Curr Neurol Neurosci Rep (2015) 15:8 DOI 10.1007/s11910-015-0525-5

SLEEP (M THORPY, M BILLIARD, SECTION EDITORS)

Parkinson’s Disease and Sleep/Wake Disturbances Keisuke Suzuki & Masayuki Miyamoto & Tomoyuki Miyamoto & Koichi Hirata

# Springer Science+Business Media New York 2015

Abstract Sleep disturbances are a common non-motor feature in patients with Parkinson’s disease (PD). Early diagnosis and appropriate management are imperative for enhancing patient quality of life. Sleep disturbances can be caused by multiple factors in addition to age-related changes in sleep, such as nocturnal motor symptoms (rigidity, resting tremor, akinesia, tardive dyskinesia, and the “wearing off” phenomenon), nonmotor symptoms (pain, hallucination, and psychosis), nocturia, and medication. Disease-related pathology involving the brainstem and changes in the neurotransmitter systems (norepinephrine, serotonin, and acetylcholine) responsible for regulating sleep structure and the sleep/wake cycle play a role in emerging excessive daytime sleepiness and sleep disturbances. Additionally, screening for sleep apnea syndrome, rapid eye movement sleep behavior disorder, and restless legs syndrome is clinically important. Questionnaire-based assessment utilizing the PD Sleep Scale-2 is useful for screening PD-related nocturnal symptoms. In this review, we focus on the current understanding and management of sleep disturbances in PD. Keywords Parkinson’s disease . Insomnia . Excessive daytime sleepiness . Restless legs syndrome . Rapid eye movement sleep behavior disorder . Sleep apnea syndrome This article is part of the Topical Collection on Sleep K. Suzuki (*) : K. Hirata Department of Neurology, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsuga, Tochigi 321-0293, Japan e-mail: [email protected] M. Miyamoto School of Nursing & Department of Neurology, Dokkyo Medical University, Tochigi, Japan T. Miyamoto Department of Neurology, Dokkyo Medical University Koshigaya Hospital, Saitama, Japan

Introduction Sleep disturbances are common disabling non-motor features of Parkinson’s disease (PD) that have a detrimental effect on health-related quality of life [1, 2•]. The cause of sleep disturbances in PD is multifactorial and overlapping (Table 1). The reported prevalence of sleep disturbances in PD varies depending on differences in the study population and methodology, with values ranging from approximately 40 to 90 % [3–6]. The frequency of sleep disturbances increases in parallel with disease progression, reflecting the impact of nocturnal motor and non-motor symptoms on sleep; however, sleep disturbances can occur at any time during the course of PD and even in the early stages. The presence of sleep disorders may predict the presence of more comorbid non-motor symptoms and reduced quality of life [7, 8]. Sleep disturbances are also associated with reduced quality of life in early-stage PD patients [2•]. Surprisingly, in a multicenter international study examining non-motor symptoms, sleep problems, such as insomnia and daytime sleepiness, were undeclared by approximately half of PD patients [9]. Some PD patients show motor improvement upon awakening in the morning and prior to taking medication. This phenomenon is called sleep benefit and is reported in 33–55 % of PD patients; however, the underlying mechanism and associated factors remain unclear [10]. Thus, comprehensively assessing sleep disturbances in PD patients and investigating possible causes are imperative for improving the quality of life of patients. In this review, we address sleep disturbances related to PD and present new information on comorbid primary sleep disorders in PD, such as rapid eye movement sleep behavior disorder (RBD), restless legs syndrome (RLS), and obstructive sleep apnea syndrome (OSAS).

8

Page 2 of 11

Table 1 Causes of sleep disturbances in Parkinson’s disease (adapted from Suzuki et al. [11]) 1. PD-related pathological changes Impairment of sleep architecture (REM and non-REM sleep) Impairment of the arousal system (orexin, serotonin, noradrenalin, acetylcholine, and dopamine) Impairment of the sleep-wake cycle, circadian rhythm sleep disorder, sundown syndrome 2. Nocturnal motor symptoms Wearing off phenomenon, rigidity, akinesia, tremor, medication-related dyskinesia, and dystonia 3. Nocturnal non-motor symptoms Neuropsychiatric symptoms (depression, psychosis, cognitive impairment) Sensory symptoms (pain, dysesthesia, restlessness of the arms or legs) Hallucinations Nightmares and vivid dreams Nocturia 4. Medication use Dopaminergic drugs, antipsychotics 5. Comorbid primary sleep disorders Sleep apnea syndrome REM sleep behavior disorder Restless legs syndrome, periodic limb movements

The Cause of Sleep Disturbances in PD Multiple factors and conditions related to PD are implicated in sleep disturbances in patients with PD (Table 1) [11]. Diseaserelated pathological changes include impairment in thalamocortical arousal and the degeneration of the brainstem regulatory centers for sleep/wakefulness maintenance and REM sleep, resulting in excessive daytime sleepiness and insomnia [12]. Additionally, sleep architecture may be altered in PD due to disease-related changes in the brainstem; the degeneration of cholinergic neurons in the basal forebrain and brainstem, including the pedunculopontine nucleus and noradrenergic neurons in the locus coeruleus, results in a reduction in REM sleep and RBD. A loss of serotonergic neurons in the raphe nucleus is associated with a decreased percentage of slow wave sleep [13]. Sleep disturbances also result from nocturnal motor symptoms, psychiatric symptoms, dementia, medication use, circadian rhythm sleep disorders, and comorbid primary sleep disorders such as SAS, RLS, and RBD. There are no firmly established treatments for PD-related sleep disturbances. Sleep problems other than nocturnal motor symptoms, insomnia, nightmares, and dopamine dysregulation syndrome may not respond well to dopaminergic treatment; however, increased severity in nocturnal motor symptoms, such as rigidity, bradykinesia, and resting tremor, may benefit from increasing the bedtime dose of dopaminergic treatment or introducing dopaminergic treatment at bedtime.

Curr Neurol Neurosci Rep (2015) 15:8

In contrast, nocturnal psychiatric symptoms including hallucinations and psychosis can be effectively treated by reducing the bedtime dose of dopaminergic treatment or adding antipsychotics. Table 2 shows treatments for sleep disturbances in patients with PD [11]. Readers may also refer to a recent review regarding the treatment of sleep disorders in PD [14].

Sleep Disturbances and Their Relationship with the Disease Course in PD Sleep disturbances increase as the disease progresses, resulting in severe nocturnal motor and non-motor symptoms. In PD patients, reduced total sleep time and sleep efficiency, increased stage 1 and 2 sleep, and decreased slow wave sleep are expected in the presence of various nocturnal motor and non-motor problems and disease-related changes involving sleep architecture. However, many studies report conflicting results of sleep architecture changes; these discrepancies are probably due to individual night-to-night variations, medication effects (dopaminergic drugs, antidepressants, and hypnotics), and differences in patient selection (disease severity). A recent review found no significant difference in sleep stage 1, 2, or slow wave sleep between PD patients and controls [15•]. Lees et al. [3] found that 98 % of 220 PD patients reported at least one sleep problem. The common problems were nocturia (79 %), inability to turn over in bed (65 %), painful leg cramps (55 %), and vivid dreams/nightmares (48 %). The most troublesome problem was inability to turn over in bed (39 %). In a community-based sleep study, sleep-onset insomnia, sleep maintenance insomnia, and early awakening were observed in 31.8, 38.9, and 23.4 % of PD patients compared with 22, 12, and 11 % of healthy controls, respectively [6]. Self-reported sleep problems occurred more frequently in patients with PD (60 %) than healthy controls (33 %) and patients with diabetes mellitus (45 %). Sleep maintenance insomnia is more common than sleep-onset insomnia in PD patients relative to healthy controls [16, 17], likely due to nocturnal motor symptoms (akinesia, tremor, rigidity, and wearing off) and non-motor symptoms (psychosis, depression, hallucination, and pain). Therefore, sleep problems are not surprisingly more common in patients in the more advanced stages than those in the early stages of PD. In contrast, in early, drug-naïve patients with PD, nocturia, nighttime cramps, dystonia, tremor, and daytime sleepiness are the most important nocturnal disabilities [18]. In another study, the Epworth Sleepiness Scale (ESS) score was abnormally high in one patient (out of 20), while short mean sleep latency was found in three other patients (out of 15), and REM sleep without atonia was a common finding; RBD was rare in early untreated PD patients [19]. Several tools are recommended for assessing sleep problems in patients with PD [20]. The PD Sleep Scale-2 (PDSS-

Curr Neurol Neurosci Rep (2015) 15:8 Table 2

Page 3 of 11 8

Management of sleep problems in patients with Parkinson’s disease (modified with permission from Suzuki et al. [11])

Type of insomnia

Cause

Treatment

Difficulty initiating sleep

Unknown cause

Difficulty maintaining sleep/early morning awakening

Drug-related (alerting effect) Unknown cause Wearing off, resting tremor, rigidity, akinesia

Hypnotics (short-acting type; eszopiclone, zolpidem, zopiclone, brotizolam) Remove or reduce dose of causative drug Hypnotics (intermediate type; flunitrazepam) Increase frequency of levodopa administration, add dopamine agonist, or switch to a different type of dopamine agonist Increase frequency of levodopa and reduce dose of levodopa administration, add dopamine agonist Antidepressant (SSRI, SNRI, tricyclic antidepressant) Antianxiety drug Dopamine agonist (D3 R) Oxybutynin, flavoxate Dopamine agonist (D1 R) Daytime rehabilitation Remove or reduce dose of causative drug (including dopamine agonist) Modafinil, caffeine Reduce dopaminergic drugs, consider Yi-Gan Sana and atypical antipsychotics Hazard avoidance (remove potentially dangerous objects from the bedroom and place a mattress on the floor), consider clonazepam and Yi-Gan Sana Continuous positive airway pressure therapy (severe case) Adjustment of dopaminergic treatment, use dopamine agonist and/or clonazepam prior to bedtime, consider iron supplement (if serum ferritin levels are below 50 μg/L) Gabapentin, pregabalin, gabapentin enacarbil, or opioids may be alternatively used

Drug-induced dyskinesia Depression, anxiety

Nocturia Excessive daytime sleepiness

Unknown cause Drug-related (sedative effect) Refractory

Hallucinations, delusions, delirium REM sleep behavior disorder

Sleep apnea syndrome Restless legs syndrome, periodic limb movements

a

Yi-Gan San is an herbal medicine that is used in Asia but that has not been approved by the FDA or EMA for use in the USA or Europe, respectively

2), consisting of 15 questions concerning various sleep and nocturnal disturbances and rated by the patients using one of five categories from 0 (never) to 4 (very frequent), can be used in outpatient clinics or at the bedside and is a useful screening tool for addressing PD-related sleep disturbances [21•]. The total score ranges from 0 (no disturbance) to 60 (maximum nocturnal disturbance). The PDSS-2 has been validated and translated into Japanese, and the PDSS-2 total score correlates with daytime sleepiness, depressive symptoms, quality of life, and motor scores [22]. The PDSS-2 could also be suitable for evaluating responses to dopaminergic treatment [23].

Excessive Daytime Sleepiness Excessive daytime sleepiness (EDS) is a common sleep problem observed in PD and affects 15 to 50 % of patients [24–26]. EDS can be caused by PD-related motor and non-motor nocturnal problems and disease-related changes in arousal

systems, including the cholinergic, serotoninergic, and noradrenergic systems, as well as the orexin system. Risk factors for EDS in PD have been reported to include male gender, longer disease duration, disease severity, and dopaminergic medication [24, 25, 27]. Genetic susceptibility to sleepiness in PD was considered; however, the effect of the COMT genotype on sleepiness was not confirmed in a subsequent study [28]. Cerebrospinal fluid (CSF) levels of orexin are decreased in patients with narcolepsy, a sleep disorder characterized by daytime sleepiness, cataplexy, hypnagogic hallucinations, and sleep paralysis. The multiple sleep latency test (MSLT) shows shorter sleep latency and emergence of REM sleep within 15 min after sleep onset, called the sleeponset REM period. Using the MSLT, a narcolepsy-like phenotype (≥2 sleep-onset REM periods) was demonstrated in a subset of patients in a study on 54 PD patients with sleepiness. Decreased orexin levels in the hypothalamus and a loss of orexin neurons have been observed in PD patients in correlation with disease progression [29, 30]. However, orexin levels

8

Page 4 of 11

in the cerebrospinal fluid of PD patients with EDS remain controversial [31–33]. In a prospective study including 232 PD patients, the EDS frequency increased from 5.6 % at baseline to 22.5 % at the 4year follow-up and to 40.8 % at the 8-year follow-up. EDS was related to age, gender, and the use of dopamine agonists in a logistic regression model, whereas in patients who had never used dopamine agonists, EDS was associated with the Hoehn and Yahr stage only, suggesting that disease-related changes in sleep-wake regulation and dopamine agonists contribute to EDS [34]. Additionally, daytime sleepiness measured using both the ESS and MSLT did not differ between drug-naïve PD patients and healthy controls, and increased daytime sleepiness emerged on both the ESS and MSLT after the introduction of dopaminergic treatment in PD patients [19, 35]; these studies support the dopaminergic medication effect on sleepiness in PD. In contrast, in a study of 134 patients with PD using ESS and MSLT, no difference was observed in type and dose of dopaminergic medication, disease severity, depression, insomnia, and nighttime sleep evaluation between the EDS and non-EDS groups, but pain complaint was more frequent and body mass index was higher in the EDS group than in the non-EDS group [36]. In animal studies, the effects of dopaminergic treatment on sleep are complicated by biphasic effects, i.e., a presynaptic sedative effect at low doses and a post-synaptic alerting effect at high doses [37]; however, in the clinic, all dopaminergic drugs have sedative effects on patients with PD. In contrast, Arnulf et al. [38] found a negative correlation between a daily dose of levodopa and the severity of sleepiness in PD patients. Divergent dosedependent effects of drug class on alertness in PD were reported; increasing dosages of dopamine agonists were associated with less daytime alertness, whereas higher levels of levodopa were associated with higher levels of alertness [39]. In PD patients, dopaminergic drugs are associated with EDS as a class effect, and the association between the specific type of dopamine agonist and EDS is unclear [40]. Sudden-onset sleep episodes while driving have been reported in 3.8–22.8 % of PD patients [24, 27, 41]. In patients with a higher ESS score (≥10), the frequency of sudden-onset sleep episodes is increased [25, 27]; therefore, screening for sudden-onset sleep episodes in patients with EDS is important. However, one should be aware that sudden-onset sleep episodes are sometimes unrecognized by patients.

Rapid Eye Movement Sleep Behavior Disorder Idiopathic RBD and PD-Related Diseases RBD was first described by Schenck in 1986 [42] and is characterized by a loss of normal muscle atonia during REM sleep and dream-enacting behavior. The clinical manifestation

Curr Neurol Neurosci Rep (2015) 15:8

includes vocalizations, such as talking and shouting, and violent, aggressive complex behavior during REM sleep in association with vivid, unpleasant, and violent dreams that can cause sleep-related injury to the patient or bed partner [43]. The prevalence of RBD in the general population is reported to be 0.5 % [44]. A link between RBD and neurodegenerative diseases has been suggested due to a novel observation by Schenck et al. [45], which demonstrated that 38 % of 29 male patients with idiopathic RBD developed PD 12.7±7.3 years after the onset of RBD. A 16-year-update from the initial study provided a conversion rate of 81 % of idiopathic RBD to neurodegenerative diseases (13 PD; 3 dementia with Lewy bodies (DLB); 2 multiple system atrophy (MSA); 1 dementia; and 2 Alzheimer’s disease (AD) with Lewy pathology) [46]. These intriguing findings have been reproduced by other investigators. Iranzo et al. [47] also found that 82 % of 44 patients with RBD developed neurodegenerative diseases over a 12-year follow-up (16 PD, 14 DLB, 1 MSA, and 5 mild cognitive impairment). Postuma et al. [48] reported that 28 % of 93 patients with idiopathic RBD developed neurodegenerative diseases (14 PD; 7 DLB; 4 AD; and 1 MSA) over a mean of 5.2 years of follow-up. The estimated risk for neurodegenerative diseases in RBD subjects was 17.7 % at 5 years of followup, 40.6 % at 10 years of follow-up, and 52.4 % at 12 years of follow-up. Furthermore, when including mild cognitive impairment, the risk of neurodegenerative diseases in RBD patients was 33.1 % at 5 years, 75.7 % at 10 years, and 90.9 % at 14 years [49]. In a clinicopathological study including 172 patients with or without coexisting neurological diseases, the neuropathological diagnoses were Lewy body disease (n= 77), combined Lewy body disease and AD (n=59), MSA (n=19), AD (n=6), progressive supranuclear palsy (n=2), and others (n = 9), demonstrating a high percentage of synucleinopathies (94 %) among RBD patients associated with neurodegenerative diseases (n=170) [50••]. Taken together, these longitudinal studies suggest that idiopathic RBD carries a substantial risk for developing synucleinopathies, particularly Lewy body-related diseases such as PD and DLB. Concomitant AD pathology has frequently been reported in patients with DLB. A study including pathologically confirmed DLB patients found a robust correlation between RBD and DLB pathology; the primary finding indicated that the presence of RBD was associated with a higher likelihood of DLB and less severe AD-related pathology in the medial temporal lobes, whereas the absence of RBD was associated with AD-like atrophy patterns on MRI and an increased phospho-tau burden [51••]. A recent multicenter international study showed that idiopathic RBD subjects have a positive family history of dreamenacting behavior [52], and the potential risk factors of idiopathic RBD include comorbid depression and antidepressant

Curr Neurol Neurosci Rep (2015) 15:8

use, ischemic heart disease [53], smoking, head injury, pesticide exposure, and farming [54]. Thus, whether antidepressant use causes secondary RBD or induces disease in subjects who are already susceptible to RBD remains unclear. The involvement of cholinergic and monoaminergic brainstem nuclei, including the pedunculopontine nuclei, laterodorsal tegmental nucleus, sublaterodorsal nuclei, and locus coeruleus as well as neuronal networks involving the limbic system and neocortex are thought to contribute to the occurrence of RBD [55]. The diagnosis of RBD in the latest version of the international classification of sleep disorders 3rd edition (ICSD-3) states the following [56]: “Criteria A-D must be met; A. Repeated episodes of sleep-related vocalization and/or complex motor behaviors; B. These behaviors are documented by polysomnography to occur during REM sleep or, based on clinical history of dream enactment, are presumed to occur during REM sleep; C. Polysomnographic recording demonstrates REM sleep without atonia (RWA); D. The disturbance is not better explained by another sleep disorder, mental disorder, medication, or substance use.” Antidepressants, such as tricyclics, selective serotonin reuptake inhibitors, and serotonin-norepinephrine reuptake inhibitors, as well as narcolepsy and brain lesions affecting the regulation of REM sleep, can cause secondary RBD. For the definition of RWA, see the AASM Manual [57]. When patients with a typical clinical history of dream-enacting behaviors also exhibit typical dream-enacting behaviors during vPSG but do not demonstrate sufficient RWA, RBD may be provisionally diagnosed. Because polysomnography (PSG) cannot be routinely performed, several screening tools for RBD have been reported to be useful in clinical practice. These include the RBD screening questionnaire [58, 59], Mayo sleep questionnaire [60], RBD questionnaire-Hong Kong [61], and Innsbruck RBD inventory [62]. RBD patients are able to recall the content of their dreams upon awakening at the time of the abnormal behavior, although details may be forgotten by noon of the following day [55]. Also of note, in patients with severe sleep apnea syndrome, apnea-induced arousals can mimic the symptoms of RBD [63]. Idiopathic RBD patients show clinical features and abnormal test results, including olfactory impairment; visual color impairment; subtle motor signs; increased systolic blood pressure drop after standing [64]; cognitive impairments, such as deficits in attention, executive functions, learning capacities, and visuospatial abilities [65]; cardiac autonomic dysfunction, including reductions in beat-to-beat RR variability [66] and u pt a ke o f c a r d i a c M I B G s ci n t i g r ap h y [ 67 ] ; a n d hyperechogenicity of the substantia nigra on transcranial sonography [68]. These features are also observed in Lewy body-related diseases, such as PD and DLB, supporting RBD as a forme fruste of Lewy body-related diseases. Therefore, identifying RBD subjects who are susceptible to the later

Page 5 of 11 8

development of neurodegenerative disease is of clinical importance for possible future disease-modifying therapy. Postuma et al. [69] reported that in idiopathic RBD subjects, increased tonic chin EMG activity during REM sleep on baseline PSG findings could predict the future development of PD. In a follow-up positron emission tomography study of a patient with idiopathic RBD, nigrostriatal presynaptic dopaminergic function was almost normal except for a slight decrease in the left posterior putamen at 1 year after the onset of RBD symptoms but was decreased by 4–6 % per year at 3.5 years after the RBD onset [70]. In a 2.5-year follow-up study of 43 idiopathic RBD subjects, of all of the subjects with reduced striatal dopamine transporter uptake and/or substantia nigra hyperechogenicity at baseline, 8 subjects developed neurodegenerative diseases (5 PD, 2 DLB, and 1 MSA), while the idiopathic RBD subjects with normal neuroimaging results at baseline remained disease-free [71]. Prior to the onset of clinically evident parkinsonism in idiopathic RBD, a Unified PD Rating Scale motor score >4 can predict prodromal parkinsonism with 88 % sensitivity and 94 % specificity 2 years prior to PD diagnosis, and other quantitative motor examinations can detect parkinsonism earlier [72]. Subtle gait changes, increased stride-to-stride variability, and decreased rhythmicity and velocity have been detected in idiopathic RBD patients in a cross-sectional study [73], which may be early signs predicting parkinsonism. Parkinson’s Disease and RBD The frequency of RBD in PD patients has been reported to be 15–60 % [43, 74] with RBD symptoms varying over time [75]. Generally, RBD frequency increases when diagnosed using PSG compared with diagnosis using clinical history without PSG because PSG can detect mild forms of RBD and abnormal behavior, which the patient does not remember upon awakening. Bolitho et al. [76] used different questionnaires to compare RWA on PSG findings in PD patients and found that PD patients who were positive for RBD on questionnaires specific to dream enactment (and not the total scores of these questionnaires) were correctly identified with higher degrees of RWA, improving the diagnostic accuracy of the questionnaires. RBD preceded the motor manifestation of PD in up to half of PD patients [77]. Gong et al. [78] reported that among PD patients with RBD, clinical features did not differ between the RBD-preceding and RBD non-preceding group. However, Gong et al. found that the RBD non-preceding subgroup had a significantly higher Unified PD Rating Scale part I score, lower quality of life score, and a lower apnea-hypopnea index during REM than PD patients in the non-RBD group. Nihei et al. [79] found that the RBD-preceding group had a higher onset age of PD, shorter PD duration, and lower levodopa equivalent dose compared with the RBD-non-preceding

8

Page 6 of 11

group. RBD comorbidity in PD patients has been associated with distinct characteristics of PD, including an akinetic rigid phenotype, increased fall frequency, poor responses to dopaminergic medications, orthostatic hypotension, and impaired color vision [80, 81]. However, the clinical motor subtype of PD associated with RBD remains controversial; several studies failed to find a clinical motor subtype that differs between PD patients with and without RBD [82, 83]. Gagnon et al. reported that mild cognitive impairment was found in 50 % of idiopathic RBD patients and 73 % of PD patients with RBD, while mild cognitive impairment was observed in 11 % of PD patients without RBD and 8 % of control subjects with mild cognitive impairment. These results showed an increased presence of mild cognitive impairment in idiopathic RBD patients and PD patients with RBD [84]. RBD was associated with mild cognitive impairment among drug-naïve patients with PD [85]. In contrast, in another study, 30 % of 57 newly diagnosed patients with PD had PSG-confirmed RBD, and non-RBD patients and RBD patients did not differ with respect to age, gender, disease duration, motor symptom subtype, and severity or cognitive performance [86]. A 4-year prospective study found that RBD at baseline was associated with increased risks of hallucination, cognitive fluctuations, and dementia in patients with PD [87]. A decreased uptake of cardiac MIBG scintigraphy was reported in PD patients with RBD compared with those without RBD [88, 89]. Among PD patients, the frequency of RBD was significantly higher in patients with older PD onset (>40 years) than the younger onset group (≤40 years) (24.8 vs. 7.8 %). PD patients with RBD had a more advanced stage, poorer sleep quality, and more frequent daytime sleepiness than those without RBD [90]. The presence of RBD in PD patients was also associated with a poorer quality of life [8, 80, 83] and increased frequencies of non-motor symptoms such as depression, fatigue, and sleep disturbances [91]. A study using neuromelanin-sensitive, structural and diffusion magnetic resonance imaging revealed a reduced signal intensity in the locus coeruleus/ subcoeruleus area, involved in the control of atonia during REM sleep, in PD patients with RBD compared with those without RBD and healthy volunteers [92].

Curr Neurol Neurosci Rep (2015) 15:8

specific to patients with PD. The relationship between RLS and PD has been investigated due to the dopaminergic dysfunction and the favorable response to dopaminergic agents observed in both diseases. However, the results of dopamine imaging studies in the brain are inconsistent. A transcranial sonography study revealed that the substantia nigra echogenicity was low in idiopathic RLS but high in PD patients [94]. Severe RLS symptoms (>15 times/month) in men were associated with the development of PD [95]. However, in approximately 70–95 % of PD patients, RLS develops after the onset of PD [96–98], and there is currently no evidence suggesting RLS as a risk for developing PD [99, 100]. Several studies have reported increased RLS comorbidity in PD patients [98, 100], but the reported prevalence of RLS in PD varies, ranging from 0 to 50 %, possibly depending on the method and patient selection used [101]. PD patients with comorbid RLS are reported to have less family history of RLS, older RLS onset [97], longer PD duration, more severe disability, and longer dopaminergic treatment duration [102, 103]. Older age [104] and younger age of PD onset [96, 98] were both related to RLS comorbidity. Additionally, Peralta et al. [98] reported that symptoms relating to the urge to move the legs and unpleasant sensations were associated with wearing off in 61 % of RLS-positive patients with PD. Many PD patients suffer from motor restlessness and sensory symptoms related to parkinsonism; these symptoms may mimic RLS, and dopaminergic treatment for PD may unmask subclinical RLS [101]. To eliminate confounding factors related to dopaminergic treatment in PD, several studies have assessed RLS prevalence in untreated PD patients; two studies (a clinical case–control study [105] and a population-based incident cohort study [106]) found no significant increase in RLS prevalence in PD patients. Shin et al. [107] reported increased RLS prevalence in drug-naïve PD patients (16.5 %) compared with that in the general population, but no controls were included in this study. Although RLS may not be prevalent, leg motor restlessness that does not fulfill the RLS criteria may be more common in untreated patients with PD than in controls [106].

Sleep Apnea Syndrome Restless Legs Syndrome RLS is a sensorimotor disorder characterized by unpleasant sensations in the legs [93]. Four essential criteria include an urge to move the legs, symptoms beginning or worsening during periods of rest or inactivity, relief by movement, and predominant occurrence in the evening or night rather than during the day. Additionally, the impact of RLS on daytime functioning and sleep problems were added to the latest ICSD-3 [56]. However, these RLS criteria pertain to the general population, and there are currently no RLS criteria

Young et al. [108] reported that habitual snorers with an apnea–hypopnea index (AHI) 5) was less frequent in 100 PD patients (50 unselected patients and 50 patients referred for sleepiness) compared with 50 in-hospital controls (27 vs. 40 %). Additionally, in the PD group, sleep apnea was not associated with daytime sleepiness, nocturia, depression, cognitive impairment, or cardiovascular events. Trotti and Bliwise [112] examined the frequency and severity of OSA in 55 PD patients compared with a large sample of normative data from the Sleep Heart Health Study (n=6,132) and found no increased risk of OSA in PD patients (AHI 5–14.9, PD 29.1 % vs. controls 28.6 %; AHI 15–29.9, 10.9 vs. 11.7 %; AHI ≥30, 3.6 vs. 6.1 %). Milder nocturnal desaturation was observed in PD with OSA patients compared with OSA controls [113–115], as was a blunted sympathetic response to OSA [116]; this may suggest that these autonomic disturbances could protect PD patients from cardiovascular diseases. Disease severity is not universally associated with OSA severity [110, 117]. These findings imply that OSA may not be a relevant issue in PD; however, a randomized, placebo-controlled, crossover study showed that in PD patients with an AHI ≥10, continuous positive airway pressure therapy improved not only sleep apnea but also daytime sleepiness, as measured using the multiple sleep latency test [118]. Therefore, OSA patients with PD, particularly in severe cases, are recommended to receive appropriate treatment [36].

Page 7 of 11 8

References Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance 1.

2.•

3. 4.

5.

6.

7.

8.

9.

Conclusion We here review the current understanding of sleep problems in PD patients. Management should consider the impacts of sleep problems on motor and non-motor symptoms as well as quality of life. Compliance with Ethics Guidelines

10.

11.

12.

Conflict of Interest Keisuke Suzuki, Masayuki Miyamoto, Tomoyuki Miyamoto, and Koichi Hirata declare that they have no conflict of interest.

13.

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.

14.

Avidan A, Hays RD, Diaz N, Bordelon Y, Thompson AW, Vassar SD, et al. Associations of sleep disturbance symptoms with healthrelated quality of life in Parkinson's disease. J Neuropsychiatry Clin Neurosci. 2013;25(4):319–26. doi:10.1176/appi. neuropsych.12070175. Duncan GW, Khoo TK, Yarnall AJ, O'Brien JT, Coleman SY, Brooks DJ, et al. Health-related quality of life in early Parkinson's disease: the impact of nonmotor symptoms. Mov Disord. 2014;29(2):195–202. doi:10.1002/mds.25664. This study demonstrated a significant impact of non-motor symptoms, such as insomnia, depression, anxiety and memory disturbance, in 158 patients with newly diagnosed PD. Lees AJ, Blackburn NA, Campbell VL. The nighttime problems of Parkinson's disease. Clin Neuropharmacol. 1988;11(6):512–9. Sixel-Döring F, Schweitzer M, Mollenhauer B, Trenkwalder C. Intraindividual variability of REM sleep behavior disorder in Parkinson's disease: a comparative assessment using a new REM sleep behavior disorder severity scale (RBDSS) for clinical routine. J Clin Sleep Med. 2011;7(1):75–80. Kumar S, Bhatia M, Behari M. Sleep disorders in Parkinson's disease. Mov Disord. 2002;17(4):775–81. doi:10.1002/mds. 10167. Tandberg E, Larsen JP, Karlsen K. A community-based study of sleep disorders in patients with Parkinson's disease. Mov Disord. 1998;13(6):895–9. doi:10.1002/mds.870130606. Neikrug AB, Maglione JE, Liu L, Natarajan L, Avanzino JA, Corey-Bloom J, et al. Effects of sleep disorders on the nonmotor symptoms of Parkinson disease. J Clin Sleep Med. 2013;9(11):1119–29. doi:10.5664/jcsm.3148. Rolinski M, Szewczyk-Krolikowski K, Tomlinson PR, Nithi K, Talbot K, Ben-Shlomo Y, et al. REM sleep behaviour disorder is associated with worse quality of life and other non-motor features in early Parkinson's disease. J Neurol Neurosurg Psychiatry. 2014;85(5):560–6. doi:10.1136/jnnp-2013-306104. Chaudhuri KR, Prieto-Jurcynska C, Naidu Y, Mitra T, FradesPayo B, Tluk S, et al. The nondeclaration of nonmotor symptoms of Parkinson's disease to health care professionals: an international study using the nonmotor symptoms questionnaire. Mov Disord. 2010;25(6):704–9. doi:10.1002/mds.22868. van Gilst MM, Bloem BR, Overeem S. "Sleep benefit" in Parkinson's disease: a systematic review. Parkinsonism Relat Disord. 2013;19(7):654–9. doi:10.1016/j.parkreldis.2013.03.014. Suzuki K, Miyamoto T, Miyamoto M, Numao A, Sakuta H, Fujita H et al. Sleep Disturbances in Patients with Parkinson’s Disease, A Synopsis of Parkinson's Disease, Dr. Abdul Qayyum Rana (Ed.), ISBN: 978-953-51-1229-7, InTech, 2014. doi:10.5772/57320. Diederich NJ, McIntyre DJ. Sleep disorders in Parkinson's disease: many causes, few therapeutic options. J Neurol Sci. 2012;314(1–2):12–9. doi:10.1016/j.jns.2011.10.025. Diederich NJ, Comella CL. Sleep disturbances in Parkinson’s disease. In: Chokroverty S, Hening WA, Walters AS, editors. Sleep and movement disorders. Philadelphia: Elsevier Science; 2003. p. 478–88. Trotti LM, Bliwise DL. Treatment of the sleep disorders associated with Parkinson's disease. Neurotherapeutics : J Am Soc Exp NeuroTher. 2014;11(1):68–77. doi:10.1007/s13311-013-0236-z.

8

Page 8 of 11

15.• Peeraully T, Yong MH, Chokroverty S, Tan EK. Sleep and Parkinson's disease: a review of case–control polysomnography studies. Mov Disord. 2012;27(14):1729–37. doi:10.1002/mds. 25197. Peeraully et al. provided a review result of case–control polysomnographic studies, which found a higher prevalence of RBD in PD patients than controls; however, the incidence of obstructive sleep apnea did not differ between controls and PD patients. Subjective sleepiness was greater in PD patients than controls, but objective sleepiness, measured using multiple sleep latency tests, did not increase in PD patients. 16. Chaudhuri KR, Pal S, DiMarco A, Whately-Smith C, Bridgman K, Mathew R, et al. The Parkinson's disease sleep scale: a new instrument for assessing sleep and nocturnal disability in Parkinson's disease. J Neurol Neurosurg Psychiatry. 2002;73(6): 629–35. 17. Suzuki K, Okuma Y, Hattori N, Kamei S, Yoshii F, Utsumi H, et al. Characteristics of sleep disturbances in Japanese patients with Parkinson's disease. A study using Parkinson's disease sleep scale. Mov Disord. 2007;22(9):1245–51. doi:10.1002/mds.21257. 18. Dhawan V, Dhoat S, Williams AJ, Dimarco A, Pal S, Forbes A, et al. The range and nature of sleep dysfunction in untreated Parkinson's disease (PD). A comparative controlled clinical study using the Parkinson's disease sleep scale and selective polysomnography. J Neurol Sci. 2006;248(1–2):158–62. doi:10. 1016/j.jns.2006.05.004. 19. Buskova J, Klempir J, Majerova V, Picmausova J, Sonka K, Jech R, et al. Sleep disturbances in untreated Parkinson's disease. J Neurol. 2011. doi:10.1007/s00415-011-6109-7. 20. Högl B, Arnulf I, Comella C, Ferreira J, Iranzo A, Tilley B, et al. Scales to assess sleep impairment in Parkinson's disease: critique and recommendations. Mov Disord. 2011;25(16):2704–16. doi: 10.1002/mds.23190. 21.• Trenkwalder C, Kohnen R, H gl B, Metta V, Sixel-Doring F, Frauscher B, et al. Parkinson's disease sleep scale-validation of the revised version PDSS-2. Mov Disord. 2011;26(4):644–52. doi:10.1002/mds.23476. Trenkwalder et al. developed the revised version of the PD sleep scale, the PDSS-2, which is the bedside screening tool for sleep problems related to PD. Applying the PDSS-2 may be useful for detecting and managing PD-related nocturnal symptoms. 22. Suzuki K, Miyamoto M, Miyamoto T, Tatsumoto M, Watanabe Y, Suzuki S, et al. Nocturnal disturbances and restlessness in Parkinson's disease: using the Japanese version of the Parkinson's disease sleep scale-2. J Neurol Sci. 2012;318(1–2): 76–81. doi:10.1016/j.jns.2012.03.022. 23. Trenkwalder C, Kies B, Rudzinska M, Fine J, Nikl J, Honczarenko K, et al. Rotigotine effects on early morning motor function and sleep in Parkinson's disease: a double-blind, randomized, placebo-controlled study (RECOVER). Mov Disord. 2011;26(1):90–9. doi:10.1002/mds.23441. 24. Ondo WG, Dat Vuong K, Khan H, Atassi F, Kwak C, Jankovic J. Daytime sleepiness and other sleep disorders in Parkinson's disease. Neurology. 2001;57(8):1392–6. 25. Suzuki K, Miyamoto T, Miyamoto M, Okuma Y, Hattori N, Kamei S, et al. Excessive daytime sleepiness and sleep episodes in Japanese patients with Parkinson's disease. J Neurol Sci. 2008;271(1–2):47–52. doi:10.1016/j.jns.2008.03.008. 26. Tandberg E, Larsen JP, Karlsen K. Excessive daytime sleepiness and sleep benefit in Parkinson's disease: a community-based study. Mov Disord. 1999;14(6):922–7. 27. Tan EK, Lum SY, Fook-Chong SM, Teoh ML, Yih Y, Tan L, et al. Evaluation of somnolence in Parkinson's disease: comparison with age- and sex-matched controls. Neurology. 2002;58(3):465–8. 28. Rissling I, Frauscher B, Kronenberg F, Tafti M, Stiasny-Kolster K, Robyr AC, et al. Daytime sleepiness and the COMT val158met

Curr Neurol Neurosci Rep (2015) 15:8 polymorphism in patients with Parkinson disease. Sleep. 2006;29(1):108–11. 29. Fronczek R, Overeem S, Lee SY, Hegeman IM, van Pelt J, van Duinen SG, et al. Hypocretin (orexin) loss in Parkinson's disease. Brain. 2007;130(Pt 6):1577–85. doi:10.1093/brain/awm090. 30. Thannickal TC, Lai YY, Siegel JM. Hypocretin (orexin) cell loss in Parkinson's disease. Brain. 2007;130(Pt 6):1586–95. doi:10. 1093/brain/awm097. 31. Overeem S, Scammell TE, Lammers GJ. Hypocretin/orexin and sleep: implications for the pathophysiology and diagnosis of narcolepsy. Curr Opin Neurol. 2002;15(6):739–45. doi:10.1097/01. wco.0000044800.53746.5a. 32. Yasui K, Inoue Y, Kanbayashi T, Nomura T, Kusumi M, Nakashima K. CSF orexin levels of Parkinson's disease, dementia with Lewy bodies, progressive supranuclear palsy and corticobasal degeneration. J Neurol Sci. 2006;250(1–2):120–3. doi:10.1016/j.jns.2006.08.004. 33. Compta Y, Santamaria J, Ratti L, Tolosa E, Iranzo A, Munoz E, et al. Cerebrospinal hypocretin, daytime sleepiness and sleep architecture in Parkinson's disease dementia. Brain. 2009;132(Pt 12):3308–17. doi:10.1093/brain/awp263. 34. Gjerstad MD, Alves G, Wentzel-Larsen T, Aarsland D, Larsen JP. Excessive daytime sleepiness in Parkinson disease: is it the drugs or the disease? Neurology. 2006;67(5):853–8. doi:10.1212/01. wnl.0000233980.25978.9d. 35. Kaynak D, Kiziltan G, Kaynak H, Benbir G, Uysal O. Sleep and sleepiness in patients with Parkinson's disease before and after dopaminergic treatment. Eur J Neurol. 2005;12(3):199–207. doi: 10.1111/j.1468-1331.2004.00971.x. 36. Cochen De Cock V, Bayard S, Jaussent I, Charif M, Grini M, Langenier MC, et al. Daytime sleepiness in Parkinson's disease: a reappraisal. PLoS One. 2014;9(9):e107278. doi:10.1371/ journal.pone.0107278. 37. Arnulf I, Leu-Semenescu S. Sleepiness in Parkinson's disease. Parkinsonism Relat Disord. 2009;15 Suppl 3:S101–4. doi:10. 1016/S1353-8020(09)70792-8. 38. Arnulf I, Konofal E, Merino-Andreu M, Houeto JL, Mesnage V, Welter ML, et al. Parkinson's disease and sleepiness: an integral part of PD. Neurology. 2002;58(7):1019–24. 39. Bliwise DL, Trotti LM, Wilson AG, Greer SA, Wood-Siverio C, Juncos JJ, et al. Daytime alertness in Parkinson's disease: potentially dose-dependent, divergent effects by drug class. Mov Disord. 2012. doi:10.1002/mds.25082. 40. Chaudhuri KR, Logishetty K. Dopamine receptor agonists and sleep disturbances in Parkinson's disease. Parkinsonism Relat Disord. 2009;15 Suppl 4:S101–4. doi:10.1016/S1353-8020(09) 70845-4. 41. Hobson DE, Lang AE, Martin WR, Razmy A, Rivest J, Fleming J. Excessive daytime sleepiness and sudden-onset sleep in Parkinson disease: a survey by the Canadian Movement Disorders Group. JAMA. 2002;287(4):455–63. 42. Schenck CH, Bundlie SR, Ettinger MG, Mahowald MW. Chronic behavioral disorders of human REM sleep: a new category of parasomnia. Sleep. 1986;9(2):293–308. 43. Arnulf I. REM sleep behavior disorder: motor manifestations and pathophysiology. Mov Disord. 2012;27(6):677–89. doi:10.1002/ mds.24957. 44. Ohayon MM, Caulet M, Priest RG. Violent behavior during sleep. J Clin Psychiatry. 1997;58(8):369–76. quiz 77. 45. Schenck CH, Bundlie SR, Mahowald MW. Delayed emergence of a parkinsonian disorder in 38 % of 29 older men initially diagnosed with idiopathic rapid eye movement sleep behaviour disorder. Neurology. 1996;46(2):388–93. 46. Schenck CH, Boeve BF, Mahowald MW. Delayed emergence of a parkinsonian disorder or dementia in 81 % of older men initially diagnosed with idiopathic rapid eye movement sleep behavior

Curr Neurol Neurosci Rep (2015) 15:8 disorder: a 16-year update on a previously reported series. Sleep Med. 2013;14(8):744–8. doi:10.1016/j.sleep.2012.10.009. 47. Iranzo A, Tolosa E, Gelpi E, Molinuevo JL, Valldeoriola F, Serradell M, et al. Neurodegenerative disease status and postmortem pathology in idiopathic rapid-eye-movement sleep behaviour disorder: an observational cohort study. Lancet Neurol. 2013;12(5):443–53. doi:10.1016/S1474-4422(13)70056-5. 48. Postuma RB, Gagnon JF, Vendette M, Fantini ML, MassicotteMarquez J, Montplaisir J. Quantifying the risk of neurodegenerative disease in idiopathic REM sleep behavior disorder. Neurology. 2009;72(15):1296–300. doi:10.1212/01.wnl. 0000340980.19702.6e. 49. Iranzo A, Fernandez-Arcos A, Tolosa E, Serradell M, Molinuevo JL, Valldeoriola F, et al. Neurodegenerative disorder risk in idiopathic REM sleep behavior disorder: study in 174 patients. PLoS One. 2014;9(2):e89741. doi:10.1371/journal.pone.0089741. 50.•• Boeve BF, Silber MH, Ferman TJ, Lin SC, Benarroch EE, Schmeichel AM, et al. Clinicopathologic correlations in 172 cases of rapid eye movement sleep behavior disorder with or without a coexisting neurologic disorder. Sleep Med. 2013;14(8):754–62. doi:10.1016/j.sleep.2012.10.015. Although a significant link between RBD and synucleinopathies has been reported in several longitudinal studies, this clinicopathological study confirmed a high percentage of pathologically proven synucleinopathies (94 %) among RBD patients associated with neurodegenerative diseases (n=170). 51.•• Murray ME, Ferman TJ, Boeve BF, Przybelski SA, Lesnick TG, Liesinger AM, et al. MRI and pathology of REM sleep behavior disorder in dementia with Lewy bodies. Neurology. 2013;81(19): 1681–9. doi:10.1212/01.wnl.0000435299.57153.f0. This study used pathologically confirmed DLB patients and found that the presence of RBD was associated with a higher likelihood of DLB pathology and less severe AD-related pathology. 52. Dauvilliers Y, Postuma RB, Ferini-Strambi L, Arnulf I, Högl B, Manni R, et al. Family history of idiopathic REM behavior disorder: a multicenter case–control study. Neurology. 2013;80(24): 2233–5. doi:10.1212/WNL.0b013e318296e967. 53. Frauscher B, Jennum P, Ju YE, Postuma RB, Arnulf I, Cochen De Cock V, et al. Comorbidity and medication in REM sleep behavior disorder: a multicenter case–control study. Neurology. 2014;82(12):1076–9. doi:10.1212/WNL.0000000000000247. 54. Postuma RB, Montplaisir JY, Pelletier A, Dauvilliers Y, Oertel W, Iranzo A, et al. Environmental risk factors for REM sleep behavior disorder: a multicenter case–control study. Neurology. 2012;79(5): 428–34. doi:10.1212/WNL.0b013e31825dd383. 55. Boeve BF. REM sleep behavior disorder: updated review of the core features, the REM sleep behavior disorder-neurodegenerative disease association, evolving concepts, controversies, and future directions. Ann N Y Acad Sci. 2010;1184:15–54. doi:10.1111/j. 1749-6632.2009.05115.x. 56. American Academy of Sleep Medicine. International classification of sleep disorders. 3rd ed. Darien: American Academy of Sleep Medicine; 2014. 57. American Academy of Sleep Medicine. The AASM Manual for the Scoring of Sleep and Associated Events: rules, terminology and technical specifications: version 2.0.3. Darien, IL, USA; 2014. 58. Miyamoto T, Miyamoto M, Iwanami M, Kobayashi M, Nakamura M, Inoue Y, et al. The REM sleep behavior disorder screening questionnaire: validation study of a Japanese version. Sleep Med. 2009;10(10):1151–4. doi:10.1016/j.sleep.2009.05.007. 59. Stiasny-Kolster K, Mayer G, Schafer S, Moller JC, HeinzelGutenbrunner M, Oertel WH. The REM sleep behavior disorder screening questionnaire—a new diagnostic instrument. Mov Disord. 2007;22(16):2386–93. doi:10.1002/mds.21740. 60. Boeve BF, Molano JR, Ferman TJ, Smith GE, Lin SC, Bieniek K, et al. Validation of the Mayo Sleep Questionnaire to screen for

Page 9 of 11 8

61.

62.

63.

64.

65.

66.

67.

68.

69.

70.

71.

72.

73.

74.

75.

76.

REM sleep behavior disorder in an aging and dementia cohort. Sleep Med. 2011;12(5):445–53. doi:10.1016/j.sleep.2010.12.009. Li SX, Wing YK, Lam SP, Zhang J, Yu MW, Ho CK, et al. Validation of a new REM sleep behavior disorder questionnaire (RBDQ-HK). Sleep Med. 2010;11(1):43–8. doi:10.1016/j.sleep. 2009.06.008. Frauscher B, Ehrmann L, Zamarian L, Auer F, Mitterling T, Gabelia D, et al. Validation of the Innsbruck REM sleep behavior disorder inventory. Mov Disord. 2012;27(13):1673–8. doi:10. 1002/mds.25223. Iranzo A, Santamaria J. Severe obstructive sleep apnea/hypopnea mimicking REM sleep behavior disorder. Sleep. 2005;28(2):203– 6. Postuma RB, Gagnon JF, Vendette M, Montplaisir JY. Markers of neurodegeneration in idiopathic rapid eye movement sleep behaviour disorder and Parkinson's disease. Brain. 2009;132(Pt 12): 3298–307. doi:10.1093/brain/awp244. Gagnon JF, Bertrand JA, Genier Marchand D. Cognition in rapid eye movement sleep behavior disorder. Front Neurol. 2012;3:82. doi:10.3389/fneur.2012.00082. Postuma RB, Lanfranchi PA, Blais H, Gagnon JF, Montplaisir JY. Cardiac autonomic dysfunction in idiopathic REM sleep behavior disorder. Mov Disord. 2010;25(14):2304–10. doi:10.1002/mds. 23347. Miyamoto T, Miyamoto M, Inoue Y, Usui Y, Suzuki K, Hirata K. Reduced cardiac 123I-MIBG scintigraphy in idiopathic REM sleep behavior disorder. Neurology. 2006;67(12):2236–8. doi:10. 1212/01.wnl.0000249313.25627.2e. Iwanami M, Miyamoto T, Miyamoto M, Hirata K, Takada E. Relevance of substantia nigra hyperechogenicity and reduced odor identification in idiopathic REM sleep behavior disorder. Sleep Med. 2010;11(4):361–5. doi:10.1016/j.sleep.2009.12.006. Postuma RB, Gagnon JF, Rompre S, Montplaisir JY. Severity of REM atonia loss in idiopathic REM sleep behavior disorder predicts Parkinson disease. Neurology. 2010;74(3):239–44. doi:10. 1212/WNL.0b013e3181ca0166. Miyamoto T, Orimo S, Miyamoto M, Hirata K, Adachi T, Hattori R, et al. Follow-up PET studies in case of idiopathic REM sleep behavior disorder. Sleep Med. 2010;11(1):100–1. doi:10.1016/j. sleep.2009.05.006. Iranzo A, Lomena F, Stockner H, Valldeoriola F, Vilaseca I, Salamero M, et al. Decreased striatal dopamine transporter uptake and substantia nigra hyperechogenicity as risk markers of synucleinopathy in patients with idiopathic rapid-eye-movement sleep behaviour disorder: a prospective study [corrected]. Lancet Neurol. 2010;9(11):1070–7. doi:10.1016/S1474-4422(10)702167. Postuma RB, Lang AE, Gagnon JF, Pelletier A, Montplaisir JY. How does parkinsonism start? Prodromal parkinsonism motor changes in idiopathic REM sleep behaviour disorder. Brain. 2012;135(Pt 6):1860–70. doi:10.1093/brain/aws093. McDade EM, Boot BP, Christianson TJ, Pankratz VS, Boeve BF, Ferman TJ, et al. Subtle gait changes in patients with REM sleep behavior disorder. Mov Disord. 2013;28(13):1847–53. doi:10. 1002/mds.25653. Trenkwalder C, Arnulf I. Principles and practice of sleep medicine. In: Kryger MH, Roth T, Dement WC, editors. Parkinsonism. 5th ed. St. Louis: Saunders; 2010. p. 980–92. Bugalho P, Viana-Baptista M. REM sleep behavior disorder and motor dysfunction in Parkinson's disease—a longitudinal study. Parkinsonism Relat Disord. 2013;19(12):1084–7. doi:10.1016/j. parkreldis.2013.07.017. Bolitho SJ, Naismith SL, Terpening Z, Grunstein RR, Melehan K, Yee BJ, et al. Investigating rapid eye movement sleep without atonia in Parkinson's disease using the rapid eye movement sleep

8

Curr Neurol Neurosci Rep (2015) 15:8

Page 10 of 11

behavior disorder screening questionnaire. Mov Disord. 2014;29(6):736–42. doi:10.1002/mds.25832. 77. Kim YE, Jeon BS. Clinical implication of REM sleep behavior disorder in Parkinson's disease. J Parkinson's Dis. 2014;4(2):237– 44. doi:10.3233/JPD-130293. 78. Gong Y, Xiong KP, Mao CJ, Shen Y, Hu WD, Huang JY, et al. Clinical manifestations of Parkinson disease and the onset of rapid eye movement sleep behavior disorder. Sleep Med. 2014;15(6): 647–53. doi:10.1016/j.sleep.2013.12.021. 79. Nihei Y, Takahashi K, Koto A, Mihara B, Morita Y, Isozumi K, et al. REM sleep behavior disorder in Japanese patients with Parkinson's disease: a multicenter study using the REM sleep behavior disorder screening questionnaire. J Neurol. 2012;259(8): 1606–12. doi:10.1007/s00415-011-6386-1. 80. Postuma RB, Gagnon JF, Vendette M, Charland K, Montplaisir J. Manifestations of Parkinson disease differ in association with REM sleep behavior disorder. Mov Disord. 2008;23(12):1665– 72. doi:10.1002/mds.22099. 81. Postuma RB, Gagnon JF, Vendette M, Charland K, Montplaisir J. REM sleep behaviour disorder in Parkinson's disease is associated with specific motor features. J Neurol Neurosurg Psychiatry. 2008;79(10):1117–21. doi:10.1136/jnnp.2008.149195. 82. Sixel-Döring F, Trautmann E, Mollenhauer B, Trenkwalder C. Associated factors for REM sleep behavior disorder in Parkinson disease. Neurology. 2011;77(11):1048–54. doi:10.1212/WNL. 0b013e31822e560e. 83. Suzuki K, Miyamoto T, Miyamoto M, Watanabe Y, Suzuki S, Tatsumoto M, et al. Probable rapid eye movement sleep behavior disorder, nocturnal disturbances and quality of life in patients with Parkinson's disease: a case-controlled study using the rapid eye movement sleep behavior disorder screening questionnaire. BMC Neurol. 2013;13:18. doi:10.1186/1471-2377-13-18. 84. Gagnon JF, Vendette M, Postuma RB, Desjardins C, MassicotteMarquez J, Panisset M, et al. Mild cognitive impairment in rapid eye movement sleep behavior disorder and Parkinson's disease. Ann Neurol. 2009;66(1):39–47. doi:10.1002/ana.21680. 85. Erro R, Santangelo G, Picillo M, Vitale C, Amboni M, Longo K, et al. Link between non-motor symptoms and cognitive dysfunctions in de novo, drug-naive PD patients. J Neurol. 2012;259(9): 1808–13. doi:10.1007/s00415-011-6407-0. 86. Plomhause L, Dujardin K, Duhamel A, Delliaux M, Derambure P, Defebvre L, et al. Rapid eye movement sleep behavior disorder in treatment-naive Parkinson disease patients. Sleep Med. 2013;14(10):1035–7. doi:10.1016/j.sleep.2013.04.018. 87. Postuma RB, Bertrand JA, Montplaisir J, Desjardins C, Vendette M, Rios Romenets S, et al. Rapid eye movement sleep behavior disorder and risk of dementia in Parkinson's disease: a prospective study. Mov Disord. 2012;27(6):720–6. doi:10.1002/mds.24939. 88. Miyamoto T, Miyamoto M, Iwanami M, Hirata K. Cardiac 123IMIBG accumulation in Parkinson's disease differs in association with REM sleep behavior disorder. Parkinsonism Relat Disord. 2011;17(3):219–20. doi:10.1016/j.parkreldis.2010.11.020. 89. Nomura T, Inoue Y, Högl B, Uemura Y, Kitayama M, Abe T, et al. Relationship between (123)I-MIBG scintigrams and REM sleep behavior disorder in Parkinson's disease. Parkinsonism Relat Disord. 2010;16(10):683–5. doi:10.1016/j.parkreldis.2010.08. 011. 90. Mahale R, Yadav R, Pal PK. Rapid eye movement sleep behaviour disorder in young- and older-onset Parkinson disease: a questionnaire-based study. Sleep Med. 2014;15(6):642–6. doi: 10.1016/j.sleep.2014.01.022. 91. Neikrug AB, Avanzino JA, Liu L, Maglione JE, Natarajan L, Corey-Bloom J, et al. Parkinson's disease and REM sleep behavior disorder result in increased non-motor symptoms. Sleep Med. 2014. doi:10.1016/j.sleep.2014.04.009.

92.

Garcia-Lorenzo D, Longo-Dos Santos C, Ewenczyk C, LeuSemenescu S, Gallea C, Quattrocchi G, et al. The coeruleus/ subcoeruleus complex in rapid eye movement sleep behaviour disorders in Parkinson's disease. Brain. 2013;136(Pt 7):2120–9. doi:10.1093/brain/awt152. 93. Allen RP, Picchietti D, Hening WA, Trenkwalder C, Walters AS, Montplaisir J. 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 Med. 2003;4(2):101–19. 94. Schmidauer C, Sojer M, Seppi K, Stockner H, Högl B, Biedermann B, et al. Transcranial ultrasound shows nigral hypoechogenicity in restless legs syndrome. Ann Neurol. 2005;58(4):630–4. doi:10.1002/ana.20572. 95. Wong JC, Li Y, Schwarzschild MA, Ascherio A, Gao X. Restless legs syndrome: an early clinical feature of Parkinson disease in men. Sleep. 2014;37(2):369–72. doi:10.5665/sleep.3416. 96. Nomura T, Inoue Y, Miyake M, Yasui K, Nakashima K. Prevalence and clinical characteristics of restless legs syndrome in Japanese patients with Parkinson's disease. Mov Disord. 2006;21(3):380–4. doi:10.1002/mds.20734. 97. Ondo WG, Vuong KD, Jankovic J. Exploring the relationship between Parkinson disease and restless legs syndrome. Arch Neurol. 2002;59(3):421–4. 98. Peralta CM, Frauscher B, Seppi K, Wolf E, Wenning GK, Högl B, et al. Restless legs syndrome in Parkinson's disease. Mov Disord. 2009;24(14):2076–80. doi:10.1002/mds.22694. 99. Iranzo A, Comella CL, Santamaria J, Oertel W. Restless legs syndrome in Parkinson's disease and other neurodegenerative diseases of the central nervous system. Mov Disord. 2007;22 Suppl 18:S424–30. doi:10.1002/mds.21600. 100. Ondo WG. Sleep/wake problems in Parkinson's disease: pathophysiology and clinicopathologic correlations. J Neural Transm. 2014. doi:10.1007/s00702-014-1239-6. 101. Möller JC, Unger M, Stiasny-Kolster K, Oertel WH. Restless legs syndrome (RLS) and Parkinson's disease (PD)-related disorders or different entities? J Neurol Sci. 2010;289(1–2):135–7. doi:10. 1016/j.jns.2009.08.035. 102. Braga-Neto P, da Silva-Junior FP, Sueli Monte F, de Bruin PF, de Bruin VM. Snoring and excessive daytime sleepiness in Parkinson's disease. J Neurol Sci. 2004;217(1):41–5. 103. Lee JE, Shin HW, Kim KS, Sohn YH. Factors contributing to the development of restless legs syndrome in patients with Parkinson disease. Mov Disord. 2009;24(4):579–82. doi:10.1002/mds.22410. 104. Krishnan PR, Bhatia M, Behari M. Restless legs syndrome in Parkinson's disease: a case-controlled study. Mov Disord. 2003;18(2):181–5. doi:10.1002/mds.10307. 105. Angelini M, Negrotti A, Marchesi E, Bonavina G, Calzetti S. A study of the prevalence of restless legs syndrome in previously untreated Parkinson's disease patients: absence of co-morbid association. J Neurol Sci. 2011;310(1–2):286–8. doi:10.1016/j.jns. 2011.08.012. 106. Gjerstad MD, Tysnes OB, Larsen JP. Increased risk of leg motor restlessness but not RLS in early Parkinson disease. Neurology. 2011;77(22):1941–6. doi:10.1212/WNL.0b013e31823a0cc8. 107. Shin HY, Youn J, Yoon WT, Kim JS, Cho JW. Restless legs syndrome in Korean patients with drug-naive Parkinson's disease: a nation-wide study. Parkinsonism Relat Disord. 2013;19(3):355–8. doi:10.1016/j.parkreldis.2012.09.009. 108. Young T, Palta M, Dempsey J, Skatrud J, Weber S, Badr S. The occurrence of sleep-disordered breathing among middle-aged adults. N Engl J Med. 1993;328(17):1230–5. doi:10.1056/ NEJM199304293281704. 109. Suzuki K, Miyamoto M, Miyamoto T, Suzuki S, Watanabe Y, Numao A, et al. Snoring is associated with an impaired motor function, disease severity and the quality of life but not with

Curr Neurol Neurosci Rep (2015) 15:8

110.

111.

112.

113.

114.

excessive daytime sleepiness in patients with Parkinson's disease. Intern Med. 2013;52(8):863–9. da Silva Jr FP, Prado GF, Barbosa ER, Tufik S, Togeiro SM. Sleep disordered breathing in Parkinson's disease: a critical appraisal. Sleep Med Rev. 2014;18(2):173–8. doi:10.1016/j.smrv.2013.04.005. De Cock VC, Abouda M, Leu S, Oudiette D, Roze E, Vidailhet M, et al. Is obstructive sleep apnea a problem in Parkinson's disease? Sleep Med. 2010;11(3):247–52. doi:10.1016/j.sleep.2009.05.008. Trotti LM, Bliwise DL. No increased risk of obstructive sleep apnea in Parkinson's disease. Mov Disord. 2010;25(13):2246–9. doi:10.1002/mds.23231. Diederich NJ, Vaillant M, Leischen M, Mancuso G, Golinval S, Nati R, et al. Sleep apnea syndrome in Parkinson's disease. A case–control study in 49 patients. Mov Disord. 2005;20(11): 1413–8. doi:10.1002/mds.20624. Maria B, Sophia S, Michalis M, Charalampos L, Andreas P, John ME, et al. Sleep breathing disorders in patients with idiopathic Parkinson's disease. Respir Med. 2003;97(10):1151–7.

Page 11 of 11 8 115.

Nomura T, Inoue Y, Kobayashi M, Namba K, Nakashima K. Characteristics of obstructive sleep apnea in patients with Parkinson's disease. J Neurol Sci. 2013;327(1–2):22–4. doi:10. 1016/j.jns.2013.01.036. 116. Valko PO, Hauser S, Werth E, Waldvogel D, Baumann CR. Heart rate variability in patients with idiopathic Parkinson's disease with and without obstructive sleep apnea syndrome. Parkinsonism Relat Disord. 2012;18(5):525–31. doi:10.1016/j.parkreldis.2012. 01.023. 117. Yong MH, Fook-Chong S, Pavanni R, Lim LL, Tan EK. Case control polysomnographic studies of sleep disorders in Parkinson's disease. PLoS One. 2011;6(7):e22511. doi:10.1371/ journal.pone.0022511. 118. Neikrug AB, Liu L, Avanzino JA, Maglione JE, Natarajan L, Bradley L, et al. Continuous positive airway pressure improves sleep and daytime sleepiness in patients with Parkinson disease and sleep apnea. Sleep. 2014;37(1):177–85. doi:10.5665/sleep. 3332.

wake disturbances.

Sleep disturbances are a common non-motor feature in patients with Parkinson's disease (PD). Early diagnosis and appropriate management are imperative...
933KB Sizes 1 Downloads 14 Views