheimer’s disease and related disorders association; NINDS-AIREN, national institute of neurological disorders and stroke and association internationale pour la recherche et l’enseignement en neurosciences vascular dementia criteria; OR, odds ratio; PSG, polysomnography; PSQI, Pittsburgh sleep quality index; P-tau, phosphorylated tau; RBANS, repeatable battery for the assessment of neuropsychological status; RR, relative risk; SDB, sleep disordered breathing; SPMSQ, short portable mental status questionnaire; TONI, test of nonverbal intelligence; Trails B, trail making b test; TST, total sleep time; T-tau, total-tau; WAIS-III, Wechsler adult intelligence scale third version; WASO, wake after sleep onset; WHIIRS, women’s health initiative insomnia rating scale; WMS-R, Wechsler memory scale-revision; w/o, without.

Page 258: Omonigho A. Michael Bubu

246

Figure 4.1: Study retrieval and selection for Obstructive Sleep Apnea, Cognitive Decline and/or Alzheimer’s disease Review

Studies screened for titles (n = 1,688)

Studies excluded (n =784)

Full-text articles assessed for eligibility

(n =193)

Full-text articles excluded, with reasons (n = 141)

- Review article (n = 15) - OSA not assessed (n =

32 ) - Relationship not

studied (n = 40) - Not peer reviewed (n =

15 ) - Alzheimer not

examined (n = 24) - Duplicate studies (n =

15) Studies included in

systematic review (n = 57)

Article identified from reference

search (n = 5)

Studies screened for abstracts

(n = 904)

Studies excluded (n = 711)

Duplicates removed (n =1029)

Studies identified through database searching (n = 2717)

(PubMed=1,112, Embase = 1,164, Web of Science = 285, Psych Info = 103, Cochrane library = 53

Studies after duplicates removed (n = 1,688)

Ide

nti

fica

tio

n

Scr

ee

nin

g

Elig

ibilit

y

Incl

ud

ed

Page 259: Omonigho A. Michael Bubu
Page 260: Omonigho A. Michael Bubu

248

Page 261: Omonigho A. Michael Bubu

249

Page 262: Omonigho A. Michael Bubu

250

APPENDIX C

IRB LETTER

10/20/2017 Omonigho Bubu, MD, MPH Epidemiology and Biostatistics 4202 East Fowler Ave. Tampa, FL 33612 RE: Not Human Subjects Research Determination IRB#: Pro00032613 Title: Obstructive Sleep Apnea and the risk of Alzheimer’s disease Dear Dr. Bubu: The Institutional Review Board (IRB) has reviewed your application. The activities presented in the application involve methods of program evaluation, quality improvement, and/or needs analysis. While potentially informative to others outside of the university community, study results would not appear to contribute to generalizable knowledge. As such, the activities do not meet the definition of human subject research under USF IRB policy, and USF IRB approval and oversight are therefore not required. While not requiring USF IRB approval and oversight, your study activities should be conducted in a manner that is consistent with the ethical principles of your profession. If the scope of your project changes in the future, please contact the IRB for further guidance. If you will be obtaining consent to conduct your study activities, please remove any references to "research" and do not include the assigned Protocol Number or USF IRB contact information. If your study activities involve collection or use of health information, please note that there may be requirements under the HIPAA Privacy Rule that apply. For further information, please

Page 263: Omonigho A. Michael Bubu

251

contact a HIPAA Program administrator at (813) 974-5638. Sincerely, E. Verena Jorgensen, M.D., Chairperson USF Institutional Review Board

Page 264: Omonigho A. Michael Bubu

252

APPENDIX D

Page 265: Omonigho A. Michael Bubu

253

Page 266: Omonigho A. Michael Bubu

254

Page 267: Omonigho A. Michael Bubu

255

Page 268: Omonigho A. Michael Bubu

256

Page 269: Omonigho A. Michael Bubu

257

Page 270: Omonigho A. Michael Bubu

258

Page 271: Omonigho A. Michael Bubu

259

Page 272: Omonigho A. Michael Bubu

260

Page 273: Omonigho A. Michael Bubu

261

Page 274: Omonigho A. Michael Bubu

262

Page 275: Omonigho A. Michael Bubu

263

Page 276: Omonigho A. Michael Bubu

264

Page 277: Omonigho A. Michael Bubu

265

Page 278: Omonigho A. Michael Bubu

266

Page 279: Omonigho A. Michael Bubu

267

Page 280: Omonigho A. Michael Bubu

268

Page 281: Omonigho A. Michael Bubu

269