Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Curr Psychiatry Rep (2015) 17:97

DOI 10.1007/s11920-015-0631-8

SLEEP DISORDERS (P GEHRMAN, SECTION EDITOR)

Sleep, Cognition and Dementia


Verna R. Porter 1 & William G. Buxton 2 & Alon Y. Avidan 3

# Springer Science+Business Media New York (outside the USA) 2015

Abstract The older patient population is growing rapidly neurodegenerative diseases, the Bsynucleinopathies^, and re-
around the world and in the USA. Almost half of seniors over quires formal polysomnographic evaluation. Untreated sleep
age 65 who live at home are dissatisfied with their sleep, and disorders may exacerbate cognitive and behavioral symptoms
nearly two-thirds of those residing in nursing home facilities in patients with dementia and are a source of considerable
suffer from sleep disorders. Chronic and pervasive sleep com- stress for bed partners and family members. When left untreat-
plaints and disturbances are frequently associated with exces- ed, sleep disturbances may also increase the risk of injury at
sive daytime sleepiness and may result in impaired cognition, night, compromise health-related quality of life, and precipi-
diminished intellect, poor memory, confusion, and psychomo- tate and accelerate social and economic burdens for
tor retardation all of which may be misinterpreted as dementia. caregivers.
The key sleep disorders impacting patients with dementia in-
clude insomnia, hypersomnolence, circadian rhythm mis- Keywords Sleep . Sleep disorders . Dementia .
alignment, sleep disordered breathing, motor disturbances of Neurodegenerative disease . Sun-downing . Aging . REM
sleep such as periodic leg movement disorder of sleep and sleep behavior disorder . Alpha synucleinopathies .
restless leg syndrome, and parasomnias, mostly in the form Alzheimer’s dementia
of rapid eye movement (REM) sleep behavior disorder
(RBD). RBD is a pre-clinical marker for a class of
Introduction
This article is part of the Topical Collection on Sleep Disorders
Sleep is a critical function that is necessary for optimal cogni-
* Alon Y. Avidan tive functioning at all ages, but as age advances, there is an
avidan@mednet.ucla.edu
increased likelihood of sleep disturbances resulting in changes
Verna R. Porter to the quality and quantity of sleep [1, 2, 3••]. As age in-
vporter@mednet.ucla.edu
creases, a number of changes occur to disrupt sleep that may
William G. Buxton result in fatigue, irritability, headaches, depression, excessive
wbuxton@mednet.ucla.edu
daytime sleepiness, and changes in motor and cognitive func-
tioning among other effects [1]. Sleep disturbances are also
1
Department of Neurology, UCLA Mary S. Easton Center for common to many neurodegenerative diseases and may in-
Alzheimer’s Disease Research, David Geffen School of Medicine at clude difficulty with insomnia, hypersomnia, parasomnias,
UCLA, 10911 Weyburn Ave., Suite 200, Los
Angeles, CA 90095-7226, USA
excessive and abnormal nocturnal motor activity, disruption
2
of circadian rhythmicities, and respiratory dysregulation [1, 4,
Department of Neurology, David Geffen School of Medicine at
UCLA, 300 UCLA Medical Plaza, Suite B200, Los
5]. As the geriatric population continues to increase, so does
Angeles, CA 90095-6975, USA the prevalence of dementia. In the year 2000, there were a
3
Department of Neurology, UCLA Sleep Disorders Center, David
reported 34 million Americans over the age of 65 years, and
Geffen School of Medicine at UCLA, 710 Westwood Blvd., Room it is anticipated that by the year 2025, this number will nearly
1-145 RNRC, Los Angeles, CA 90095-1769, USA double to more than 62 million [6]. In 2010, it was estimated
97 Page 2 of 11 Curr Psychiatry Rep (2015) 17:97

that worldwide, 35.6 million people were affected by demen- with cognitive decline [15••, 16]. A recent study showed that
tia with an anticipated near-doubling of that number every these factors may be linked since age-related medial prefrontal
20 years to a projected 65.7 million affected in 2030 and cortex gray matter (mPFC) atrophy is associated with reduced
115.4 million affected in 2050 [7]. The medical and economic NREM slow-wave activity (SWA) that further appears to ac-
impact of dementia is profound, and it was estimated that as of count statistically for the impairment of overnight sleep-
2010, the worldwide economic impact of dementia had related memory consolidation. Together, these data support a
reached an estimated annual cost of greater than $604 billion model in which age-related mPFC atrophy decreases SWA,
(USD); this is equivalent to 1 % of the world’s gross domestic which disrupts long-term memory consolidation. Such find-
product [7, 8]. Effective management of sleep disturbances in ings suggest that sleep disruption in the elderly, accompanied
the elderly population with dementia has important social and by structural brain changes, represents a contributing factor to
clinical implications that may help bridge the gap between age-related cognitive decline in older individuals [17]. Sleep is
disease modifying therapies and current interventions de- also thought to serve a variety of physiological functions in-
signed to promote optimal cognitive functioning and delay cluding restoration of tissues and brain-metabolite clearance.
progression of dementia [9–11]. The etiology of different neu- Recently, the Bglymphatic system^ has been proposed in
rodegenerative conditions is diverse and complex. Similarly, mouse models to facilitate biomolecule clearance using a con-
the etiologies of different sleep disturbances within these dif- vective flow between the cerebrospinal fluid (CSF) and inter-
ferent neurodegenerative conditions also are varied [10]. As stitial fluid (ISF) to remove toxic metabolites in the brain
our understanding of the pathophysiology of different neuro- [18••]. There have been reports in mice that this glymphatic
degenerative conditions improves, so will our understanding system is strongly activated by sleep and may potentially help
of the effects of these conditions on the sleep-wake cycle of to clear toxic proteins known to contribute to the pathology of
the aging adult and the varied presentation of sleep distur- Alzheimer’s disease (AD) such as beta-amyloid (Aβ) [18••,
bances in different neurodegenerative conditions. 19••]. Additionally, soluble Aβ 40 and 42 levels have been
shown to be consistently higher during wakefulness and lower
during sleep; with age and with increasing amyloid deposi-
Sleep and Aging tion, as determined by PiB-PET scans, this sleep-wake varia-
tion decreases [15••, 20•, 21•]. This diurnal Aβ pattern has
Sleep is a complex phenomenon with physiological roots in also been observed in murine models (e.g., human amyloid
nervous system regions such as the hypothalamus, brainstem, precursor protein (APP) transgenic mice, Tg2576, that devel-
and thalamus [6]. At least one chronic sleep complaint has op amyloid deposition) indicating that chronic sleep restric-
been reported in more than 50 % of community-dwelling tion significantly enhances brain soluble concentrations of Aβ
adults over the age of 65 and in nearly two-thirds of nursing [22]. APP transgenic mice also have higher concentrations of
home residents [6, 12•]. The most common complaints are Aβ levels during wakefulness, and these levels decrease dur-
inability to initiate and maintain asleep (insomnia), followed ing sleep [22]. The investigators also showed that the use of an
by chronic complaints of excessive daytime sleepiness (EDS) orexin-receptor dual antagonist to induce sleep decreased Aβ
resulting in frequent naps [1, 6, 11]. plaque deposition; whereas sleep deprivation increased ISF
The quality and architecture of sleep clearly changes with concentrations of Aβ and accelerated Aβ deposition into
healthy aging, and insomnia and excessive daytime sleepiness plaques [22]. Recent human studies have suggested that Aβ
are frequently reported in the elderly population. Sleep also accumulation disrupts human non-REM sleep (NREM) and
increasingly fragments with age and is characterized by more associated slow waves (SWA), which may contribute to
nighttime awakenings and an increased desire for daytime hippocampal-dependent cognitive decline [17, 23••]. This
naps [6, 12•]. The homeostatic mechanisms and circadian provides for an interesting theoretical model in which Aβ
rhythms and architecture of sleep also alter with normal aging deposition is associated with diminished NREM sleep and
[13]. This includes reductions in slow-wave sleep (SWS) per- SWA, with NREM sleep disruption reciprocally promoting
cent and spectral power of sleep in the electroencephalogram an increased synaptic tone and metabolic activity in the brain
(EEG), number and amplitude of sleep spindles, rapid eye that in turn leads to the accumulation of toxic metabolites
movement (REM) concentration, and amplitude of circadian (Aβ) in the CSF, resulting in a subsequent Aβ plaque depo-
rhythms and phase advance (earlier onset) of the brain’s cir- sition that further worsens sleep [15••, 22, 23••].
cadian clock [14]. The notable role of sleep in facilitating cognitive function-
Of interest, nearly all people over the age of 60 have ing, memory stabilization, integration, and consolidation has
disrupted sleep architecture and decreased slow wave sleep long been recognized by psychological scientists [3••]. Con-
(SWS); and SWS is critical for appropriate consolidation of versely, inadequate sleep has been recognized even in middle-
long-term memory. Further, aging is associated with regional aged adults to contribute to disruption in attention, executive-
brain atrophy in midline frontal lobe regions that is associated control tasks, working memory, episodic memory, and an
Curr Psychiatry Rep (2015) 17:97 Page 3 of 11 97

increased frequency of cognitive complaints [3••]. Prolonged observed that insomnia frequency was identical in AD and
accumulation of sleep debt also impairs physical, mental, and frontotemporal dementia (FTD) patients, but about 2.5 and
emotional well-being and may give rise to the emergence of 1.5% more frequent, respectively, in vascular dementia
behavioral and mood disorders [6]. (VaD) and DLB/PD [9]. Finally, that same study suggested
The etiology of sleep disturbances in the elderly may be that patients with AD and mild cognitive impairment (MCI)
divided into internal (patient-related) and external had an almost identical frequency of any given sleep distur-
(environmental) factors. Internal factors consist of primary bance which did not allow any prediction concerning the con-
sleep disorders such as obstructive sleep apnea [24–26], peri- version from MCI to AD [9]. Sleep disordered breathing has
odic limb movements of sleep (PLMDS) [27], restless legs been shown to be more prevalent in VaD, and severe daytime
syndrome (Willis Ekbom Disease, WED) [28, 29], and sleepiness appears to be strongly predictive of vascular de-
parasomnias (e.g., rapid eye movement sleep behavior disor- mentia, but is not predictive of other non-vascular dementias
der, RBD) [30–32]. Sleep disturbances in the elderly may also [9, 45]. REM behavioral disorder is more frequently present in
arise because of comorbid medical conditions, psychiatric DLB and PD [9, 31, 44].
conditions, polypharmacy, or other psychosocial factors [11]. In some cases, use of medications that are designed to
Conversely, the presence of sleep disorders may also exacer- modify various aspects of neurodegenerative conditions
bate underlying medical and psychiatric conditions [6]. Large (e.g., benzodiazepines, selective serotonin reuptake inhibitors)
prospective studies have also suggested that excessive day- may induce side effects of sleepiness, disrupted sleep archi-
time sleepiness may be independently associated with an in- tecture, or even precipitate RBD [32]. For caregivers, disrup-
creased risk of cognitive decline in the elderly population, thus tive behaviors include being awakened at night and patient
suggesting a potential target for early intervention [2, 33–35]. wandering, confusion (e.g., Bsun-downing^), getting out of
External factors may include increased ambient lighting dur- bed repeatedly and talking in bed. These comprise some of
ing the night, excessive noise in the sleep environment, or the most disruptive behaviors that may lead to earlier nursing
temperatures that are too warm or too cold. These external home placement [46].
factors are relatively simple to remedy as one formulates a
treatment plan [36].
Alzheimer’s Disease

Sleep Disturbances in Patients With AD is the most common form of age-related dementia and is
Neurodegenerative Disease characterized by progressive memory loss and cognitive dete-
rioration. Familial, early-onset (<60 years of age), autosomal-
Dementia (major neurocognitive disorder), as defined by dominant forms of AD (FAD) have now been shown to asso-
DSM-V criteria, is characterized by a substantial cognitive ciate with mutations in the genes encoding for the amyloid
decline from a previous level of performance in one or more precursor protein (APP), presenilin 1 (PSEN1) and presenilin
cognitive domains and a decline in neurocognitive perfor- 2 (PSEN2) [47]. Sporadic AD typically presents at a later
mance, typically involving a test performance in the range of onset (>60 years) and is due to multifactorial genetic and
two or more standard deviations below appropriate norms environmental risk factors [48]. Memory disturbances are
(i.e., below the third percentile) on formal testing or an equiv- the most common initial presenting symptoms of AD, and in
alent clinical evaluation. Further, these cognitive deficits are particular the condition is characterized by difficulty remem-
sufficient to interfere with independence (i.e., necessitating bering recent events (short-term memory loss). As the disease
assistance with instrumental activities of daily living) [37, progresses, patients present with worsening episodic memory,
38]. Dementia is currently estimated to affect about 6 % of executive dysfunction, language compromise (e.g., naming or
individuals over the age of 65 and is strongly correlated with semantic problems), loss of visual orientation, behavioral
advancing age [39, 40]. changes, loss of motivation, and eventually, poor self-care
Alzheimer’s disease (AD) is the most common cause of and behavioral issues. Patients with AD are often affected
dementia, accounting for about 70 % of all causes of dementia by primary circadian dysrhythmia as suggested by the pres-
and is followed in prevalence by dementia with Lewy bodies ence of insomnia, hypersomnia, and a tendency to sleep more
that accounts for about 20 to 25 % of cases [6]. Nocturnal and during the day and less at night [6]. As a patient’s condition
daytime sleep disturbances affect as many as 44 % of AD progresses and cognition decreases, she or he often withdraws
patients in clinic and community-based samples [39, 41]. from family and society. Although the speed of progression
However, the highest frequency of sleep disturbances occurs can vary notably, the average life expectancy following diag-
in individuals with dementia with Lewy bodies (DLB) and nosis is three to nine years [5, 40, 49]. Pathologically, AD is
Parkinson’s disease (PD) with nearly 90 % of individuals af- characterized by the presence of senile plaques, which are
fected [5, 12•, 43–45]. A recent prospective cohort study comprised of extracellular amyloid beta (Aβ) deposits that
97 Page 4 of 11 Curr Psychiatry Rep (2015) 17:97

have historically been felt to be a fundamental pathological suprachiasmatic nucleus (SCN) may be responsible for alter-
hallmark of AD either by overproduction or some failed clear- ations in circadian rhythmicity, Bsun-downing^ and other
ance mechanism (i.e., amyloid hypothesis); in 2009, this the- sleep-wake disturbances [1, 6]. Degeneration of the brainstem
ory was updated, suggesting that an associated amyloid pro- respiratory neurons and supramedullary respiratory pathways
tein (N-APP) may be a major culprit in the pathophysiology of may contribute to sleep disordered breathing (SDB) and other
the disease [50, 51]. The other pathological hallmark of AD is respiratory dysregulation problems affecting sleep in AD [1,
neurofibrillary tangles, which are composed of a filamentous 6]. Even in the setting of amnestic mild cognitive impairment,
aggregation of hyperphosphorylated tau (p-tau) proteins [47, there are already known clinical (increased insomnia and sleep
48]. Most recently (2015), novel stem cell 3D gel culture disordered breathing) and electroencephalographic (increased
systems have further helped elucidate the sequence of events slowing in wakefulness and REM sleep and attenuation of
in AD pathogenesis, starting with extracellular aggregation sleep spindles and slow-wave sleep) abnormalities, many of
amyloid-β (Aβ) and followed by the accumulation of which are associated with decreases in sleep-dependent mem-
hyperphosphorylated tau. The 3D culture generation takes ory consolidation [9, 14, 59].
1–2 days and is associated by aggregation of Aβ observed Patients with Alzheimer’s disease suffer from a number of
after 6 weeks of differentiation, followed by robust tau pathol- sleep disruptions including insomnia, frequent nighttime
ogy after 10–14 weeks [47]. This has further solidified the awakenings, decreased total nighttime sleep, increased day-
notion that amyloid-β (Aβ) accumulation is the inciting time sleep, and evening agitation [60]. Patients with AD have
event, followed by the accumulation of hyperphosphorylated significant sleep architectural disruption including reduced
tau and neurofibrillary tangle formation, with neuronal cell sleep efficiency, increased non-REM stage N1 sleep, in-
dysfunction and death likely perpetuated by the inflammatory creased arousal and awakening frequency, decreased total
milieu that contributes to dysregulation of clearance of toxic sleep time (TST), and reduced sleep spindles and K com-
metabolites, other mechanisms that lead to misfolded or dam- plexes [6]. Some of the most disruptive behaviors in patients
aged neuronal proteins, and to tau-associated disruptions of with AD include the profound disruption in sleep-wake rhyth-
axonal transport integrity [47, 48, 52]. micity that occurs early in the disease and results in sleep
Individuals with the apolipoprotein E (APOE- 4) allele are fragmentation, increased daytime sleepiness, insomnia, noc-
predisposed to accumulate Aβ and are at an increased risk for turnal wandering, progressive cognitive decline, increase in
AD [11]. Interestingly, the APOE- 4 allele is the most proin- the number of daytime naps, increased time spent in bed (in-
flammatory allele and has also been associated with as much cluding awake time in bed), and increased nocturnal wander-
as a two-fold higher risk for the development of obstructive ing, disorientation, and confusion [1, 5, 12•, 58, 61–63].
sleep apnea (OSA), with OSA conferring a higher risk for Therefore, in patients with AD, support exists that sleep
subsequent cognitive impairment [11, 53–55]. Conversely, disruption results in poorer outcomes across multiple do-
better sleep consolidation diminishes the effects APOE geno- mains: (1) poor sleep quality is predictive of cognitive decline
type on the incidence of AD and the subsequent development [14, 64, 65••], (2) good sleep quality may help mitigate against
of neurofibrillary tangle pathology [53]. Chronic systemic in- genetic susceptibility factors, e.g., APOE genotype, possibly
flammation may therefore link many AD risk factors (i.e., by increasing clearance of Aβ from the brain [18••, 66], and
APOE genotype, cardiovascular disease, Type II diabetes (3) poor sleep quality is proinflammatory and is associated
mellitus, poor diet, a sedentary lifestyle, and poor sleep qual- with a higher risk of metabolic (e.g., diabetes) and cardiovas-
ity), all of which may contribute to the proinflammatory path- cular disease, which have been suggested to be independent
ophysiology of this condition [11]. risk factors for AD [11]. To highlight this, Fig. 1 illustrates the
Central to the pathophysiological changes of AD is the loss schematic association between circadian dysregulation and
of cholinergic cells of the nucleus basalis complex and pro- poor sleep in the context of AD pathogenesis [11]. Production
gressive degeneration of the cortical and subcortical structures of soluble Aβ may be relatively increased during the sleep
[6]. Interestingly, the use of the cholinesterase inhibitor, period by reduced NREM SWA in the context of aging and
donepezil, improved OSA by improving the apnea- associated sleep disturbances (e.g., sleep apnea or insomnia).
hypopnea index (AHI) and oxygen saturation after donepezil These sleep disruptions result in increased time awake during
treatment [56, 57]. Sleep-wake disturbances such as increased the relative sleep period, and may link Aβ pathology with
daytime napping and insomnia at night affect 25 to 40 % of hippocampal-dependent cognitive decline [17, 23••].
patients with mild-to-moderate AD [58]. Degeneration of the During sleep, the default mode network (DMN), which is
cholinergic neurons in the nucleus basalis of Meynert, the active during non-task performance and necessary for episod-
pedunculopontine and laterodorsal tegmental nuclei, and the ic memory retrieval, deactivates during sleep and particularly
noradrenergic neurons of the brainstem may be responsible for SWS; it is this domain that is also adversely affected in early
the decreased rapid eye movement sleep (REM) in patients AD. With decreased sleep efficiency and associated decreased
with AD [1, 6]. Degeneration of neurons of the SWS, there is a lack of deactivation of the DMN which results
Curr Psychiatry Rep (2015) 17:97 Page 5 of 11 97

and paralimbic) in both Parkinson’s disease (PD) and DLB;


however, Kosaka et al. have identified Lewy bodies in the
cerebral cortex and subcortical white matter of patients with
DLB, which do not necessarily demonstrate the characteristic
halo but do stain positive for α-synuclein and ubiquitin [67].
Typical clinical features of the condition include fluctuating
alertness, visual hallucinations and predilection to the develop-
ment of RBD and Parkinsonian motor symptoms with a de-
mentia characterized by both cortical and subcortical features.
It is the second most common form of dementia after AD and
accounts for 25 % of all dementias [68]. The prevalence of this
condition is estimated to be 0.7 % of the population over the
Fig. 1 AD pathophysiology as a product of circadian dysregulation and age of 65 years with a greater male predominance of the con-
poor sleep: Recent data illustrates that the production of a soluble Aβ may dition, 1.9:1 [69–71]. Risk factors for the development of DLB
be relatively accelerated during sleep due to the reduced NREM SWA in
the context of aging and associated sleep disturbances (e.g., sleep apnea
include advanced age, hypertension, dyslipidemia, and a car-
or insomnia). Disturbed sleep results in an increased time awake during rier of one or more APOE 4 alleles [71–73]. As in PD, there is
the relative sleep period, and may link Aβ pathology with the a higher proportion of the CYP2D6B allele and mutations in
hippocampal-dependent cognitive decline [15••] glucocerebrosidase 1 (GBA1), which are significant risk fac-
tors for DLB [74, 75]. In DLB, a greater heterozygous frequen-
in an increase in synaptic and metabolic neuronal activity; this cy of GBA1 exists than in PD, 3.5 versus 2.9 %, supporting the
results in increased soluble Aβ levels in the CSF during the notion that GBA1 mutations likely play an even larger role in
sleep period, and subsequent increased Aβ aggregation and the genetic etiology of DLB than in PD [74, 75].
deposition into plaques with attenuation in the normal Aβ Sleep disturbances with nighttime sleep disruptions are
diurnal rhythms [15••]. Further, a decrease in SWS is associ- common in DLB, and in fact, are more common in DLB than
ated with age-related atrophy of the medial prefrontal cortex in AD patients [43]. Patients with DLB suffer more movement
(mPFC) gray matter, which may also give rise to cognitive disorders during sleep and more daytime sleepiness [43]. In
deterioration and decline [17, 23••]. Although changes in the patients with DLB, RBD may precede cognitive and motor
sleep-wake cycle have been associated with markers for AD impairments by many years [70, 71, 73]. RBD is a REM
pathology (e.g., amyloid deposition), the precise associations parasomnia that is strongly associated with alpha
between advancing AD pathology and sleep-wave cycle alter- synucleinopathies. It is demarcated by vivid, often frighten-
ations must still be further investigated [17, 23••]. ing, quasi dream-enactments associated with motor behaviors
An alternative explanation is that sleep changes are markers during REM sleep, which may result in injuries to the patient
of AD pathology rather than an initiating event in the cascade or his/her bed partners [76]. The polysomnographic features
of AD pathogenesis per se. This model suggests that many of RBD include abnormal muscle augmentation during REM
changes occur during aging including brain atrophy (regional sleep (known as REM sleep without atonia) [76]. Key epide-
atrophy in the mFPC) and changes in sleep parameters (e.g., miological studies suggest that the majority of patients with
increased wake after sleep onset, decreased sleep efficiency, RBD are older men generally in the sixth or seventh decade of
decreased SWS and increased incidence of various sleep dis- life [59]. More recent longitudinal studies have suggested a
orders). These factors interact within an individual with genet- high rate of phenoconversion of patients with idiopathic RBD
ic and environmental factors that ultimately determine an in- (iRBD) to α-synucleinopathies such as PD, DLB, and multi-
dividual’s relative risk of and venerability to developing AD. system atrophy (MSA) [32, 43, 77–79]. Several markers of
In a given individual, depending on the interplay of these other neurodegeneration have been identified in RBD, including
factors, sleep will play a greater or lesser determinant in cognitive impairments such as deficits in attention, executive
whether that individual develops subsequent AD [15••]. functions, learning capacities, and visuospatial skills [5, 32,
43, 62, 77–79]. Approximately 50 % of RBD patients also
present with features of mild cognitive impairment (MCI) [9,
Dementia With Lewy Bodies 62, 64, 77]. RBD has also been highly associated with cogni-
tive decline in PD [32, 77, 78, 80]. A previous study docu-
Dementia with Lewy bodies (DLB) is an atypical Parkinso- mented that 38 % of neurologically normal men older than
nian condition characterized as an alpha synucleinopathy with 50 years with RBD subsequently develop a Parkinsonian dis-
the cardinal neuropathological finding of the Lewy body, a order [81].
neuronal inclusion composed of aggregated α-synuclein Of interest, the association of EEG slowing and the pres-
[67]. Lewy bodies are found in the brainstem nuclei (limbic ence of MCI, which is frequently seen in the early stages of
97 Page 6 of 11 Curr Psychiatry Rep (2015) 17:97

DLB, has been an area of active investigation in an effort to related quality of life (HRQOL) measures in PD shows that
identify objective indicators of those individuals at risk for essentially all measures of HRQOL in a community-dwelling
conversion to α-synucleinopathies such as PD, DLB, and sample of PD patients were negatively affected by reports of
MSA [76]. In a recent study (2010), patients with idiopathic significant sleep disturbances, particularly sleep maintenance
RBD (iRBD) without MCI and healthy controls were follow- insomnia, daytime sleepiness, and inadequate sleep, as well as
ed for 2.4 years. Patients with RBD who developed MCI had awakening short of breath or with a headache [42, 82]. Sleep
marked EEG slowing (theta and delta) in central and occipital spindles and slow waves at baseline have been shown to con-
regions during wakefulness and REM sleep in baseline studies tribute to brain plasticity and cognitive performance [80]. Re-
as compared to controls and iRBD patients who had not de- sults of recent studies suggest that non-rapid eye movement
veloped MCI by the time of follow-up. Further, the EEG spec- sleep irregularities noted on EEG in PD correlate with cogni-
tral pattern of RBD+MCI group was similar to that found in tive decline and risk of developing dementia [83]. Specifical-
patients with DLB or PD associated with dementia [59]. These ly, sleep spindle activity was notably impaired in PD patients
findings suggest that EEG slowing on spectral analysis may who subsequently developed dementia, with a more posterior
be predictive of the development of MCI within a short period topographical pattern [80]. This supports the notion that sleep
in patients with an initial diagnosis of iRBD. spindle alterations are associated with subsequent develop-
In contrast to the dorsal prefrontal pathology seen in AD ment of dementia in patients with PD [80]. Further, a high
and its relationship to sleep, the aberration of REM sleep seen percentage of PD patients without frank sleep complaints
in RBD has its pathophysiologic correlates in lower brain may have subclinical or clinical RBD, and RBD may precede
structures (i.e., pontine and midbrain nuclei). Since the the onset of motor manifestations of Parkinsonism by many
pedunculopontine nucleus and the locus coeruleus represent years, particularly in older male patients [1]. Studies have also
the largest clusters of cholinergic and noradrenergic neurons shown that cognitive impairment in non-demented patients
in the brainstem reticular system, and are known to play a role with PD is closely correlated with the presence of RBD [79].
in arousal and cortical activation, it has been proposed that a
dysfunctional cholinergic system may be contributing to cog-
nitive impairment and EEG slowing in iRBD patients [76]. Vascular Dementia
Further, other studies have shown (by single-photon emission
computer tomography studies) iRBD+MCI patients demon- Ischemic or hemorrhagic cerebrovascular disease (CVD)
strate hypoperfusion in the occipital and temporal regions causes injury to brain regions that are important for executive
compared to iRBC−MCI and controls. Other brain regions functions, behavior and memory, leading to decline in cogni-
(e.g., left superior temporal lobe, right occipital lobe, both tive functions and vascular dementia (VaD). The causes of
hippocampi and the left parahippocampal gyrus) have also cerebrovascular disease are diverse (e.g., ischemic, embolic,
been implicated in other imaging studies investigating patients thrombotic, and microvascular) and include VaD from hypo-
with iRBD [76]. Taken together, these studies imply that struc- perfusion of susceptible brain areas. CVD may also worsen
tural changes in the brain and brainstem are responsible for degenerative dementias such as Alzheimer’s disease (AD)
EEG slowing and cognitive decline. Therefore, a subset of [49, 73]. Currently, the broader diagnostic category for cogni-
patients with EEG slowing and the sleep disturbances associ- tive impairment of vascular origin is vascular cognitive disor-
ated with RBD are at increased risk of dementia, and the basis der (VCD). VCD ranges from vascular cognitive impairment
is quite distinct from the link between slow wave sleep and (VCI) to VaD. The term VCI is used for cases of cognitive
Alzheimer’s disease. impairment of vascular etiology without frank dementia; VCI
is equivalent to vascular mild cognitive impairment (MCI).
Risk factors for VaD include age, hypertension, diabetes,
Parkinson’s Disease smoking, cardiovascular disease (coronary heart disease, con-
gestive heart failure, peripheral vascular disease), atrial fibril-
Parkinson’s disease (PD) is the most common neurodegener- lation, left ventricular hypertrophy, hyperhomocysteinemia,
ative movement disorder with an annual incidence of 13.4 per orthostatic hypotension, cardiac arrhythmias,
100,000 in the US [42]. Sleep disturbances and cognitive im- hyperfibrinogenemia, sleep apnea, infection, and high C-
pairment are common non-motor manifestations of PD [80]. reactive protein [84]. Patients with vascular dementia experi-
Various sleep-related conditions that interfere with quality of ence disruption in sleep-wake cycles and decreased sleep ef-
life in PD patients include inability to fall asleep and stay ficiency [85]. As previously reported in the Caerphilly cohort
asleep, an urge to move the legs in the evening time (restless of older men, sleep disturbances were predictive of risk of
legs syndrome), abnormal and disruptive leg jerks at night ischemic stroke (relative odds of up to 1.97; 95 % confidence
(periodic leg movement disorder) and excessive daytime som- limits: 1.26, 3.09), and daytime sleepiness is associated with
nolence [42, 82]. A recent study of the associations of health- an increase in vascular disease events (relative odds: 1.41;
Curr Psychiatry Rep (2015) 17:97 Page 7 of 11 97

1.04, 1.92) [86]. Research on biomarkers revealed increased addition, bright-light therapy (BLT) (i.e., using a full-spectrum
CSF-neurofilament protein levels in VaD, with CSF-tau levels light box) for a minimum of half-an-hour daily, usually in the
that were normal. VaD also shows low CSF acetylcholinester- morning, may enhance sleep in the AD population [90–93].
ase levels; this condition is known to show a response to Most studies have suggested that BLT has positive effects on
acetylcholinesterase inhibitors, confirming the role of cholin- normalizing circadian rhythms and improving sleep function
ergic deficit in its pathogenesis. Evidence strongly suggests by reducing sleep fragmentation and increasing the duration
that the control of vascular risk factors, particularly hyperten- of sleep and improving some cognitive and noncognitive
sion, could help prevent VaD [84]. Recent studies further sug- symptoms of dementia [90, 91, 93–95].
gest that sleep disturbances, and specifically excessive day- Pharmacological therapies may include use of melatonin,
time sleepiness, appear to be strongly correlated with a higher which has been proposed to have cytoprotective, antioxidant,
predictive power for subsequent VaD, but not for non-vascular and perhaps even anti-amyloid effects as well as modest ef-
dementias [45]. fects on improving day/nighttime sleep ratios and decreasing
nocturnal activity and sun-downing in patients with AD [10,
60, 93, 96, 97•, 98, 99]. Also, patients with PD have been
Frontotemporal Dementia shown to have a significantly less increase in nighttime mel-
atonin release compared to controls. Additionally, nighttime
Frontotemporal dementia refers to a range of neurodegenera- melatonin levels were shown to be significantly lower in the
tive disorders that are characterized by focal atrophy in the subset of patients with PD experiencing excessive daytime
frontal and/or temporal lobes of the brain that often results in sleepiness than in the subset without daytime sleepiness, thus
profound behavioral, cognitive, and emotional symptoms suggesting the potential utility of supplemental melatonin in
[87]. Two subtypes of FTD are the behavioral variant patients with PD [100••].
(bvFTD) and semantic dementia (SD). Much less is known Other pharmacological treatments that have been utilized
about sleep disturbances in FTD, but sleep disruptions that are with variable success include antipsychotics (with notable side
characterized by increased nocturnal activity, decreased morn- effects of sedation, increased fall risk and serious cardiac side
ing activity, and excessive daytime sleepiness have all been effects with second-generation antipsychotics), hypnotics
reported in patients with FTD [1, 87–89]. (promote sedation at the expense of altering sleep architec-
ture), antidepressants (side effects include somnolence, seda-
tion, anticholinergic effects and dizziness), antihistamines
Treatments for Sleep Disturbances in Patients With (side effects include sedation, anticholinergic effects, dizzi-
Dementia ness and somnolence), and acetylcholinesterase inhibitors
(may increase REM sleep associated parasomnias, but may
The normal human aging process produces biological changes increase REM sleep percentage and density [30, 32, 57,
in circadian rhythms that produce sleep disorders and disrup- 97•]). Newer agents such as melatonin receptor agonists and
tions of the sleep-wake cycle. The prevalence of sleep disor- an orexin-receptor antagonist may be more specific in their
ders is higher in patients with dementia, and sleep disorders mechanism of action [96, 100••, 101]. The appropriate use of a
are often the cause of increased caregiver burden. In treat- combination of pharmacological and non-pharmacological
ments of sleep disorders in this cohort, it is important to re- measures to enhance appropriate sleep may successfully in-
view sleep disorders carefully with the bed partners or family crease a patient’s quality of life, improve daytime activity,
members in order to understand the behavioral and psycho- increase sleep efficiency, decrease agitated behaviors, reduce
logical impact of dementia on sleep. In clinical practice, non- depressive symptoms, and even reduce cognitive compromise
pharmacological therapies, including cognitive-behavioral in patients with dementia [10, 90, 94].
and psycho-educational strategies, should be utilized first Maximizing environmental safety and protecting the
and consist of interventions such as combined daytime phys- sleeping environment by removing sharp objects from the
ical activity, sleep hygiene, and an enhanced nighttime envi- bedroom area are cornerstone strategies for patients with both-
ronment. In these enhanced environmental programs, patients ersome dream enactment in the setting of RBD. Suggested
are encouraged to (1) exercise regularly (ideally for 30 min) pharmacotherapy for RBD may be in the form of melatonin
during the daytime hours, (2) walk outside in natural light as (3–12 mg P.O. Q.H.S) or clonazepam (0.25 to 1 mg P.O.
often as possible, (3) decrease time spent in bed awake at night Q.H.S). The former is effective in up to 90 % of patients,
and napping during the daytime, (4) maintain regular bedtimes and while there is little evidence of tolerance or abuse with
and arising times, (5) keep the bedroom reserved for sleep and this pharmacological approach, caution should be exercised
a quiet environment with minimal noise/light/TVor light emit- when using it in patients with chronic respiratory diseases or
ting electronics, and (6) minimize use of cholinesterase inhib- impaired renal function [102]. The exact mechanism of action
itors and stimulating drugs during the pre-sleep time [90]. In of melatonin in improving RBD is unknown, but it probably
97 Page 8 of 11 Curr Psychiatry Rep (2015) 17:97

exerts its therapeutic effect by restoring REM sleep atonia. Compliance with Ethical Standards
One study reported that melatonin was effective in 87 % of
Conflict of Interest Verna R. Porter and William G. Buxton declare
patients taking 3 to 9 mg at bedtime [103], whereas follow-up that they have no conflict of interest.
data reported resolution in those taking 6 to 12 mg of melato- Alon Y. Avidan is a consultant for Vanda, Merck, is on the speaker’s
nin at bedtime [104]. The readers should be reminded that bureau for Merck and Xenoport and received stipends as an author and
lecturer on behalf of the Elsevier, LWW, Best Doctors, AAN, AASM,
melatonin, a food supplement, is not approved by the Food
ACCP, and CHEST.
and Drug Administration and has poor regulation in terms of
pharmacologic preparation. Human and Animal Rights and Informed Consent This article does
not contain any studies with human or animal subjects performed by any
of the authors.

Conclusion

Sleep changes profoundly with advancing age. Across both


subjective and objective measures, data confirms an increase References
in sleep-wake disturbances, sleep fragmentation, less deep
sleep, and a tendency towards early morning awakening. In
Papers of particular interest, published recently, have been
older adults with dementia, the prevalence of sleep-related
highlighted as:
breathing disorder, periodic limb movement disorder of sleep
• Of importance
and restless leg syndrome increases and may lead to excessive
•• Of major importance
daytime sleepiness, insomnia and neurocognitive compromise
that may exacerbate and accelerate the neurodegenerative pro- 1. Bhatt MH, Podder N, Chokroverty S. Sleep and neurodegenera-
cess. Since many of the sleep disorders are potentially revers- tive diseases. Semin Neurol. 2005;25:39–51.
ible, health care providers should incorporate a review of sleep 2. Keage HA, Banks S, Yang KL, et al. What sleep characteristics
disturbances into a carefully integrated and diagnostic predict cognitive decline in the elderly? Sleep Med. 2012;13:886–
92.
decision-making process with the aim to benefit the patient 3.•• Scullin MK, Bliwise DL. Sleep, cognition, and normal aging:
and their family. Sleep problems of the older adult with early integrating a half century of multidisciplinary research. Perspect
dementia may contribute significantly to the decision to even- Psychol Sci. 2015;10:97–137. A nicely written article reviewing
tually institutionalize the patient, which could magnify the a perplexing question on whether the alterations in sleep phys-
iology can impact cognition. The authors review 50 years of
social and economic burden of chronic nursing home care. It research across diverse experimental protocols concluding
is now understood that sleep disturbances may lead to Aß that maintenance of adequate sleep in younger-middle age
deposition, and therefore fundamentally contribute to the promotes better cognitive functioning and may even be pro-
pathogenesis of conditions such as AD. In patients with alpha tective against age-dependent cognitive declines.
4. Bliwise DL. Sleep in normal aging and dementia. Sleep. 1993;16:
synucleinopathies, the presence of sleep disorders such as
40–81.
RBD may help predict those with a unique vulnerability to 5. Bliwise DL. Sleep disorders in Alzheimer’s disease and other
phenoconversion to a dementia by up to a decade before the dementias. Clin Cornerstone. 2004;6(Suppl 1A):S16–28.
dementia is clinically apparent. In both scenarios, sleep is 6. Avidan AY. Sleep and neurologic problems in the elderly. Sleep
central to the notion that (1) disrupted sleep may directly con- Med Clin. 2006;1:273–92.
7. Prince M, Bryce R, Albanese E, et al. The global prevalence of
tribute to dementia, and (2) the presence of sleep pathology dementia: a systematic review and metaanalysis. Alzheimers
can predict, with a high degree of certainty, a patient’s risk of Dement. 2013;9:63–75. e62.
conversion to clinical dementia. In the first scenario, this ar- 8. Wimo A, Jonsson L, Bond J, et al. The worldwide economic
gues strongly for the necessity to treat and manage disrupted impact of dementia 2010. Alzheimers Dement. 2013;9:1–11. e13.
9. Guarnieri B, Adorni F, Musicco M, et al. Prevalence of sleep
sleep. In the second scenario, the scientific community is en- disturbances in mild cognitive impairment and dementing disor-
couraged to develop neuroprotective agents and interventions ders: a multicenter Italian clinical cross-sectional study on 431
for those patients who present with sleep pathology. Sleep is patients. Dement Geriatr Cogn Disord. 2012;33:50–8.
pivotal to the health of the population, and its disruption con- 10. Guarnieri B, Musicco M, Caffarra P, et al. Recommendations of
the Sleep Study Group of the Italian Dementia Research
tributes meaningfully to an individual’s relative risk of demen-
Association (SINDem) on clinical assessment and management
tia within the population. A careful assessment of a patient’s of sleep disorders in individuals with mild cognitive impairment
sleep patterns should thus be the new Bvital sign^ for every and dementia: a clinical review. Neurol Sci. 2014;35:1329–48.
clinical visit. 11. Landry GJ, Liu-Ambrose T. Buying time: a rationale for examin-
ing the use of circadian rhythm and sleep interventions to delay
progression of mild cognitive impairment to Alzheimer’s disease.
Acknowledgments This material is the result of work supported with Front Aging Neurosci. 2014;6:325.
the resources and use of facilities at the UCLA Department of Neurology 12.• Cipriani G, Lucetti C, Danti S, Nuti A. Sleep disturbances and
in Los Angeles, California, USA. dementia. Psychogeriatrics. 2015;15:65–74. A timely review
Curr Psychiatry Rep (2015) 17:97 Page 9 of 11 97

covering the etiology and consequences of and potential inter- disruption and memory impairment in older adults and
ventions for disrupted sleep in patients with dementia. connecting disturbed sleep as a potential mechanism
13. Motohashi Y, Maeda A, Wakamatsu H, et al. Circadian rhythm by which β-amyloid pathology could induce cognitive decline
abnormalities of wrist activity of institutionalized dependent elder- in older adults.
ly persons with dementia. J Gerontol A Biol Sci Med Sci. 24. Lee SD, Kang SH, Ju G, et al. The prevalence of and risk factors
2000;55:M740–3. for sleep-disordered breathing in an elderly Korean population.
14. Pace-Schott EF, Spencer RM. Sleep-dependent memory consoli- Respiration. 2014;87:372–8.
dation in healthy aging and mild cognitive impairment. Curr Top 25. Olson EJ, Park JG, Morgenthaler TI. Obstructive sleep apnea-
Behav Neurosci. 2015;25:307–30. hypopnea syndrome. Prim Care. 2005;32:329–59.
15.•• Lucey BP, Bateman RJ. Amyloid-beta diurnal pattern: possible 26. Sforza E, Roche F, Thomas-Anterion C, et al. Cognitive function
role of sleep in Alzheimer’s disease pathogenesis. Neurobiol and sleep related breathing disorders in a healthy elderly popula-
Aging. 2014;35 Suppl 2:S29–34. An important paper from tion: the SYNAPSE study. Sleep. 2010;33:515–21.
the Washington University group examining the circadian 27. Carelli G, Krieger J, Calvi-Gries F, Macher JP. Periodic limb
patterns of Aβ concentrations, reviewing animal models that movements and obstructive sleep apneas before and after contin-
depict that changes in sleep time alter Aβ deposition. The uous positive airway pressure treatment. J Sleep Res. 1999;8:211–
investigators suggest that sleep may play an important role 6.
in AD pathogenesis, and propose a hypothetical model for
28. Berger K, Luedemann J, Trenkwalder C, et al. Sex and the risk of
examining the role of sleep and the Aβ diurnal pattern in
restless legs syndrome in the general population. Arch Intern Med.
promoting to the pathogenesis of AD.
2004;164:196–202.
16. Redline S, Kirchner HL, Quan SF, et al. The effects of age, sex,
ethnicity, and sleep-disordered breathing on sleep architecture. 29. Trenkwalder C, Hening WA, Montagna P, et al. Treatment of
Arch Intern Med. 2004;164:406–18. restless legs syndrome: an evidence-based review and implica-
tions for clinical practice. Mov Disord. 2008;23:2267–302.
17. Mander BA, Rao V, Lu B, et al. Prefrontal atrophy, disrupted
NREM slow waves and impaired hippocampal-dependent mem- 30. Aurora RN, Zak RS, Maganti RK, et al. Best practice guide for the
ory in aging. Nat Neurosci. 2013;16:357–64. treatment of REM sleep behavior disorder (RBD). J Clin Sleep
18.•• Mendelsohn AR, Larrick JW. Sleep facilitates clearance of metab- Med. 2010;6:85–95.
olites from the brain: glymphatic function in aging and neurode- 31. Malhotra R, Avidan AY. Neurodegenerative disease and REM
generative diseases. Rejuvenation Res. 2013;16:518–23. A beau- behavior disorder. Curr Treat Options Neurol. 2012;14:474–92.
tifully illustrated review summarizing the glymphatic system 32. Trotti LM. REM sleep behaviour disorder in older individuals:
in humans which opens up new possibilities for the therapy epidemiology, pathophysiology and management. Drugs Aging.
and prevention of dementia. The paper illustrates how healthy 2010;27:457–70.
sleep as a public health initiative in middle-aged and older 33. Foley D, Monjan A, Masaki K, et al. Daytime sleepiness is asso-
people could play a vital role in protecting from and ciated with 3-year incident dementia and cognitive decline in older
preventing the future development of AD, especially among Japanese-American men. J Am Geriatr Soc. 2001;49:1628–32.
those with the apolipoprotein E (ApoE) ɛ4 allele. 34. Jaussent I, Bouyer J, Ancelin ML, et al. Excessive sleepiness is
19.•• Kress BT, Iliff JJ, Xia M, et al. Impairment of paravascular clear- predictive of cognitive decline in the elderly. Sleep. 2012;35:
ance pathways in the aging brain. Ann Neurol. 2014;76:845–61. A 1201–7.
pivotal investigation proposing that an impaired glymphatic 35. Merlino G, Piani A, Gigli GL, et al. Daytime sleepiness is associ-
clearance could contribute to cognitive decline among older ated with dementia and cognitive decline in older Italian adults: a
patients, which could constitute a novel therapeutic target population-based study. Sleep Med. 2010;11:372–7.
for the management of neurodegenerative diseases associated 36. Vitiello MV, Borson S. Sleep disturbances in patients with
with the abnormal accumulation of misfolded protein Alzheimer’s disease: epidemiology, pathophysiology and treat-
aggregates. ment. CNS Drugs. 2001;15:777–96.
20.• Huang Y, Potter R, Sigurdson W, et al. Effects of age and amyloid 37. Association AP. Diagnostic and statistical manual of mental dis-
deposition on Aβ dynamics in the human central nervous system. orders. 5th ed. DC:: Washington; 2013.
Arch Neurol. 2012;69:51–8. A succinct review, appraising the
38. Tay L, Lim WS, Chan M et al. New DSM-V neurocognitive
current literature related to the mechanisms by which dis-
disorders criteria and their impact on diagnostic classifications of
turbed sleep and circadian rhythms could play a role in the
mild cognitive impairment and dementia in a memory clinic set-
pathogenesis of AD. The authors discuss potential therapeutic
ting. Am J Geriatr Psychiatry 2015.
strategies directed at these systems for the prevention of AD.
39. Avidan AY. Clinical neurology of insomnia in neurodegenerative
21.• Roh JH, Huang Y, Bero AW, et al. Disruption of the sleep-wake
and other disorders of neurological function. Rev Neurol Dis.
cycle and diurnal fluctuation of beta-amyloid in mice with
2007;4:21–34.
Alzheimer’s disease pathology. Sci Transl Med. 2012;4:150ra–
122. In this comprehensive review, the authors argue that fluc- 40. Beydoun MA, Beydoun HA, Gamaldo AA, et al. Epidemiologic
tuations in sleep-wake behavior and diurnal variations of Aβ studies of modifiable factors associated with cognition and demen-
in the CNS could be surrogate functional and biochemical tia: systematic review and meta-analysis. BMC Public Health.
biomarkers of Aβ -associated pathology. The authors suggest 2014;14:643.
that abnormalities in sleep-wake rhythmicity should be ex- 41. McCurry SM, Gibbons LE, Logsdon RG, et al. Nighttime insom-
plored given potential therapeutic opportunities. nia treatment and education for Alzheimer’s disease: a random-
22. Kang JE, Lim MM, Bateman RJ, et al. Amyloid-beta dynamics ized, controlled trial. J Am Geriatr Soc. 2005;53:793–802.
are regulated by orexin and the sleep-wake cycle. Science. 42. Avidan A, Hays RD, Diaz N, et al. Associations of sleep distur-
2009;326:1005–7. bance symptoms with health-related quality of life in Parkinson’s
23.•• Mander BA, Marks SM, Vogel JW et al. beta-amyloid disrupts disease. J Neuropsychiatry Clin Neurosci. 2013;25:319–26.
human NREM slow waves and related hippocampus-dependent 43. Bliwise DL, Mercaldo ND, Avidan AY, et al. Sleep disturbance in
memory consolidation. Nat Neurosci 2015. Fascinating new data dementia with Lewy bodies and Alzheimer’s disease: a multicen-
demonstrates amyloid pathology with non-REM sleep ter analysis. Dement Geriatr Cogn Disord. 2011;31:239–46.
97 Page 10 of 11 Curr Psychiatry Rep (2015) 17:97

44. Boeve BF, Silber MH, Ferman TJ, et al. REM sleep behavior compared with those without sleep-disordered breathing.
disorder and degenerative dementia: an association likely Possible mechanisms could include hypoxia, sleep fragmenta-
reflecting Lewy body disease. Neurology. 1998;51:363–70. tion, or sleep duration.
45. Elwood PC, Bayer AJ, Fish M, et al. Sleep disturbance and day- 66. Yoon SS, Jo SA. Mechanisms of amyloid-beta peptide clearance:
time sleepiness predict vascular dementia. J Epidemiol potential therapeutic targets for Alzheimer’s disease. Biomol Ther
Community Health. 2011;65:820–4. (Seoul). 2012;20:245–55.
46. Pollak CP, Perlick D, Linsner JP, et al. Sleep problems in the 67. Mayo MC, Bordelon Y. Dementia with Lewy bodies. Semin
community elderly as predictors of death and nursing home place- Neurol. 2014;34:182–8.
ment. J Community Health. 1990;15:123–35. 68. Heidebrink JL. Is dementia with Lewy bodies the second most
47. Kim YH, Choi SH, D’Avanzo C et al. A 3D human neural cell common cause of dementia? J Geriatr Psychiatry Neurol.
culture system for modeling Alzheimer’s disease. In: Nat Protoc. 2002;15:182–7.
2015/06/13. 2015. pp. 985–1006. 69. Gagnon JF, Bertrand JA, Genier MD. Cognition in rapid eye
48. Ringman JM, Goate A, Masters CL, et al. Genetic heterogeneity in movement sleep behavior disorder. Front Neurol. 2012;3:82.
Alzheimer disease and implications for treatment strategies. Curr 70. Kosaka K. Diffuse Lewy body disease. Neuropathology.
Neurol Neurosci Rep. 2014;14:499. 2000;20(Suppl):S73–8.
49. Del Ser T, Hachinski V, Merskey H, Munoz DG. Alzheimer’s 71. Kosaka K. Lewy body disease and dementia with Lewy bodies.
disease with and without cerebral infarcts. J Neurol Sci. Proc Jpn Acad Ser B Phys Biol Sci. 2014;90:301–6.
2005;231:3–11. 72. Benjamin R, Leake A, Edwardson JA, et al. Apolipoprotein E
50. Jahn H. Memory loss in Alzheimer’s disease. Dialogues Clin genes in Lewy body and Parkinson’s disease. Lancet. 1994;343:
Neurosci. 2013;15:445–54. 1565.
51. Nikolaev A, McLaughlin T, O’Leary DD, Tessier-Lavigne M. 73. Gardner RC, Valcour V, Yaffe K. Dementia in the oldest old: a
APP binds DR6 to trigger axon pruning and neuron death via multi-factorial and growing public health issue. Alzheimers Res
distinct caspases. Nature. 2009;457:981–9. Ther. 2013;5:27.
52. Heneka MT, Carson MJ, El Khoury J, et al. Neuroinflammation in 74. Mata IF, Samii A, Schneer SH, et al. Glucocerebrosidase gene
Alzheimer’s disease. Lancet Neurol. 2015;14:388–405. mutations: a risk factor for Lewy body disorders. Arch Neurol.
53. Lim AS, Yu L, Kowgier M, et al. Modification of the relationship 2008;65:379–82.
of the apolipoprotein E epsilon4 allele to the risk of Alzheimer
75. Nalls MA, Duran R, Lopez G, et al. A multicenter study of
disease and neurofibrillary tangle density by sleep. JAMA Neurol.
glucocerebrosidase mutations in dementia with Lewy bodies.
2013;70:1544–51.
JAMA Neurol. 2013;70:727–35.
54. O’Hara R, Schroder CM, Kraemer HC, et al. Nocturnal sleep
76. Inoue Y, Sasai T, Hirata K. Electroencephalographic finding in
apnea/hypopnea is associated with lower memory performance
idiopathic REM sleep behavior disorder. Neuropsychobiology.
in APOE epsilon4 carriers. Neurology. 2005;65:642–4.
2015;71:25–33.
55. Osorio RS, Ayappa I, Mantua J, et al. Interaction between sleep-
77. Claassen DO, Josephs KA, Ahlskog JE, et al. REM sleep behavior
disordered breathing and apolipoprotein E genotype on cerebro-
disorder preceding other aspects of synucleinopathies by up to half
spinal fluid biomarkers for Alzheimer’s disease in cognitively nor-
a century. Neurology. 2010;75:494–9.
mal elderly individuals. Neurobiol Aging. 2014;35:1318–24.
56. Moraes W, Poyares D, Sukys-Claudino L, et al. Donepezil im- 78. Munhoz RP, Teive HA. REM sleep behaviour disorder: how use-
proves obstructive sleep apnea in Alzheimer disease: a double- ful is it for the differential diagnosis of parkinsonism? Clin Neurol
blind, placebo-controlled study. Chest. 2008;133:677–83. Neurosurg. 2014;127:71–4.
57. Sukys-Claudino L, Moraes W, Guilleminault C, et al. Beneficial 79. Vendette M, Gagnon JF, Decary A, et al. REM sleep behavior
effect of donepezil on obstructive sleep apnea: a double-blind, disorder predicts cognitive impairment in Parkinson disease with-
placebo-controlled clinical trial. Sleep Med. 2012;13:290–6. out dementia. Neurology. 2007;69:1843–9.
58. Moran M, Lynch CA, Walsh C, et al. Sleep disturbance in mild to 80. Latreille V, Carrier J, Lafortune M, et al. Sleep spindles in
moderate Alzheimer’s disease. Sleep Med. 2005;6:347–52. Parkinson’s disease may predict the development of dementia.
59. Iranzo A, Isetta V, Molinuevo JL, et al. Electroencephalographic Neurobiol Aging. 2015;36:1083–90.
slowing heralds mild cognitive impairment in idiopathic REM 81. Schenck CH, Bundlie SR, Mahowald MW. Delayed emergence of
sleep behavior disorder. Sleep Med. 2010;11:534–9. a parkinsonian disorder in 38% of 29 older men initially diagnosed
60. Dowling GA, Burr RL, Van Someren EJ, et al. Melatonin and with idiopathic rapid eye movement sleep behaviour disorder.
bright-light treatment for rest-activity disruption in institutional- Neurology. 1996;46:388–93.
ized patients with Alzheimer’s disease. J Am Geriatr Soc. 82. Ondo WG, Vuong KD, Jankovic J. Exploring the relationship
2008;56:239–46. between Parkinson disease and restless legs syndrome. Arch
61. Ancoli-Israel S, Poceta JS, Stepnowsky C, et al. Identification and Neurol. 2002;59:421–4.
treatment of sleep problems in the elderly. Sleep Med Rev. 1997;1: 83. Ferini-Strambi L, Marelli S, Galbiati A, et al. REM sleep behavior
3–17. disorder (RBD) as a marker of neurodegenerative disorders. Arch
62. Bombois S, Derambure P, Pasquier F, Monaca C. Sleep disorders Ital Biol. 2014;152:129–46.
in aging and dementia. J Nutr Health Aging. 2010;14:212–7. 84. Roman GC. Vascular dementia. Advances in nosology, diagnosis,
63. Ju YE, McLeland JS, Toedebusch CD, et al. Sleep quality and treatment and prevention. Panminerva Med. 2004;46:207–15.
preclinical Alzheimer disease. JAMA Neurol. 2013;70:587–93. 85. Aharon-Peretz J, Masiah A, Pillar T, et al. Sleep-wake cycles in
64. Beaulieu-Bonneau S, Hudon C. Sleep disturbances in older adults multi-infarct dementia and dementia of the Alzheimer type.
with mild cognitive impairment. Int Psychogeriatr. 2009;21:654– Neurology. 1991;41:1616–9.
66. 86. Elwood P, Hack M, Pickering J, et al. Sleep disturbance, stroke,
65.•• Yaffe K, Laffan AM, Harrison SL, et al. Sleep-disordered breath- and heart disease events: evidence from the Caerphilly cohort. J
ing, hypoxia, and risk of mild cognitive impairment and dementia Epidemiol Community Health. 2006;60:69–73.
in older women. JAMA. 2011;306:613–9. Important data dem- 87. Merrilees J, Hubbard E, Mastick J, et al. Sleep in persons with
onstrating that older women with sleep-disordered breathing frontotemporal dementia and their family caregivers. Nurs Res.
had an increased risk of developing cognitive impairment, 2014;63:129–36.
Curr Psychiatry Rep (2015) 17:97 Page 11 of 11 97

88. Anderson KN, Hatfield C, Kipps C, et al. Disrupted sleep and routine management of sleep problems in AD, including the
circadian patterns in frontotemporal dementia. Eur J Neurol. benzodiazepine and non-benzodiazepine hypnotics and noting
2009;16:317–23. lack of evidence to support the use of melatonin in patients
89. Yamakawa M, Shigenobu K, Makimoto K, et al. Environmental with AD, patients who present with sleep disturbances in the
control interventions for frontotemporal dementia with reversed setting of moderate to severe dementia.
sleep-wake cycles. Am J Alzheimers Dis Other Demen. 98. Medeiros CA, Carvalhedo de Bruin PF, Lopes LA, et al. Effect of
2008;23:470–6. exogenous melatonin on sleep and motor dysfunction in
90. Peter-Derex L, Yammine P, Bastuji H, Croisile B. Sleep and Parkinson’s disease. A randomized, double blind, placebo-
Alzheimer’s disease. Sleep Med Rev. 2015;19:29–38. controlled study. J Neurol. 2007;254:459–64.
91. Lack L, Wright H. The effect of evening bright light in delaying 99. Wade AG, Farmer M, Harari G, et al. Add-on prolonged-release
the circadian rhythms and lengthening the sleep of early morning melatonin for cognitive function and sleep in mild to moderate
awakening insomniacs. Sleep. 1993;16:436–43. Alzheimer’s disease: a 6-month, randomized, placebo-controlled,
92. McCurry SM, Pike KC, Vitiello MV, et al. Increasing walking and multicenter trial. Clin Interv Aging. 2014;9:947–61.
bright light exposure to improve sleep in community-dwelling 100.•• Videnovic A, Lazar AS, Barker RA, Overeem S. BThe clocks that
persons with Alzheimer’s disease: results of a randomized, con- time us^—circadian rhythms in neurodegenerative disorders. Nat
trolled trial. J Am Geriatr Soc. 2011;59:1393–402. Rev Neurol. 2014;10:683–93. A very elegant review examining
93. Riemersma-van der Lek RF, Swaab DF, Twisk J, et al. Effect of whether circadian disturbances could represent cause or a con-
bright light and melatonin on cognitive and noncognitive function sequence of neurodegeneration. The investigators argue that
in elderly residents of group care facilities: a randomized con- AD, PD, and HD share an underlying Bchronodegeneration^
trolled trial. JAMA. 2008;299:2642–55. as a common feature. The authors conclude that novel interven-
94. Deschenes CL, McCurry SM. Current treatments for sleep distur- tions targeting the chronodegeneration may help improve
bances in individuals with dementia. Curr Psychiatry Rep. symptoms, quality of life, and perhaps even delay or slow down
2009;11:20–6. disease progression.
95. Sloane PD, Williams CS, Mitchell CM, et al. High-intensity envi- 101. Asai H, Hirano M, Furiya Y, et al. Cerebrospinal fluid-orexin
ronmental light in dementia: effect on sleep and activity. J Am levels and sleep attacks in four patients with Parkinson’s disease.
Geriatr Soc. 2007;55:1524–33. Clin Neurol Neurosurg. 2009;111:341–4.
96. Lin L, Huang QX, Yang SS, et al. Melatonin in Alzheimer’s dis-
102. Mahowald MW, Schenck CH. REM sleep behavior disorder.
ease. Int J Mol Sci. 2013;14:14575–93.
Philadelphia: WB Saunders; 1994.
97.• McCleery J, Cohen DA, Sharpley AL. Pharmacotherapies for
sleep disturbances in Alzheimer’s disease. Cochrane Database 103. Takeuchi N, Uchimura N, Hashizume Y, et al. Melatonin therapy
Syst Rev. 2014;3:CD009178. An extensive meta-analysis noting for REM sleep behavior disorder. Psychiatry Clin Neurosci.
a lack of evidence to help direct pharmacological interventions 2001;55:267–9.
for the management of sleep problems in AD. Authors note the 104. Boeve B. Melatonin for treatment of REM sleep behavior disor-
lack of RCTs examining therapies commonly used in the der: response in 8 patients. Sleep. 2001;24(suppl):A35.

You might also like