1
|
Pounders JD, McCarter SJ. Sleep and Prodromal Synucleinopathies. Semin Neurol 2025. [PMID: 40097019 DOI: 10.1055/a-2544-1482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
α-synucleinopathies are a complex group of progressive neurodegenerative disorders with an increasingly recognized long prodromal period, during which sleep dysfunction is a hallmark. Sleep disorders during the prodromal synucleinopathy period, primarily isolated rapid eye movement (REM) sleep behavior disorder (iRBD) and daytime hypersomnolence correlate best with the recently proposed "body-first" Lewy body disease progression. iRBD is the most widely recognized form of prodromal α-synucleinopathy, and patients with iRBD show abnormal α-synuclein in tissues and biofluids even in the absence of cognitive or motor symptoms. More importantly, individuals with iRBD have an elevated risk for near-term development of a clinically diagnosable symptomatic synucleinopathy. Other sleep disorders such as hypersomnia and circadian rhythm dysfunction also occur across the synucleinopathy spectrum, although their prognostic significance is less well understood than iRBD. Finally, isolated REM sleep without atonia may represent an even earlier stage of prodromal synucleinopathy, but further studies are needed.
Collapse
Affiliation(s)
- Johnson D Pounders
- Mayo Clinic Department of Neurology, Division of Behavioral Neurology Rochester, Minnesota
| | - Stuart J McCarter
- Mayo Clinic Department of Neurology, Division of Behavioral Neurology Rochester, Minnesota
- Mayo Clinic Center for Sleep Medicine, Division of Pulmonology, Rochester, Minnesota
| |
Collapse
|
2
|
Salsone M, Agosta F, Filippi M, Ferini-Strambi L. Sleep disorders and Parkinson's disease: is there a right direction? J Neurol 2024; 271:6439-6451. [PMID: 39133321 DOI: 10.1007/s00415-024-12609-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/23/2024] [Accepted: 07/27/2024] [Indexed: 08/13/2024]
Abstract
In the last years, the hypothesis of a close relationship between sleep disorders (SDs) and Parkinson's disease (PD) has significantly strengthened. Whether this association is causal has been also highlighted by recent evidence demonstrating a neurobiological link between SDs and PD. Thus, the question is not whether these two chronic conditions are mutually connected, but rather how and when this relationship is expressed. Supporting this, not all SDs manifest with the same temporal sequence in PD patients. Indeed, SDs can precede or occur concomitantly with the onset of the clinical manifestation of PD. This review discusses the existing literature, putting under a magnifying glass the timing of occurrence of SDs in PD-neurodegeneration. Based on this, here, we propose two possible directions for studying the SDs-PD relationship: the first direction, from SDs to PD, considers SDs as potential biomarker/precursor of future PD-neurodegeneration; the second direction, from PD to SDs, considers SDs as concomitant symptoms in manifest PD, mainly related to primary PD-neuropathology and/or parkinsonian drugs. Furthermore, for each direction, we questioned SDs-PD relationship in terms of risk factors, neuronal circuits/mechanisms, and impact on the clinical phenotype and disease progression. Future research is needed to investigate whether targeting sleep may be the winning strategy to treat PD, in the context of a personalized precision medicine.
Collapse
Affiliation(s)
- Maria Salsone
- Vita-Salute San Raffaele University, Milan, Italy.
- IRCCS Istituto Policlinico San Donato, Milan, Italy.
| | - Federica Agosta
- Vita-Salute San Raffaele University, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Filippi
- Vita-Salute San Raffaele University, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luigi Ferini-Strambi
- Vita-Salute San Raffaele University, Milan, Italy
- Division of Neuroscience, Sleep Disorders Center, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
3
|
Semenova EI, Rudenok MM, Rybolovlev IN, Shulskaya MV, Lukashevich MV, Partevian SA, Budko AI, Nesterov MS, Abaimov DA, Slominsky PA, Shadrina MI, Alieva AK. Effects of Age and MPTP-Induced Parkinson's Disease on the Expression of Genes Associated with the Regulation of the Sleep-Wake Cycle in Mice. Int J Mol Sci 2024; 25:7721. [PMID: 39062963 PMCID: PMC11276692 DOI: 10.3390/ijms25147721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Parkinson's disease (PD) is characterized by a long prodromal period, during which patients often have sleep disturbances. The histaminergic system and circadian rhythms play an important role in the regulation of the sleep-wake cycle. Changes in the functioning of these systems may be involved in the pathogenesis of early stages of PD and may be age-dependent. Here, we have analyzed changes in the expression of genes associated with the regulation of the sleep-wake cycle (Hnmt, Hrh1, Hrh3, Per1, Per2, and Chrm3) in the substantia nigra (SN) and striatum of normal male mice of different ages, as well as in young and adult male mice with an MPTP-induced model of the early symptomatic stage (ESS) of PD. Age-dependent expression analysis in normal mouse brain tissue revealed changes in Hrh3, Per1, Per2, and Chrm3 genes in adult mice relative to young mice. When gene expression was examined in mice with the MPTP-induced model of the ESS of PD, changes in the expression of all studied genes were found only in the SN of adult mice with the ESS model of PD. These data suggest that age is a significant factor influencing changes in the expression of genes associated with sleep-wake cycle regulation in the development of PD.
Collapse
Affiliation(s)
- Ekaterina I. Semenova
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Margarita M. Rudenok
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Ivan N. Rybolovlev
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Marina V. Shulskaya
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Maria V. Lukashevich
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Suzanna A. Partevian
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Alexander I. Budko
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Maxim S. Nesterov
- Scientific Center for Biomedical Technologies of the Federal Biomedical Agency of Russia, 119435 Krasnogorsk, Russia;
| | - Denis A. Abaimov
- Research Center of Neurology, Volokolamskoye Shosse 80, 125367 Moscow, Russia;
| | - Petr A. Slominsky
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Maria I. Shadrina
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| | - Anelya Kh. Alieva
- National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia; (M.M.R.); (I.N.R.); (M.V.S.); (M.V.L.); (S.A.P.); (A.I.B.); (P.A.S.); (M.I.S.); (A.K.A.)
| |
Collapse
|
4
|
Krugliakova E, Karpovich A, Stieglitz L, Huwiler S, Lustenberger C, Imbach L, Bujan B, Jedrysiak P, Jacomet M, Baumann CR, Fattinger S. Exploring the local field potential signal from the subthalamic nucleus for phase-targeted auditory stimulation in Parkinson's disease. Brain Stimul 2024; 17:769-779. [PMID: 38906529 DOI: 10.1016/j.brs.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 05/26/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Enhancing slow waves, the electrophysiological (EEG) manifestation of non-rapid eye movement (NREM) sleep, could potentially benefit patients with Parkinson's disease (PD) by improving sleep quality and slowing disease progression. Phase-targeted auditory stimulation (PTAS) is an approach to enhance slow waves, which are detected in real-time in the surface EEG signal. OBJECTIVE We aimed to test whether the local-field potential of the subthalamic nucleus (STN-LFP) can be used to detect frontal slow waves and assess the electrophysiological changes related to PTAS. METHODS We recruited patients diagnosed with PD and undergoing Percept™ PC neurostimulator (Medtronic) implantation for deep brain stimulation of STN (STN-DBS) in a two-step surgery. Patients underwent three full-night recordings, including one between-surgeries recording and two during rehabilitation, one with DBS+ (on) and one with DBS- (off). Surface EEG and STN-LFP signals from Percept PC were recorded simultaneously, and PTAS was applied during sleep in all three recording sessions. RESULTS Our results show that during NREM sleep, slow waves of the cortex and STN are time-locked. PTAS application resulted in power and coherence changes, which can be detected in STN-LFP. CONCLUSION Our findings suggest the feasibility of implementing PTAS using solely STN-LFP signal for slow wave detection, thus without a need for an external EEG device alongside the implanted neurostimulator. Moreover, we propose options for more efficient STN-LFP signal preprocessing, including different referencing and filtering to enhance the reliability of cortical slow wave detection in STN-LFP recordings.
Collapse
Affiliation(s)
- Elena Krugliakova
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Artyom Karpovich
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lennart Stieglitz
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Stephanie Huwiler
- Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Caroline Lustenberger
- Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Lukas Imbach
- Swiss Epilepsy Center, Clinic Lengg, Zurich, Switzerland
| | - Bartosz Bujan
- Neurorehabilitation, Clinic Lengg, Zurich, Switzerland
| | | | - Maria Jacomet
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Christian R Baumann
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sara Fattinger
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
5
|
Bonato G, Cimino P, Pistonesi F, Salviati L, Bertolin C, Carecchio M. Non-Motor Symptoms in Primary Familial Brain Calcification. J Clin Med 2024; 13:3873. [PMID: 38999439 PMCID: PMC11242504 DOI: 10.3390/jcm13133873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/17/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
Background/Objectives: Primary Familial Brain Calcification is a rare neurodegenerative disorder of adulthood characterized by calcium deposition in the basal ganglia and other brain areas; the main clinical manifestations include movement disorders, mainly parkinsonism. Non-motor symptoms are not well defined in PFBC. This work aims at defining the burden of non-motor symptoms in PFBC. Methods: A clinical, genetic and neuropsychological evaluation of a cohort of PFBC patients, COMPASS-31 scale administration. Results: A total of 50 PFBC patients were recruited; in 25, the genetic test was negative; 10 carried mutations in SLC20A2 gene, 8 in MYORG, 3 in PDGFB, 1 in PDGFRB, 2 in JAM2 (single mutations), and one test is still ongoing. The main motor manifestation was parkinsonism. Headache was reported in 26% of subjects (especially in PDGFB mutation carriers), anxiety or depression in 62%, psychosis or hallucinations in 10-12%, sleep disturbances in 34%; 14% of patients reported hyposmia, 32% constipation, and 34% urinary disturbances. A neuropsychological assessment revealed cognitive involvement in 56% (sparing memory functions, to some extent). The COMPASS-31 mean score was 20.6, with higher sub-scores in orthostatic intolerance and gastrointestinal problems. MYORG patients and subjects with cognitive decline tended to have higher scores and bladder involvement compared to other groups. Conclusions: The presence of non-motor symptoms is frequent in PFBC and should be systematically assessed to better meet patients' needs.
Collapse
Affiliation(s)
- Giulia Bonato
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, 35128 Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, 35128 Padova, Italy
| | - Paola Cimino
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, 35128 Padova, Italy
| | - Francesca Pistonesi
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, 35128 Padova, Italy
| | - Leonardo Salviati
- Medical Genetics Unit, Department of Women and Children's Health, University of Padova, 35128 Padova, Italy
| | - Cinzia Bertolin
- Medical Genetics Unit, Department of Women and Children's Health, University of Padova, 35128 Padova, Italy
| | - Miryam Carecchio
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, 35128 Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, 35128 Padova, Italy
| |
Collapse
|
6
|
Hauser RA, Videnovic A, Soares-da-Silva P, Liang GS, Olson K, Jen E, Rocha JF, Klepitskaya O. OFF-times before, during, and after nighttime sleep periods in Parkinson's disease patients with motor fluctuations and the effects of opicapone: A post hoc analysis of diary data from BIPARK-1 and -2. Parkinsonism Relat Disord 2024; 123:106971. [PMID: 38631081 DOI: 10.1016/j.parkreldis.2024.106971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/05/2024] [Accepted: 04/07/2024] [Indexed: 04/19/2024]
Abstract
INTRODUCTION In BIPARK-1 and BIPARK-2, addition of once-daily opicapone to levodopa/carbidopa significantly reduced daily "OFF"-time relative to placebo in adults with Parkinson's disease (PD) and motor fluctuations. Diary data from these studies were pooled and analyzed post hoc to characterize "OFF"-times around nighttime sleep and to explore the effects of opicapone 50 mg. METHODS "OFF" before sleep (OBS), "OFF during the nighttime sleep period" (ODNSP), early morning "OFF" (EMO), and duration of nighttime sleep and awake periods were analyzed descriptively at baseline. Mean changes from baseline to Week 14/15 (end of double-blind treatment) were analyzed using two-sided t-tests in participants with data for both visits. RESULTS At baseline, 88.3 % (454/514) of participants reported having OBS (34.0 %), ODNSP (17.1 %), or EMO (79.6 %). Those with ODNSP had substantially shorter mean duration of uninterrupted sleep (4.4 h) than the overall pooled population (7.1 h). At Week 14/15, mean decrease from baseline in ODNSP duration was significantly greater with opicapone than with placebo (-0.9 vs. -0.4 h, P < 0.05). In participants with ODNSP at baseline, the decrease in total time spent awake during the night-time sleep period was significantly greater with opicapone than with placebo (-1.0 vs. -0.4 h, P < 0.05), as was the reduction in percent time spent awake during the night-time sleep period (-12.8 % vs. -4.5 %, P < 0.05). CONCLUSION "OFF"-times around nighttime sleep were common in BIPARK-1 and BIPARK-2. Opicapone may improve sleep by decreasing the amount of time spent awake during the night in patients with PD who have night-time sleep period "OFF" episodes.
Collapse
Affiliation(s)
- Robert A Hauser
- University of South Florida, Parkinson's Disease & Movement Disorders Center, 4001 E. Fletcher Ave, 6th Floor, Tampa, FL, 33613, USA.
| | - Aleksandar Videnovic
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, 165 Cambridge Street, Suite 650, Boston, MA, 02214, USA
| | - Patrício Soares-da-Silva
- BIAL-Portela & C(a), S.A., À Avenida da Siderurgia Nacional, 4745-457, Coronado, Portugal; University of Porto, Praça Gomes Teixeira, 4099-002, Porto, Portugal
| | - Grace S Liang
- Neurocrine Biosciences, Inc., 12780 El Camino Real, San Diego, CA, 92130, USA
| | - Kurt Olson
- Neurocrine Biosciences, Inc., 12780 El Camino Real, San Diego, CA, 92130, USA
| | - Eric Jen
- Neurocrine Biosciences, Inc., 12780 El Camino Real, San Diego, CA, 92130, USA
| | - José-Francisco Rocha
- BIAL-Portela & C(a), S.A., À Avenida da Siderurgia Nacional, 4745-457, Coronado, Portugal
| | - Olga Klepitskaya
- Neurocrine Biosciences, Inc., 12780 El Camino Real, San Diego, CA, 92130, USA
| |
Collapse
|
7
|
Dodet P, Houot M, Leu-Semenescu S, Corvol JC, Lehéricy S, Mangone G, Vidailhet M, Roze E, Arnulf I. Sleep disorders in Parkinson's disease, an early and multiple problem. NPJ Parkinsons Dis 2024; 10:46. [PMID: 38424131 PMCID: PMC10904863 DOI: 10.1038/s41531-024-00642-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 01/15/2024] [Indexed: 03/02/2024] Open
Abstract
In Parkinson's disease (PD), it remains unclear whether sleep disorders including insomnia, REM sleep behavior disorder (RBD), excessive daytime sleepiness (EDS), restless legs syndrome (RLS) and sleep-disordered breathing (SDB), are isolated or combined, interact with each other and are associated with clinical factors. We sought to determine the prevalence and combinations of the main sleep disorders, and their clinical and polysomnographic associations in early stage PD. Sleep disorders were systematically diagnosed after medical interview and video-polysomnography in 162 participants with early stage PD and 58 healthy controls from the baseline of the longitudinal ICEBERG cohort. Demographic, clinical (motor, cognitive, autonomic, psychological and sensory tests), therapeutic and polysomnographic associations of sleep disorders were investigated. Sleep disorders were frequent (71%) and combined in half of the patients. The number of sleep disorders increased with disease duration and dysautonomia. Insomnia was the most common (41%), followed by definite RBD (25%), EDS (25%), and RLS (16%). These disorders were more frequent than in controls whereas SDB was rare, moderate and similar in both groups. In patients, insomnia (mainly difficulties maintaining sleep) was associated with female gender, shorter sleep time and RLS, but not with motor or psychological symptoms. RBD was associated with dysautonomia and advanced age, but not with motor and cognitive measures. EDS was associated with psychiatric and motor symptoms as well as the sedative effects of dopamine agonists but not with other sleep disturbances. Sleep disturbances are frequent and combined in early patients with PD. Their determinants and markers are more organic than psychological.
Collapse
Affiliation(s)
- Pauline Dodet
- Service des Pathologies du Sommeil et Centre de Référence National des Narcolepsies et Hypersomnies rares, Assistance Publique-Hôpitaux de Paris-Sorbonne (AP-HP-Sorbonne), Hôpital la Pitié-Salpêtrière, Paris, France.
- Paris Brain Institute (ICM), Sorbonne University, Inserm U1227, CNRS 7225, Paris, France.
| | - Marion Houot
- Center of Excellence of Neurodegenerative Disease (CoEN), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- Assistance Publique Hôpitaux de Paris, Inserm, Clinical Investigation Centre (CIC) Neuroscience, Paris Brain Institute - ICM, Pitié-Salpêtrière Hospital, Paris, France
| | - Smaranda Leu-Semenescu
- Service des Pathologies du Sommeil et Centre de Référence National des Narcolepsies et Hypersomnies rares, Assistance Publique-Hôpitaux de Paris-Sorbonne (AP-HP-Sorbonne), Hôpital la Pitié-Salpêtrière, Paris, France
- Paris Brain Institute (ICM), Sorbonne University, Inserm U1227, CNRS 7225, Paris, France
| | - Jean-Christophe Corvol
- Paris Brain Institute (ICM), Sorbonne University, Inserm U1227, CNRS 7225, Paris, France
- Center of Excellence of Neurodegenerative Disease (CoEN), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- Assistance Publique Hôpitaux de Paris, Inserm, Clinical Investigation Centre (CIC) Neuroscience, Paris Brain Institute - ICM, Pitié-Salpêtrière Hospital, Paris, France
| | - Stéphane Lehéricy
- Paris Brain Institute (ICM), Sorbonne University, Inserm U1227, CNRS 7225, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié -Salpêtrière, Department of Neuroradiology, 75013, Paris, France
| | - Graziella Mangone
- Paris Brain Institute (ICM), Sorbonne University, Inserm U1227, CNRS 7225, Paris, France
| | - Marie Vidailhet
- Paris Brain Institute (ICM), Sorbonne University, Inserm U1227, CNRS 7225, Paris, France
- Center of Excellence of Neurodegenerative Disease (CoEN), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Emmanuel Roze
- Paris Brain Institute (ICM), Sorbonne University, Inserm U1227, CNRS 7225, Paris, France
- Center of Excellence of Neurodegenerative Disease (CoEN), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Isabelle Arnulf
- Service des Pathologies du Sommeil et Centre de Référence National des Narcolepsies et Hypersomnies rares, Assistance Publique-Hôpitaux de Paris-Sorbonne (AP-HP-Sorbonne), Hôpital la Pitié-Salpêtrière, Paris, France
- Paris Brain Institute (ICM), Sorbonne University, Inserm U1227, CNRS 7225, Paris, France
| |
Collapse
|
8
|
Peng C, Wang Z, Sun Y, Mo Y, Hu K, Li Q, Hou X, Zhu Z, He X, Xue S, Zhang S. Subthalamic nucleus dynamics track microlesion effect in Parkinson's disease. Front Cell Dev Biol 2024; 12:1370287. [PMID: 38434618 PMCID: PMC10906266 DOI: 10.3389/fcell.2024.1370287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 02/06/2024] [Indexed: 03/05/2024] Open
Abstract
Parkinson's Disease (PD) is characterized by the temporary alleviation of motor symptoms following electrode implantation (or nucleus destruction), known as the microlesion effect (MLE). Electrophysiological studies have explored different PD stages, but understanding electrophysiological characteristics during the MLE period remains unclear. The objective was to examine the characteristics of local field potential (LFP) signals in the subthalamic nucleus (STN) during the hyperacute period following implantation (within 2 days) and 1 month post-implantation. 15 patients diagnosed with PD were enrolled in this observational study, with seven simultaneous recordings of bilateral STN-LFP signals using wireless sensing technology from an implantable pulse generator. Recordings were made in both on and off medication states over 1 month after implantation. We used a method to parameterize the neuronal power spectrum to separate periodic oscillatory and aperiodic components effectively. Our results showed that beta power exhibited a significant increase in the off medication state 1 month after implantation, compared to the postoperative hyperacute period. Notably, this elevation was effectively attenuated by levodopa administration. Furthermore, both the exponents and offsets displayed a decrease at 1 month postoperatively when compared to the hyperacute postoperative period. Remarkably, levodopa medication exerted a modulatory effect on these aperiodic parameters, restoring them back to levels observed during the hyperacute period. Our findings suggest that both periodic and aperiodic components partially capture distinct electrophysiological characteristics during the MLE. It is crucial to adequately evaluate such discrepancies when exploring the mechanisms of MLE and optimizing adaptive stimulus protocols.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Sha Xue
- Neurosurgery Center, Department of Functional Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shizhong Zhang
- Neurosurgery Center, Department of Functional Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Lima MMS, Targa ADS, Dos Santos Lima GZ, Cavarsan CF, Torterolo P. Macro and micro-sleep dysfunctions as translational biomarkers for Parkinson's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 174:187-209. [PMID: 38341229 DOI: 10.1016/bs.irn.2023.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Sleep disturbances are highly prevalent among patients with Parkinson's disease (PD) and often appear from the early-phase disease or prodromal stages. In this chapter, we will discuss the current evidence addressing the links between sleep dysfunctions in PD, focusing most closely on those data from animal and mathematical/computational models, as well as in human-based studies that explore the electrophysiological and molecular mechanisms by which PD and sleep may be intertwined, whether as predictors or consequences of the disease. It is possible to clearly state that leucine-rich repeat kinase 2 gene (LRRK2) is significantly related to alterations in sleep architecture, particularly affecting rapid eye movement (REM) sleep and non-REM sleep, thus impacting sleep quality. Also, decreases in gamma power, observed after dopaminergic lesions, correlates negatively with the degree of injury, which brings other levels of understanding the impacts of the disease. Besides, abnormal synchronized oscillations among basal ganglia nuclei can be detrimental for information processing considering both motor and sleep-related processes. Altogether, despite clear advances in the field, it is still difficult to definitely establish a comprehensive understanding of causality among all the sleep dysfunctions with the disease itself. Although, certainly, the search for biomarkers is helping in shortening this road towards a better and faster diagnosis, as well as looking for more efficient treatments.
Collapse
Affiliation(s)
- Marcelo M S Lima
- Neurophysiology Laboratory, Department of Physiology, Federal University of Paraná, Curitiba, Paraná, Brazil.
| | - Adriano D S Targa
- CIBER of Respiratory diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain; Translational Research in Respiratory Medicine, Hospital Universitari Arnau de Vilanova-Santa Maria, Biomedical Research Institute of Lleida (IRBLleida), Lleida, Spain
| | - Gustavo Z Dos Santos Lima
- Science and Technology School, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Clarissa F Cavarsan
- College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Pablo Torterolo
- Laboratory of Sleep Neurobiology, Department of Physiology, School of Medicine, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
10
|
Dubessy AL, Arnulf I. Sleepiness in neurological disorders. Rev Neurol (Paris) 2023; 179:755-766. [PMID: 37598089 DOI: 10.1016/j.neurol.2023.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 08/21/2023]
Abstract
Sleepiness is a frequent and underrecognized symptom in neurological disorders, that impacts functional outcomes and quality of life. Multiple and potentially additive factors might contribute to sleepiness in neurological disorders, including sleep quality alterations, circadian rhythm disorders, drugs, and sleep disorders including sleep apnea or central disorders of hypersomnolence. Physician awareness of the possible symptoms of hypersomnolence, and associated causes is of crucial importance to allow proper identification and treatment of underlying causes. This review first provides a brief overview on clinical aspects of excessive daytime sleepiness, and diagnosis tools, then examines its frequency and mechanisms in various neurological disorders, including neurodegenerative disorders, multiple sclerosis, autoimmune encephalitis, epilepsy, and stroke.
Collapse
Affiliation(s)
- A-L Dubessy
- Saint Antoine Hospital, Assistance publique des Hôpitaux de Paris (AP-HP), Paris, France.
| | - I Arnulf
- Sleep Disorder Unit, Pitié-Salpêtrière Hospital and Sorbonne University, Paris, France; National Reference Network for Orphan Diseases: Narcolepsy and Rare Hypersomnias, Paris, France
| |
Collapse
|
11
|
Thangaleela S, Sivamaruthi BS, Kesika P, Mariappan S, Rashmi S, Choeisoongnern T, Sittiprapaporn P, Chaiyasut C. Neurological Insights into Sleep Disorders in Parkinson's Disease. Brain Sci 2023; 13:1202. [PMID: 37626558 PMCID: PMC10452387 DOI: 10.3390/brainsci13081202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/07/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Parkinson's disease (PD) is a common multidimensional neurological disorder characterized by motor and non-motor features and is more prevalent in the elderly. Sleep disorders and cognitive disturbances are also significant characteristics of PD. Sleep is an important physiological process for normal human cognition and physical functioning. Sleep deprivation negatively impacts human physical, mental, and behavioral functions. Sleep disturbances include problems falling asleep, disturbances occurring during sleep, abnormal movements during sleep, insufficient sleep, and excessive sleep. The most recognizable and known sleep disorders, such as rapid-eye-movement behavior disorder (RBD), insomnia, excessive daytime sleepiness (EDS), restless legs syndrome (RLS), sleep-related breathing disorders (SRBDs), and circadian-rhythm-related sleep-wake disorders (CRSWDs), have been associated with PD. RBD and associated emotional disorders are common non-motor symptoms of PD. In individuals, sleep disorders and cognitive impairment are important prognostic factors for predicting progressing neurodegeneration and developing dementia conditions in PD. Studies have focused on RBD and its associated neurological changes and functional deficits in PD patients. Other risks, such as cognitive decline, anxiety, and depression, are related to RBD. Sleep-disorder diagnosis is challenging, especially in identifying the essential factors that disturb the sleep-wake cycle and the co-existence of other concomitant sleep issues, motor symptoms, and breathing disorders. Focusing on sleep patterns and their disturbances, including genetic and other neurochemical changes, helps us to better understand the central causes of sleep alterations and cognitive functions in PD patients. Relations between α-synuclein aggregation in the brain and gender differences in sleep disorders have been reported. The existing correlation between sleep disorders and levels of α-synuclein in the cerebrospinal fluid indicates the risk of progression of synucleinopathies. Multidirectional approaches are required to correlate sleep disorders and neuropsychiatric symptoms and diagnose sensitive biomarkers for neurodegeneration. The evaluation of sleep pattern disturbances and cognitive impairment may aid in the development of novel and effective treatments for PD.
Collapse
Affiliation(s)
- Subramanian Thangaleela
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (S.T.); (B.S.S.); (P.K.)
| | - Bhagavathi Sundaram Sivamaruthi
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (S.T.); (B.S.S.); (P.K.)
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Periyanaina Kesika
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (S.T.); (B.S.S.); (P.K.)
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Subramanian Rashmi
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (S.T.); (B.S.S.); (P.K.)
| | - Thiwanya Choeisoongnern
- Neuropsychological Research Laboratory, Neuroscience Research Center, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand
| | - Phakkharawat Sittiprapaporn
- Neuropsychological Research Laboratory, Neuroscience Research Center, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Bangkok 10110, Thailand
| | - Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (S.T.); (B.S.S.); (P.K.)
| |
Collapse
|
12
|
Ma R, Yin Z, Chen Y, Yuan T, An Q, Gan Y, Xu Y, Jiang Y, Du T, Yang A, Meng F, Zhu G, Zhang J. Sleep outcomes and related factors in Parkinson's disease after subthalamic deep brain electrode implantation: a retrospective cohort study. Ther Adv Neurol Disord 2023; 16:17562864231161163. [PMID: 37200769 PMCID: PMC10185976 DOI: 10.1177/17562864231161163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/15/2023] [Indexed: 05/20/2023] Open
Abstract
Background Subthalamic nucleus deep brain stimulation (STN-DBS) improves sleep qualities in Parkinson's disease (PD) patients; however, it remains elusive whether STN-DBS improves sleep by directly influencing the sleep circuit or alleviates other cardinal symptoms such as motor functions, other confounding factors including stimulation intensity may also involve. Studying the effect of microlesion effect (MLE) on sleep after STN-DBS electrode implantation may address this issue. Objective To examine the influence of MLE on sleep quality and related factors in PD, as well as the effects of regional and lateral specific correlations with sleep outcomes after STN-DBS electrode implantation. Study Design Case-control study; Level of evidence, 3. Data Sources and Methods In 78 PD patients who underwent bilateral STN-DBS surgery in our center, we compared the sleep qualities, motor performances, anti-Parkinsonian drug dosage, and emotional conditions at preoperative baseline and postoperative 1-month follow-up. We determined the related factors of sleep outcomes and visualized the electrodes position, simulated the MLE-engendered volume of tissue lesioned (VTL), and investigated sleep-related sweet/sour spots and laterality in STN. Results MLE improves sleep quality with Pittsburgh Sleep Quality Index (PSQI) by 13.36% and Parkinson's Disease Sleep Scale-2 (PDSS-2) by 17.95%. Motor (P = 0.014) and emotional (P = 0.001) improvements were both positively correlated with sleep improvements. However, MLE in STN associative subregions, as an independent factor, may cause sleep deterioration (r = 0.348, P = 0.002), and only the left STN showed significance (r = 0.327, P = 0.004). Sweet spot analysis also indicated part of the left STN associative subregion is the sour spot indicative of sleep deterioration. Conclusion The MLE of STN-DBS can overall improve sleep quality in PD patients, with a positive correlation between motor and emotional improvements. However, independent of all other factors, the MLE in the STN associative subregion, particularly the left side, may cause sleep deterioration.
Collapse
Affiliation(s)
- Ruoyu Ma
- Department of Neurosurgery, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
| | - Zixiao Yin
- Department of Neurosurgery, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
| | - Yingchuan Chen
- Department of Neurosurgery, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
| | - Tianshuo Yuan
- Department of Neurosurgery, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
| | - Qi An
- Department of Neurosurgery, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
| | - Yifei Gan
- Department of Neurosurgery, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
| | - Yichen Xu
- Department of Neurosurgery, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
| | - Yin Jiang
- Department of Functional Neurosurgery, Beijing
Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Tingting Du
- Department of Functional Neurosurgery, Beijing
Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Anchao Yang
- Department of Neurosurgery, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
| | - Fangang Meng
- Department of Functional Neurosurgery, Beijing
Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Neurostimulation,
Beijing, China
| | - Guanyu Zhu
- Department of Neurosurgery, Beijing Tiantan
Hospital, Capital Medical University, No. 119 South 4th Ring West Road,
Fengtai District, Beijing 100070, China
| | - Jianguo Zhang
- Department of Neurosurgery, Beijing Tiantan
Hospital, Capital Medical University, No. 119 South 4th Ring West Road,
Fengtai District, Beijing 100070, China
- Department of Functional Neurosurgery, Beijing
Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Neurostimulation,
Beijing, China
| |
Collapse
|
13
|
Silvani A, Baldelli L, Giannini G, Guaraldi P, Sambati L, Cecere A, Mignani F, Cortelli P, Calandra-Buonaura G, Provini F. Pervasive and diffuse muscle activity during REM sleep and non-REM sleep characterises multiple system atrophy in comparison with Parkinson's disease. J Sleep Res 2023; 32:e13721. [PMID: 36054178 DOI: 10.1111/jsr.13721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/05/2022] [Accepted: 08/07/2022] [Indexed: 12/01/2022]
Abstract
Multiple system atrophy (MSA) and Parkinson's disease (PD) may share overlapping features particularly at early disease stage, including sleep alterations, but have profoundly different prognoses. Certain sleep phenomena and disorders of motor control are more prevalent in multiple system atrophy, such as REM sleep behaviour disorder (RBD). We quantitatively tested whether pervasive muscle activity during sleep occurs in subjects with multiple system atrophy versus Parkinson's disease. Laboratory polysomnographic studies were performed in 50 consecutive subjects with Parkinson's disease and 26 age- and gender-matched subjects with multiple system atrophy at <5 years from disease onset. The distributions of normalised electromyographic activity of submentalis, wrist extensor, and tibialis anterior muscles in different wake-sleep states during the night were analysed. Subjects with multiple system atrophy had significantly higher activity of submentalis, wrist extensor, and tibialis anterior muscles than subjects with Parkinson's disease during non-REM sleep, including separately in stages N1, N2, and N3, and during REM sleep, but not during nocturnal wakefulness. The activity of wrist extensor and tibialis anterior muscles during non-REM sleep and the activity of tibialis anterior muscles during REM sleep were also significantly higher in subjects with multiple system atrophy and RBD than in subjects with Parkinson's disease and RBD. In conclusion, with respect to Parkinson's disease, multiple system atrophy is characterised by a pervasive and diffuse muscle overactivity that involves axial and limb muscles and occurs not only during REM sleep, but also during non-REM sleep and between subjects with comorbid RBD.
Collapse
Affiliation(s)
- Alessandro Silvani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Luca Baldelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giulia Giannini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Pietro Guaraldi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Luisa Sambati
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Annagrazia Cecere
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Francesco Mignani
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Pietro Cortelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Giovanna Calandra-Buonaura
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Federica Provini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
14
|
McCarter SJ, Camerucci E, Mullan AF, Stang CD, Turcano P, St. Louis EK, Boeve BF, Savica R. Sleep Disorders in Early-Onset Parkinsonism: A Population-Based Study. JOURNAL OF PARKINSON'S DISEASE 2023; 13:1175-1183. [PMID: 37742659 PMCID: PMC10657686 DOI: 10.3233/jpd-230045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/25/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND Sleep disturbances are common in parkinsonian disorders; however, whether sleep disorders affect individuals with early-onset parkinsonism and whether they differ from individuals with typical-onset parkinsonism is unknown. OBJECTIVE To compare the prevalence and incidence of sleep disorders before and after parkinsonian motor symptom onset between individuals with early onset parkinsonism (age ≤50 at motor symptom onset) and typical-onset parkinsonism (age >50 at motor symptom onset). METHODS We used a population-based, 1991 to 2015 incident-cohort study of parkinsonism including 38 patients with early-onset and 1,001 patients with typical-onset parkinsonism. Presence or absence and type of sleep disorder as well as the relationship between motor and sleep symptoms were abstracted from the medical records. Rates of sleep disorders before and after onset of parkinsonism were compared with logistic regression and Cox proportional hazards models. RESULTS The prevalence of sleep disorders prior to the onset of parkinsonism in early vs. typical parkinsonism (24% vs. 16% p = 0.19) and incidence of sleep disorders after parkinsonism onset (5.85 cases per 100 person-years vs. 4.11 cases per 100 person-years; HR 1.15 95% CI: 0.74-1.77) were similar between the two groups. Early-onset parkinsonism had a higher risk for developing post-motor insomnia compared with typical-onset parkinsonism (HR 1.73, 95% CI: 1.02-2.93); the risk for developing all other sleep disorders considered was similar between groups. CONCLUSION Sleep disorders are common in individuals with early-onset parkinsonism and occur with similar frequency to those with typical-onset parkinsonism, except for insomnia, which was more frequent in the early-onset group.
Collapse
Affiliation(s)
- Stuart J. McCarter
- Mayo Clinic Department of Neurology, Rochester, MN, USA
- Mayo Clinic Center for Sleep Medicine, Rochester, MN, USA
| | | | - Aidan F. Mullan
- Mayo Clinic Department of Quantitative Health Sciences, Rochester, MN, USA
| | - Cole D. Stang
- Mayo Clinic Department of Neurology, Rochester, MN, USA
| | | | - Erik K. St. Louis
- Mayo Clinic Department of Neurology, Rochester, MN, USA
- Mayo Clinic Center for Sleep Medicine, Rochester, MN, USA
| | | | - Rodolfo Savica
- Mayo Clinic Department of Neurology, Rochester, MN, USA
- Mayo Clinic Department of Quantitative Health Sciences, Rochester, MN, USA
| |
Collapse
|
15
|
Komagamine T, Suzuki K, Kokubun N, Komagamine J, Kawasaki A, Funakoshi K, Hirata K. Sleep-related hallucinations in patients with Parkinson's disease. PLoS One 2022; 17:e0276736. [PMID: 36282859 PMCID: PMC9595548 DOI: 10.1371/journal.pone.0276736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/12/2022] [Indexed: 01/24/2023] Open
Abstract
Given that sleep-wake cycle dysfunction can cause hallucinations in Parkinson's disease patients, sleep-related hallucinations may be a different subtype from hallucinations that occur only during full wakefulness. However, few studies that distinguish the onset situations of hallucinations related to sleep from those that occur in full wakefulness have been conducted to investigate hallucinations in Parkinson's disease patients. Therefore, we conducted a multicenter observational study to investigate the prevalence of and factors associated with sleep-related hallucinations in patients with Parkinson's disease. Information on hallucinations was collected by using a questionnaire and face-to-face interviews. Of 100 consecutive patients with Parkinson's disease, 29 (29%) reported sleep-related hallucinations, and 16 (16%) reported hallucinations only in the full wakefulness. A longer duration of Parkinson's disease treatment (OR 1.35, 95% CI 1.07 to 1.72), higher Beck Depression Inventory-II scores (OR 1.07; 95% CI 1.01 to 1.14), and higher rapid eye movement sleep behavior disorder scores (OR 5.60; 95% CI 1.54 to 20.38) were independent factors associated with the presence of sleep-related hallucinations in a multivariable analysis. Sleep-related hallucinations, but not daytime hallucinations, were associated with probable rapid eye movement sleep behavior disorder. Phenomenological discrimination between sleep-related hallucinations and daytime hallucinations is important for elucidating the full pathology in Parkinson's disease and the mechanisms underlying hallucinations.
Collapse
Affiliation(s)
- Tomoko Komagamine
- Department of Neurology, Dokkyo Medical University, Tochigi, Japan
- Department of Internal Medicine, National Hospital Organization Tochigi Medical Center, Tochigi, Japan
| | - Keisuke Suzuki
- Department of Neurology, Dokkyo Medical University, Tochigi, Japan
| | - Norito Kokubun
- Department of Neurology, Dokkyo Medical University, Tochigi, Japan
| | - Junpei Komagamine
- Department of Internal Medicine, National Hospital Organization Tochigi Medical Center, Tochigi, Japan
| | - Akiko Kawasaki
- Department of Neurology, Dokkyo Medical University, Tochigi, Japan
| | - Kei Funakoshi
- Department of Neurology, Dokkyo Medical University, Tochigi, Japan
| | - Koichi Hirata
- Department of Neurology, Dokkyo Medical University, Tochigi, Japan
| |
Collapse
|
16
|
Mizrahi-Kliger AD, Feldmann LK, Kühn AA, Bergman H. Etiologies of insomnia in Parkinson's disease - Lessons from human studies and animal models. Exp Neurol 2022; 350:113976. [PMID: 35026228 DOI: 10.1016/j.expneurol.2022.113976] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/27/2021] [Accepted: 01/06/2022] [Indexed: 12/28/2022]
Abstract
Sleep disorders are integral to Parkinson's disease (PD). Insomnia, an inability to maintain stable sleep, affects most patients and is widely rated as one of the most debilitating facets of this disease. PD insomnia is often perceived as a multifactorial entity - a consequence of several of the disease symptoms, comorbidities and therapeutic strategies. Yet, this view evolved against a backdrop of a relative scarcity of works trying to directly dissect the underlying neural correlates and mechanisms in animal models. The last years have seen the emergence of a wealth of new evidence regarding the neural underpinnings of insomnia in PD. Here, we review early and recent reports from patients and animal models evaluating the etiology of PD insomnia. We start by outlining the phenomenology of PD insomnia and continue to analyze the evidence supporting insomnia as emanating from four distinct subdivisions of etiologies - the symptoms and comorbidities of the disease, the medical therapy, the degeneration of non-dopaminergic cell groups and subsequent alterations in circadian rhythms, and the degeneration of dopaminergic neurons in the brainstem and its resulting effect on the basal ganglia. Finally, we review emerging neuromodulation-based therapeutic avenues for PD insomnia.
Collapse
Affiliation(s)
- Aviv D Mizrahi-Kliger
- Department of Neurobiology, Institute of Medical Research Israel-Canada, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 91120, Israel.
| | - Lucia K Feldmann
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, Berlin 10117, Germany
| | - Andrea A Kühn
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, Berlin 10117, Germany; NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin 10117, Germany; Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin, Germany
| | - Hagai Bergman
- Department of Neurobiology, Institute of Medical Research Israel-Canada, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 91120, Israel; The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem 91904, Israel; Department of Neurosurgery, Hadassah University Hospital, Jerusalem 91120, Israel
| |
Collapse
|
17
|
Kataoka H, Isogawa M, Inoue T, Hasebe M, Takashima R, Kasama S, Nanaura H, Kiriyama T, Kasahara M, Sugie K. Zonisamide for the Efficacy of Sleep Abnormality in Parkinson's Disease (ZEAL Study): A Protocol for Randomized Controlled Trials. Front Neurol 2022; 12:741307. [PMID: 34970203 PMCID: PMC8713296 DOI: 10.3389/fneur.2021.741307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 11/05/2021] [Indexed: 11/15/2022] Open
Abstract
Background: Sleep disorders are one of the most frequent non-motor symptoms of Parkinson's disease (PD), and the efficacy of dopaminergic agents remains controversial. Clinical randomized control trials for the treatment of sleep disorders in PD are limited. Zonisamide (1,2-benzisoxazole-3-methanesulfonamide) improved motor symptoms and wearing-off in patients with PD. Patients with PD were reported to have dream-enacting behavior that was resolved after treatment with zonisamide. This study aimed to verify the safety and efficacy of zonisamide for sleep disorders and rapid eye movement (REM) sleep behavioral disorders using a mobile two-channel electroencephalography (EEG)/electrooculography (EOG) recording system. Methods and Analysis: The present study is a randomized placebo-controlled trial to determine the efficacy of zonisamide for sleep disorders in patients with PD. This study was designed to be single-blind, but the subject allocation is randomized by an independent allocation manager via computer-generated block randomization. The subjects in the treatment group took zonisamide (25 mg per day) before bedtime for 28 days. The sleep index is analyzed using a portable EEG/EOG recording system collected on two consecutive nights within 7 days prior to the intervention and reobtained on one night within 2 days after the 28-day administration of zonisamide. The amount of change in sleep efficiency before and after the 28-day administration will be compared between the zonisamide treatment group and placebo group concerning the primary endpoint. As for the secondary endpoint, the change in the ratio of other sleep parameters, including REM sleep without atonia, or sleep architecture will be evaluated. Ethics and Dissemination: The protocol was approved by the Nara Medical University Certified Review Board (CRB5200002). The trial was notified and registered with the Japan Registry of Clinical Trials (jRCTs051200160). Written informed consent will be obtained from every participant using informed consent approved by the CRB. The results of this trial will be disseminated through peer-reviewed scientific journals.
Collapse
Affiliation(s)
- Hiroshi Kataoka
- Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Masahiro Isogawa
- Institute for Clinical and Translational Science, Nara Medical University Hospital, Kashihara, Japan
| | - Takashi Inoue
- Institute for Clinical and Translational Science, Nara Medical University Hospital, Kashihara, Japan
| | - Miyoko Hasebe
- Institute for Clinical and Translational Science, Nara Medical University Hospital, Kashihara, Japan
| | - Ryuzo Takashima
- Institute for Clinical and Translational Science, Nara Medical University Hospital, Kashihara, Japan
| | - Shu Kasama
- Institute for Clinical and Translational Science, Nara Medical University Hospital, Kashihara, Japan
| | - Hitoki Nanaura
- Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Takao Kiriyama
- Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Masato Kasahara
- Institute for Clinical and Translational Science, Nara Medical University Hospital, Kashihara, Japan
| | - Kazuma Sugie
- Department of Neurology, Nara Medical University, Kashihara, Japan
| |
Collapse
|
18
|
Liu H, Li J, Wang X, Huang J, Wang T, Lin Z, Xiong N. Excessive Daytime Sleepiness in Parkinson's Disease. Nat Sci Sleep 2022; 14:1589-1609. [PMID: 36105924 PMCID: PMC9464627 DOI: 10.2147/nss.s375098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Excessive daytime sleepiness (EDS) is one of the most common sleep disorders in Parkinson's disease (PD). It has attracted much attention due to high morbidity, poor quality of life, increased risk for accidents, obscure mechanisms, comorbidity with PD and limited therapeutic approaches. In this review, we summarize the current literature on epidemiology of EDS in PD to address the discrepancy between subjective and objective measures and clarify the reason for the inconsistent prevalence in previous studies. Besides, we focus on the effects of commonly used antiparkinsonian drugs on EDS and related pharmacological mechanisms to provide evidence for rational clinical medication in sleepy PD patients. More importantly, degeneration of wake-promoting nuclei owing to primary neurodegenerative process of PD is the underlying pathogenesis of EDS. Accordingly, altered wake-promoting nerve nuclei and neurotransmitter systems in PD patients are highlighted to providing clues for identifying EDS-causing targets in the sleep and wake cycles. Future mechanistic studies toward this direction will hopefully advance the development of novel and specific interventions for EDS in PD patients.
Collapse
Affiliation(s)
- Hanshu Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jingwen Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xinyi Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zhicheng Lin
- Laboratory of Psychiatric Neurogenomics, McLean Hospital; Harvard Medical School, Belmont, MA, 02478, USA
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
19
|
Nodel M, Shevtsova K, Kovrov G. The factors impact on the urgent daytime sleepiness degree in the Parkinson’s disease. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:102-108. [DOI: 10.17116/jnevro2022122101102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
20
|
Morawska MM, Moreira CG, Ginde VR, Valko PO, Weiss T, Büchele F, Imbach LL, Masneuf S, Kollarik S, Prymaczok N, Gerez JA, Riek R, Baumann CR, Noain D. Slow-wave sleep affects synucleinopathy and regulates proteostatic processes in mouse models of Parkinson's disease. Sci Transl Med 2021; 13:eabe7099. [PMID: 34878820 DOI: 10.1126/scitranslmed.abe7099] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Marta M Morawska
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland.,University of Zurich (UZH), Neuroscience Center Zurich (ZNZ), Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Carlos G Moreira
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland.,ETH Zurich, Neuroscience Center Zurich (ZNZ), Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Varun R Ginde
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland
| | - Philipp O Valko
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland
| | - Tobias Weiss
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland
| | - Fabian Büchele
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland
| | - Lukas L Imbach
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland
| | - Sophie Masneuf
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland
| | - Sedef Kollarik
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland.,University of Zurich (UZH), Neuroscience Center Zurich (ZNZ), Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Natalia Prymaczok
- ETH Zurich, Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, Vladimir-Prelog-Weg 2, Zurich 8093, Switzerland
| | - Juan A Gerez
- ETH Zurich, Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, Vladimir-Prelog-Weg 2, Zurich 8093, Switzerland
| | - Roland Riek
- ETH Zurich, Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, Vladimir-Prelog-Weg 2, Zurich 8093, Switzerland
| | - Christian R Baumann
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland.,University of Zurich (UZH), Neuroscience Center Zurich (ZNZ), Winterthurerstrasse 190, Zurich 8057, Switzerland.,Center of Competence Sleep and Health Zurich, University of Zurich, Frauenklinikstrasse 26, Zurich 8091, Switzerland
| | - Daniela Noain
- Department of Neurology, University Hospital Zurich (USZ), Frauenklinikstrasse 26, Zurich 8091, Switzerland.,University of Zurich (UZH), Neuroscience Center Zurich (ZNZ), Winterthurerstrasse 190, Zurich 8057, Switzerland.,Center of Competence Sleep and Health Zurich, University of Zurich, Frauenklinikstrasse 26, Zurich 8091, Switzerland
| |
Collapse
|
21
|
Videnovic A, Amara AW, Comella C, Schweitzer PK, Emsellem H, Liu K, Sterkel AL, Gottwald MD, Steinerman JR, Jochelson P, Zomorodi K, Hauser RA. Solriamfetol for Excessive Daytime Sleepiness in Parkinson's Disease: Phase 2 Proof-of-Concept Trial. Mov Disord 2021; 36:2408-2412. [PMID: 34191352 PMCID: PMC8596433 DOI: 10.1002/mds.28702] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 11/29/2022] Open
Abstract
Background Solriamfetol is approved (US and EU) for excessive daytime sleepiness (EDS) in narcolepsy and obstructive sleep apnea. Objectives Evaluate solriamfetol safety/efficacy for EDS in Parkinson's disease (PD). Methods Phase 2, double‐blind, 4‐week, crossover trial: adults with PD and EDS were randomized to sequence A (placebo, solriamfetol 75, 150, 300 mg/d), B (solriamfetol 75, 150, 300 mg/d, placebo), or C (placebo). Outcomes (safety/tolerability [primary]; Epworth Sleepiness Scale [ESS]; Maintenance of Wakefulness Test [MWT]) were assessed weekly. P values are nominal. Results Common adverse events (n = 66): nausea (10.7%), dizziness (7.1%), dry mouth (7.1%), headache (7.1%), anxiety (5.4%), constipation (5.4%), dyspepsia (5.4%). ESS decreased both placebo (−4.78) and solriamfetol (−4.82 to −5.72; P > 0.05). MWT improved dose‐dependently with solriamfetol, increasing by 5.05 minutes with 300 mg relative to placebo (P = 0.0098). Conclusions Safety/tolerability was consistent with solriamfetol's known profile. There were no significant improvements on ESS; MWT results suggest possible benefit with solriamfetol in PD. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society
Collapse
Affiliation(s)
- Aleksandar Videnovic
- Movement Disorders Unit and Division of Sleep Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Amy W Amara
- Division of Movement Disorders, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Cynthia Comella
- Parkinson's Disease and Movement Disorders Program, Rush University, Chicago, Illinois, USA
| | - Paula K Schweitzer
- Sleep Medicine and Research Center, St. Luke's Hospital, Chesterfield, Missouri, USA
| | - Helene Emsellem
- The Center for Sleep & Wake Disorders, Chevy Chase, Maryland, USA
| | - Kris Liu
- Jazz Pharmaceuticals, Palo Alto, California, USA
| | | | | | | | | | | | - Robert A Hauser
- Parkinson's Disease and Movement Disorders Center, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
22
|
Mc Carthy CE. Sleep Disturbance, Sleep Disorders and Co-Morbidities in the Care of the Older Person. Med Sci (Basel) 2021; 9:medsci9020031. [PMID: 34063838 PMCID: PMC8162526 DOI: 10.3390/medsci9020031] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 01/14/2023] Open
Abstract
Sleep complaints can be both common and complex in the older patient. Their consideration is an important aspect of holistic care, and may have an impact on quality of life, mortality, falls and disease risk. Sleep assessment should form part of the comprehensive geriatric assessment. If sleep disturbance is brought to light, consideration of sleep disorders, co-morbidity and medication management should form part of a multifaceted approach. Appreciation of the bi-directional relationship and complex interplay between co-morbidity and sleep in older patients is an important element of patient care. This article provides a brief overview of sleep disturbance and sleep disorders in older patients, in addition to their association with specific co-morbidities including depression, heart failure, respiratory disorders, gastro-oesophageal reflux disease, nocturia, pain, Parkinson's disease, dementia, polypharmacy and falls. A potential systematic multidomain approach to assessment and management is outlined, with an emphasis on non-pharmacological treatment where possible.
Collapse
Affiliation(s)
- Christine E. Mc Carthy
- Department of Geriatric Medicine, University Hospital Galway, Galway, Ireland;
- HRB-Clinical Research Facility, National University of Ireland, Galway, Co., Galway, Ireland
| |
Collapse
|
23
|
Saini D, Mukherjee A, Roy A, Biswas A. A Comparative Study of the Behavioral Profile of the Behavioral Variant of Frontotemporal Dementia and Parkinson's Disease Dementia. Dement Geriatr Cogn Dis Extra 2020; 10:182-194. [PMID: 33569074 PMCID: PMC7841718 DOI: 10.1159/000512042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Executive dysfunction is the common thread between pure cortical dementia like the behavioral variant of frontotemporal dementia (bvFTD) and subcortical dementia like Parkinson's disease dementia (PDD). Although there are clinical and cognitive features to differentiate cortical and subcortical dementia, the behavioral symptoms differentiating these 2 conditions are still not well known. OBJECTIVE To evaluate the behavioral profile of bvFTD and PDD and compare them to find out which behavioral symptoms can differentiate between the two. METHODS Twenty consecutive patients with bvFTD (>1 year after diagnosis) and 20 PDD patients were recruited according to standard diagnostic criteria. Behavioral symptoms were collected from the reliable caregiver by means of a set of questionnaires and then compared between the 2 groups. RESULTS bvFTD patients had more severe disease and more behavioral symptoms than PDD. bvFTD patients were different from PDD patients due to their significantly greater: loss of basic emotion (p < 0.001, odds ratio [OR] 44.33), loss of awareness of pain (p < 0.001, OR 44.33), disinhibition (p < 0.001, OR 35.29), utilization phenomenon (p = 0.008, OR 22.78), loss of taste discrimination (p < 0.001, OR 17), neglect of hygiene (p = 0.001, OR 13.22), loss of embarrassment (p = 0.003, OR 10.52), wandering (p = 0.004, OR 9.33), pacing (p = 0.014, OR 9), selfishness (p = 0.014, OR 9), increased smoking (p = 0.014, OR 9), increased alcohol consumption (p = 0.031, OR 7.36), social avoidance (p = 0.012, OR 6.93), mutism (p = 0.041, OR 5.67), and failure to recognize objects (p = 0.027, OR 4.33). The bvFTD patients were also significantly less suspicious (p = 0.001, OR 0.0295), less inclined to have a false belief that people were in their home (p = 0.014, OR 0.11) and had fewer visual illusions/hallucinations (p = 0.004, OR 0.107) than PDD patients. CONCLUSION Behavioral symptoms are helpful to distinguish bvFTD from PDD, and thus also cortical dementia with frontal-lobe dysfunction from subcortical dementia.
Collapse
Affiliation(s)
| | | | | | - Atanu Biswas
- Department of Neurology, Bangur Institute of Neurosciences, and Institute of Post Graduate Medical Education and Research, Kolkata, India
| |
Collapse
|
24
|
Hermann W, Schmitz-Peiffer H, Kasper E, Fauser M, Franke C, Wienecke M, Otto K, Löhle M, Brandt MD, Reichmann H, Storch A. Sleep Disturbances and Sleep Disordered Breathing Impair Cognitive Performance in Parkinson's Disease. Front Neurosci 2020; 14:689. [PMID: 32903712 PMCID: PMC7438827 DOI: 10.3389/fnins.2020.00689] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 06/05/2020] [Indexed: 01/30/2023] Open
Abstract
Background Sleep disturbances and impairment of cognitive function are among the most frequent non-motor symptoms in Parkinson’s disease (PD) with negative implications on quality of life of patients and caregivers. Despite the fact that sleep disturbances are a major issue in PD patients, only limited data are available regarding interactions of sleep disturbances and cognitive performance. Objective This post hoc analysis of the RaSPar trial was therefore designed to further elucidate sleep disturbances and their impact on cognition in PD. Methods Twenty-six PD patients with sleep disturbances were evaluated thoroughly including assessments of patients’ subjective and objective sleep quality by interview, questionnaires, and polysomnography (PSG). Cognitive performance was assessed by Parkinson Neuropsychometric Dementia Assessment (PANDA) and Test of Attentional Performance (TAP), and associations of sleep and cognitive function were evaluated. Results We did not detect differences in cognitive performance between patients with and without rapid eye movement (REM) sleep behavior disorder (RBD). Instead, cognitive impairment, particularly affecting cognitive domains attention, executive function/working memory, and semantic memory, was associated with impaired PSG-measured sleep quality (e.g., sleep efficiency) and sleep disordered breathing (SDB) (Apnea-Hypopnea Index > 5/h). Global cognitive performance was decreased in patients with SDB (PANDA score 23.2 ± 3.5 vs. 26.9 ± 2.2, P = 0.020, unpaired two-sided t-test). Conclusion Sleep apnea and other sleep disturbances impair cognitive performance in PD and should be evaluated in routine care, and treatment options such as continuous airway pressure therapy should be considered.
Collapse
Affiliation(s)
- Wiebke Hermann
- Department of Neurology, Technische Universität Dresden, Dresden, Germany.,Sleep Centre, Department of Neurology and Department of Internal Medicine, Technische Universität Dresden, Dresden, Germany.,Department of Neurology, University of Rostock, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE), Research Site Rostock, Rostock, Germany
| | | | - Elisabeth Kasper
- Department of Neurology, University of Rostock, Rostock, Germany
| | - Mareike Fauser
- Department of Neurology, University of Rostock, Rostock, Germany
| | - Christiana Franke
- Department of Neurology, Technische Universität Dresden, Dresden, Germany
| | - Miriam Wienecke
- Department of Neurology, Technische Universität Dresden, Dresden, Germany.,Sleep Centre, Department of Neurology and Department of Internal Medicine, Technische Universität Dresden, Dresden, Germany
| | - Karolin Otto
- Department of Neurology, Technische Universität Dresden, Dresden, Germany
| | - Matthias Löhle
- Department of Neurology, University of Rostock, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE), Research Site Rostock, Rostock, Germany
| | - Moritz D Brandt
- Department of Neurology, Technische Universität Dresden, Dresden, Germany.,Sleep Centre, Department of Neurology and Department of Internal Medicine, Technische Universität Dresden, Dresden, Germany.,German Center for Neurodegenerative Diseases (DZNE), Research Site Dresden, Dresden, Germany
| | - Heinz Reichmann
- Department of Neurology, Technische Universität Dresden, Dresden, Germany
| | - Alexander Storch
- Department of Neurology, University of Rostock, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE), Research Site Rostock, Rostock, Germany
| |
Collapse
|
25
|
Basal ganglia beta oscillations during sleep underlie Parkinsonian insomnia. Proc Natl Acad Sci U S A 2020; 117:17359-17368. [PMID: 32636265 DOI: 10.1073/pnas.2001560117] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Sleep disorders are among the most debilitating comorbidities of Parkinson's disease (PD) and affect the majority of patients. Of these, the most common is insomnia, the difficulty to initiate and maintain sleep. The degree of insomnia correlates with PD severity and it responds to treatments that decrease pathological basal ganglia (BG) beta oscillations (10-17 Hz in primates), suggesting that beta activity in the BG may contribute to insomnia. We used multiple electrodes to record BG spiking and field potentials during normal sleep and in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinsonism in nonhuman primates. MPTP intoxication resulted in severe insomnia with delayed sleep onset, sleep fragmentation, and increased wakefulness. Insomnia was accompanied by the onset of nonrapid eye movement (NREM) sleep beta oscillations that were synchronized across the BG and cerebral cortex. The BG beta oscillatory activity was associated with a decrease in slow oscillations (0.1-2 Hz) throughout the cortex, and spontaneous awakenings were preceded by an increase in BG beta activity and cortico-BG beta coherence. Finally, the increase in beta oscillations in the basal ganglia during sleep paralleled decreased NREM sleep, increased wakefulness, and more frequent awakenings. These results identify NREM sleep beta oscillation in the BG as a neural correlate of PD insomnia and suggest a mechanism by which this disorder could emerge.
Collapse
|
26
|
A 3-year observation of excessive daytime sleepiness after subthalamic deep brain stimulation in patients with Parkinson’s disease. Clin Neurol Neurosurg 2020; 192:105721. [DOI: 10.1016/j.clineuro.2020.105721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 01/12/2020] [Accepted: 02/03/2020] [Indexed: 11/21/2022]
|
27
|
Taguchi T, Ikuno M, Yamakado H, Takahashi R. Animal Model for Prodromal Parkinson's Disease. Int J Mol Sci 2020; 21:ijms21061961. [PMID: 32183024 PMCID: PMC7139491 DOI: 10.3390/ijms21061961] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/18/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra and subsequent motor symptoms, but various non-motor symptoms (NMS) often precede motor symptoms. Recently, NMS have attracted much attention as a clue for identifying patients in a prodromal stage of PD, which is an excellent point at which to administer disease-modifying therapies (DMTs). These prodromal symptoms include olfactory loss, constipation, and sleep disorders, especially rapid eye movement sleep behavior disorder (RBD), all of which are also important for elucidating the mechanisms of the initiation and progression of the disease. For the development of DMTs, an animal model that reproduces the prodromal stage of PD is also needed. There have been various mammalian models reported, including toxin-based, genetic, and alpha synuclein propagation models. In this article, we review the animal models that exhibit NMS as prodromal symptoms and also discuss an appropriate prodromal model and its importance for the development of DMT of PD.
Collapse
Affiliation(s)
| | | | - Hodaka Yamakado
- Correspondence: (H.Y.); (R.T.); Tel.: +81-75-751-3767 (H.Y.); Tel.: +81-75-751-4397 (R.T.); Fax: +81-75-761-9780 (H.Y.); Fax: +81-75-761-9780 (R.T.)
| | - Ryosuke Takahashi
- Correspondence: (H.Y.); (R.T.); Tel.: +81-75-751-3767 (H.Y.); Tel.: +81-75-751-4397 (R.T.); Fax: +81-75-761-9780 (H.Y.); Fax: +81-75-761-9780 (R.T.)
| |
Collapse
|
28
|
Pal P, Mahale R, Yadav R. Does quality of sleep differ in familial and sporadic Parkinson’s disease? ANNALS OF MOVEMENT DISORDERS 2020. [DOI: 10.4103/aomd.aomd_7_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
29
|
Effect of ALDH2 on Sleep Disturbances in Patients with Parkinson's Disease. Sci Rep 2019; 9:18950. [PMID: 31831791 PMCID: PMC6908732 DOI: 10.1038/s41598-019-55427-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/07/2019] [Indexed: 12/15/2022] Open
Abstract
Monoamine neurotransmitters play essential roles in the regulation of arousal and sleep. Impaired metabolism of monoamine neurotransmitters could result in the accumulation of neurotoxic aldehyde metabolites and, hence, neuronal degeneration. Aldehyde dehydrogenases play an important role in the metabolism of the neurotoxic aldehyde metabolites, including the aldehyde metabolites of dopamine, serotonin, and noradrenaline. Deficient aldehyde dehydrogenase 2 (ALDH2) has been suggested to result in the accumulation of these biogenic aldehydes. An ALDH2 single nucleotide polymorphism (SNP), rs671 (A), results in significantly reduced ALDH2 enzyme activity. A total of 83 Parkinson’s disease (PD) patients were recruited in this study. In addition to the genotypes of rs671, the patients were assessed with the PD sleep scale-2nd version (PDSS-2) and the Epworth sleepiness scale (ESS) for symptoms of daytime and nocturnal sleep disturbances. The patients carrying rs671 (A) had more frequent dozing while lying down to rest in the afternoon (ESS item5) (F = 7.308, p = 0.008) than the rs671 (GG) patients. The patients with rs671 (A) reported a trend toward more frequent difficulty staying asleep than the patients with rs671 (GG). (F = 3.278, p = 0.074). The results indicate that patients carrying allele rs671 (A) are more likely to experience impairment in the regulation of arousal and sleep. The results also support the hypothesis that the accumulation of neurotoxic monoamine neurotransmitter aldehyde metabolites secondary to reduced ALDH2 enzyme activity may cause more severe monoaminergic neuronal loss and, hence, more severe symptoms in the regulation of wakefulness and sleep.
Collapse
|
30
|
Iyer V, Vo Q, Mell A, Chinniah S, Zenerovitz A, Venkiteswaran K, Kunselman AR, Fang J, Subramanian T. Acute levodopa dosing around-the-clock ameliorates REM sleep without atonia in hemiparkinsonian rats. NPJ PARKINSONS DISEASE 2019; 5:27. [PMID: 31815176 PMCID: PMC6884572 DOI: 10.1038/s41531-019-0096-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 10/21/2019] [Indexed: 01/08/2023]
Abstract
Rapid-eye-movement (REM) sleep without atonia (RSWA), a marker of REM sleep behavior disorder (RBD), is frequently comorbid with Parkinson's disease (PD). Although rodent models are commonly used for studying PD, the neurobiological and behavioral correlates of RBD remain poorly understood. Therefore, we developed a behavior-based criteria to identify RSWA in the hemiparkinsonian rat model of PD. Video recordings of rats were analyzed, to develop a criteria consisting of behavioral signs that occurred during polysomnographically confirmed epochs of sleep-wake stages. The sleep-slouch, a postural shift of the body or head caused only by gravity, was identified as a unique behavioral sign of REM sleep onset and was altered in hemiparkinsonian rats during RSWA. There was a significant correlation between the behavior-based criteria and polysomnograms for all sleep-wake stages in control but not hemiparkinsonian rats indicating a deterioration of sleep-wake architecture in parkinsonism. We then tested the efficacy of levodopa in ameliorating RSWA using intermittent and around-the-clock (ATC) dosing regimens. ATC levodopa dosing at 4 mg/kg for 48 h caused a significant reduction of RSWA as measured by polysomnography and the behavioral-based criteria along with an amelioration of forelimb motor deficits. Our findings show that the phenomenological correlates of RSWA can be reliably characterized in the hemiparkinsonian rat model. ATC levodopa administration ameliorates RSWA in this model without deleterious consequences to the overall sleep-wake architecture and therapeutic benefits for parkinsonian motor deficits. These findings suggest that further study may allow for the application of a similar approach to treat RBD in PD patients.
Collapse
Affiliation(s)
- Vishakh Iyer
- 1Program in Neuroscience, Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN USA
| | - Quynh Vo
- 2Department of Neurology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Anthony Mell
- 3Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA USA
| | - Siven Chinniah
- 3Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA USA
| | - Ashley Zenerovitz
- 3Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA USA
| | - Kala Venkiteswaran
- 3Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA USA
| | - Allen R Kunselman
- 4Department of Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, PA USA
| | - Jidong Fang
- 5Department of Psychiatry, The Pennsylvania State University College of Medicine, Hershey, PA USA
| | - Thyagarajan Subramanian
- 3Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA USA
| |
Collapse
|
31
|
Medeiros DDC, Lopes Aguiar C, Moraes MFD, Fisone G. Sleep Disorders in Rodent Models of Parkinson's Disease. Front Pharmacol 2019; 10:1414. [PMID: 31827439 PMCID: PMC6892229 DOI: 10.3389/fphar.2019.01414] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
Sleep disorders are frequently diagnosed in Parkinson's disease and manifested in the prodromal and advanced stages of the disease. These conditions, which in some cases affect more than 50% of Parkinson's disease (PD) patients, include hypersomnia, often manifested as excessive daytime sleepiness, insomnia, characterized by delayed initiation and fragmentation of sleep at night, and disruption of rapid eye movement (REM) sleep, resulting in loss of atonia and dream enactment. Standard dopamine replacement therapies for the treatment of motor symptoms are generally inadequate to combat sleep abnormalities, which seriously affect the quality of life of PD patients. Rodent models still represent a major tool for the study of many aspects of PD. They have been primarily designed to eliminate midbrain dopamine neurons and elicit motor impairment, which are the traditional pathological features of PD. However, rodent models are increasingly employed to investigate non-motor symptoms, which are often caused by degenerative processes affecting multiple monoaminergic and peptidergic structures. This review describes how neurotoxic and genetic manipulations of rats and mice have been utilized to reproduce some of the major sleep disturbances associated with PD and to what extent these abnormalities can be linked to nondopaminergic dysfunction, affecting for instance noradrenaline, serotonin, and orexin transmission. Strengths and limitations are discussed, as well as the consistency of results obtained so far, and the need for models that better reproduce the multisystemic neurodegenerative nature of PD, thereby allowing to replicate the complex etiology of sleep-related disorders.
Collapse
Affiliation(s)
- Daniel de Castro Medeiros
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cleiton Lopes Aguiar
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Márcio Flávio Dutra Moraes
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
32
|
Excessive daytime sleepiness and fatigue in neurological disorders. Sleep Breath 2019; 24:413-424. [DOI: 10.1007/s11325-019-01921-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 08/06/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022]
|
33
|
Parkinson's Disease is Associated with Dysregulations of a Dopamine-Modulated Gene Network Relevant to Sleep and Affective Neurobehaviors in the Striatum. Sci Rep 2019; 9:4808. [PMID: 30886221 PMCID: PMC6423036 DOI: 10.1038/s41598-019-41248-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 02/07/2019] [Indexed: 12/11/2022] Open
Abstract
In addition to the characteristic motor symptoms, Parkinson’s disease (PD) often involves a constellation of sleep and mood symptoms. However, the mechanisms underlying these comorbidities are largely unknown. We have previously reconstructed gene networks in the striatum of a population of (C57BL/6J x A/J) F2 mice and associated the networks to sleep and affective phenotypes, providing a resource for integrated analyses to investigate perturbed sleep and affective functions at the gene network level. Combining this resource with PD-relevant transcriptomic datasets from humans and mice, we identified four networks that showed elevated gene expression in PD patients, including a circadian clock and mitotic network that was altered similarly in mouse models of PD. We then utilized multiple types of omics data from public databases and linked this gene network to postsynaptic dopamine signaling in the striatum, CDK1-modulated transcriptional regulation, and the genetic susceptibility of PD. These findings suggest that dopamine deficiency, a key aspect of PD pathology, perturbs a circadian/mitotic gene network in striatal neurons. Since the normal functions of this network were relevant to sleep and affective behaviors, these findings implicate that dysregulation of functional gene networks may be involved in the emergence of non-motor symptoms in PD. Our analyses present a framework for integrating multi-omics data from diverse sources in mice and humans to reveal insights into comorbid symptoms of complex diseases.
Collapse
|
34
|
Liguori C, Mercuri NB, Albanese M, Olivola E, Stefani A, Pierantozzi M. Daytime sleepiness may be an independent symptom unrelated to sleep quality in Parkinson's disease. J Neurol 2019; 266:636-641. [PMID: 30607535 DOI: 10.1007/s00415-018-09179-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/28/2018] [Accepted: 12/31/2018] [Indexed: 12/31/2022]
Abstract
Excessive daytime sleepiness (EDS) may represent a disabling non-motor symptom in patients affected by Parkinson's disease (PD). This is a secondary analysis of a previous study documenting the improvement of nocturnal sleep in PD patients treated by rotigotine vs placebo. Here we tested the supposition that EDS may represent a distinct PD non-motor symptom occurring independently of other sleep-wake disorders; moreover, we verified whether EDS can be influenced by the improvement of nocturnal sleep in PD. In the present study, we evaluated the daytime sleepiness of PD patients treated with nocturnal administration of rotigotine (PD-Rot) vs placebo (PD-Pla), as measured by both subjective (Epworth Sleepiness Scale-ESS) and objective (Multiple Sleep Latency Test-MSLT) tools. We included 21 PD-Rot compared to 21 PD-Pla patients and documented no significant changes of both ESS and MSLT data between baseline and follow-up visits in both groups. Moreover, we found no correlations between nocturnal sleep improvement and diurnal sleepiness. Therefore, these data suggest that the improvement of nocturnal sleep in PD patients does not modify the daytime sleepiness, thus suggesting that diurnal sleepiness may occur independently of nocturnal sleep disturbances in PD patients.
Collapse
Affiliation(s)
- Claudio Liguori
- Department of Systems Medicine, Sleep Medicine Centre, University of Rome "Tor Vergata", Viale Oxford 81, 00133, Rome, Italy. .,UOSD Parkinson, University Hospital of Rome "Tor Vergata", Rome, Italy. .,Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.
| | - Nicola Biagio Mercuri
- Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.,Fondazione Santa Lucia IRCCS, Rome, Italy
| | - Maria Albanese
- Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Enrica Olivola
- Parkinson and Movement Disorders Unit, Neurology Department, IRCCS Neuromed, Pozzilli, IS, Italy
| | - Alessandro Stefani
- UOSD Parkinson, University Hospital of Rome "Tor Vergata", Rome, Italy.,Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Mariangela Pierantozzi
- UOSD Parkinson, University Hospital of Rome "Tor Vergata", Rome, Italy.,Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
35
|
Nodel MR, Shevtsova KV, Kovrov GV, Yakhno NN. Nighttime sleep disorders in patients with daytime sleepiness in Parkinson s disease. NEUROLOGY, NEUROPSYCHIATRY, PSYCHOSOMATICS 2018. [DOI: 10.14412/2074-2711-2018-4-23-29] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Daytime sleepiness is one of the clinically significant non-motor manifestations of Parkinson's disease (PD). One of its insufficiently studied aspects is a relationship between daytime sleepiness and nighttime sleep disorders.Objective:to clarify the clinical characteristics of PD in patients with different types of daytime sleepiness and to estimate of the ratio of daytime sleepiness to clinical and polysomnographic characteristics of nighttime sleep in patients with advanced stages of PD.Patients and methods. The investigation included 110 patients (56 men and 54 women) (mean age, 63.78+0.6 years) with PD (Hoehn and Yahr stage 2.6+0.2; disease duration, 6.3+3.2 years) without dementia. All the patients received therapy with levodopa at a mean daily dose of 667.8 mg; 98 of them had the drug in combination with dopamine receptor agonists at a stable dose. The unified PD rating scale, the PD sleep scale (PDSS), and the Epworth sleepiness scale (ESS) were applied. Nocturnal polysomnography (PSG) and the multiple sleep latency test (MSLT) were performed.Results and discussion. There was daytime sleepiness in 44% of the patients: permanent sleepiness in 15%, sudden daytime sleep attacks (along with low daytime sleepiness (ESS) in 14%, and permanent drowsiness concurrent with sleep attacks in 15%. The PSG findings showed a decrease in sleep efficiency, an increase in the duration of the first stage of sleep, a reduction in the duration of the second and third sleep stages, an extension of rapid eye movement (REM) sleep latency, and frequent awakenings (sleep fragmentation). PSG also demonstrated REM sleep behavior disorders (RBD) in half of the examinees.Patients with sleep attacks differed from those with permanent drowsiness without sleep attacks with more severe sleep disorders (PDSS) and shorter sleep latency (MSLT). Patients with the RBD phenomenon had shorter sleep latency (MTLS) than those without this parasomnia. Patients with moderate or severe sleepiness (ESS scores of >10) differed from those with milder drowsiness (ESS scores of 410) and a lower representation of the third sleep stage.Conclusion.There is evidence for the association of daytime sleepiness in PD with reduced efficiency, changes in the nighttime sleep pattern, and RBD.
Collapse
Affiliation(s)
- M. R. Nodel
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia; N.I. Pirogov Russian National Research Medical University, Ministry of Health of Russia
| | - K. V. Shevtsova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia
| | - G. V. Kovrov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia
| | - N. N. Yakhno
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia
| |
Collapse
|
36
|
Christensen E, Abosch A, Thompson JA, Zylberberg J. Inferring sleep stage from local field potentials recorded in the subthalamic nucleus of Parkinson's patients. J Sleep Res 2018; 28:e12806. [DOI: 10.1111/jsr.12806] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/31/2018] [Accepted: 11/07/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Elijah Christensen
- Department of Physiology and Biophysics; University of Colorado School of Medicine; Aurora Colorado
- Medical Scientist Training Program; University of Colorado School of Medicine; Aurora Colorado
| | - Aviva Abosch
- Department of Neurosurgery; University of Colorado School of Medicine; Aurora Colorado
| | - John A. Thompson
- Department of Neurosurgery; University of Colorado School of Medicine; Aurora Colorado
| | - Joel Zylberberg
- Department of Physiology and Biophysics; University of Colorado School of Medicine; Aurora Colorado
- Computational Biosciences Program; University of Colorado School of Medicine; Aurora Colorado
| |
Collapse
|
37
|
Laryngopharyngeal motor dysfunction and obstructive sleep apnea in Parkinson’s disease. Sleep Breath 2018; 23:543-550. [DOI: 10.1007/s11325-018-1729-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 09/18/2018] [Accepted: 09/21/2018] [Indexed: 12/28/2022]
|
38
|
ER Lipid Defects in Neuropeptidergic Neurons Impair Sleep Patterns in Parkinson's Disease. Neuron 2018; 98:1155-1169.e6. [PMID: 29887339 DOI: 10.1016/j.neuron.2018.05.022] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/16/2018] [Accepted: 05/14/2018] [Indexed: 01/23/2023]
Abstract
Parkinson's disease patients report disturbed sleep patterns long before motor dysfunction. Here, in parkin and pink1 models, we identify circadian rhythm and sleep pattern defects and map these to specific neuropeptidergic neurons in fly models and in hypothalamic neurons differentiated from patient induced pluripotent stem cells (iPSCs). Parkin and Pink1 control the clearance of mitochondria by protein ubiquitination. Although we do not observe major defects in mitochondria of mutant neuropeptidergic neurons, we do find an excess of endoplasmic reticulum-mitochondrial contacts. These excessive contact sites cause abnormal lipid trafficking that depletes phosphatidylserine from the endoplasmic reticulum (ER) and disrupts the production of neuropeptide-containing vesicles. Feeding mutant animals phosphatidylserine rescues neuropeptidergic vesicle production and acutely restores normal sleep patterns in mutant animals. Hence, sleep patterns and circadian disturbances in Parkinson's disease models are explained by excessive ER-mitochondrial contacts, and blocking their formation or increasing phosphatidylserine levels rescues the defects in vivo.
Collapse
|
39
|
Mantovani S, Smith SS, Gordon R, O'Sullivan JD. An overview of sleep and circadian dysfunction in Parkinson's disease. J Sleep Res 2018; 27:e12673. [PMID: 29493044 DOI: 10.1111/jsr.12673] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/15/2018] [Accepted: 01/15/2018] [Indexed: 12/18/2022]
Abstract
Sleep and circadian alterations are amongst the very first symptoms experienced in Parkinson's disease, and sleep alterations are present in the majority of patients with overt clinical manifestation of Parkinson's disease. However, the magnitude of sleep and circadian dysfunction in Parkinson's disease, and its influence on the pathophysiology of Parkinson's disease remains often unclear and a matter of debate. In particular, the confounding influences of dopaminergic therapy on sleep and circadian dysfunction are a major challenge, and need to be more carefully addressed in clinical studies. The scope of this narrative review is to summarise the current knowledge around both sleep and circadian alterations in Parkinson's disease. We provide an overview on the frequency of excessive daytime sleepiness, insomnia, restless legs, obstructive apnea and nocturia in Parkinson's disease, as well as addressing sleep structure, rapid eye movement sleep behaviour disorder and circadian features in Parkinson's disease. Sleep and circadian disorders have been linked to pathological conditions that are often co-morbid in Parkinson's disease, including cognitive decline, memory impairment and neurodegeneration. Therefore, targeting sleep and circadian alterations could be one of the earliest and most promising opportunities to slow disease progression. We hope that this review will contribute to advance the discussion and inform new research efforts to progress our knowledge in this field.
Collapse
Affiliation(s)
- Susanna Mantovani
- Faculty of Medicine, The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, Australia.,Wesley Medical Research, Auchenflower, QLD, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Simon S Smith
- Institute for Social Science Research (ISSR), The University of Queensland, Indooroopilly, Australia
| | - Richard Gordon
- Faculty of Medicine, The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, Australia.,Wesley Medical Research, Auchenflower, QLD, Australia
| | - John D O'Sullivan
- Faculty of Medicine, The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, Australia.,Wesley Medical Research, Auchenflower, QLD, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| |
Collapse
|
40
|
Sasai-Sakuma T, Nishio Y, Yokoi K, Mori E, Inoue Y. Pareidolias in REM Sleep Behavior Disorder: A Possible Predictive Marker of Lewy Body Diseases? Sleep 2017; 40:2666487. [PMID: 28364496 DOI: 10.1093/sleep/zsw045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2016] [Indexed: 11/14/2022] Open
Abstract
Study Objectives To investigate conditions and clinical significance of pareidolias in patients with idiopathic rapid eyemovent (REM) sleep behavior disorder (iRBD). Methods This cross-sectional study examined 202 patients with iRBD (66.8 ± 8.0 yr, 58 female) and 46 healthy control subjects (64.7 ± 5.8 years, 14 females). They underwent the Pareidolia test, a newly developed instrument for evoking pareidolias, video polysomnography, olfactory tests, and Addenbrooke's cognitive examination-revised. Results Results show that 53.5% of iRBD patients exhibited one or more pareidolic responses: The rate was higher than control subjects showed (21.7%). The pictures evoking pareidolic responses were more numerous for iRBD patients than for control subjects (1.2 ± 1.8 vs. 0.4 ± 0.8, p < .001). Subgroup analyses revealed that iRBD patients with pareidolic responses had higher amounts of REM sleep without atonia (RWA), with lower sleep efficiency, lower cognitive function, and older age than subjects without pareidolic responses. Results of multivariate analyses show the number of pareidolic responses as a factor associated with decreased cognitive function in iRBD patients with better predictive accuracy. Morbidity length and severity of iRBD, olfactory function, and the amount of RWA were not factors associated with better predictive accuracy. Conclusions Half or more of the iRBD patients showed pareidolic responses. The responses were proven to be associated more intimately with their cognitive decline than clinical or physiological variables related to RBD. Pareidolias in iRBD are useful as a predictive marker of future development of Lewy body diseases.
Collapse
Affiliation(s)
- Taeko Sasai-Sakuma
- Department of Somnology, Tokyo Medical University, Tokyo, Japan.,Division of Biomedical Laboratory Sciences, Department of Life Sciences and Bio-informatics, Graduate School of Health Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshiyuki Nishio
- Department of Behavioral Neurology and Cognitive Neuroscience, Tohoku University School of Medicine, Sendai, Japan
| | - Kayoko Yokoi
- Department of Behavioral Neurology and Cognitive Neuroscience, Tohoku University School of Medicine, Sendai, Japan.,Department of Occupational Therapy, Yamagata Prefectural University of Health Sciences, Yamagata, Japan
| | - Etsuro Mori
- Department of Behavioral Neurology and Cognitive Neuroscience, Tohoku University School of Medicine, Sendai, Japan
| | - Yuichi Inoue
- Department of Somnology, Tokyo Medical University, Tokyo, Japan.,Japan Somnology Center, Neuropsychiatric Research Institute, Tokyo, Japan
| |
Collapse
|
41
|
Baumann-Vogel H, Imbach LL, Sürücü O, Stieglitz L, Waldvogel D, Baumann CR, Werth E. The Impact of Subthalamic Deep Brain Stimulation on Sleep–Wake Behavior: A Prospective Electrophysiological Study in 50 Parkinson Patients. Sleep 2017; 40:3079010. [DOI: 10.1093/sleep/zsx033] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2017] [Indexed: 11/12/2022] Open
|
42
|
Fifel K. Alterations of the circadian system in Parkinson's disease patients. Mov Disord 2016; 32:682-692. [PMID: 27859638 DOI: 10.1002/mds.26865] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/28/2016] [Accepted: 10/05/2016] [Indexed: 01/21/2023] Open
Abstract
Alterations of circadian rhythms are among the most debilitating non-motor symptoms in Parkinson's Disease (PD). Although a growing awareness towards these symptoms has occurred during the last decade, their underlying neuropathophysiology remains poorly understood and consequently no effective therapeutic strategies are available to alleviate these problems. Recent studies have investigated multiple circadian rhythms at different stages of PD. The advances made have allowed an accurate evaluation of the affected underlying pathways and mechanisms. Here I dissect, over disease progression, the relative causal contribution to health impairments in PD patients of dysfunctions in the different components of the neural network governing circadian rhythms. A deeper understanding of these mechanisms will provide not only a greater understanding of disease neuropathology, but also hold the promise for effective therapies. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Karim Fifel
- Laboratory of Neurophysiology, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
43
|
Distinct roles for primate caudate dopamine D1 and D2 receptors in visual discrimination learning revealed using shRNA knockdown. Sci Rep 2016; 6:35809. [PMID: 27805010 PMCID: PMC5090965 DOI: 10.1038/srep35809] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 10/06/2016] [Indexed: 12/13/2022] Open
Abstract
The striatum plays important motor, associative and cognitive roles in brain functions. However, the rodent dorsolateral (the primate putamen) and dorsomedial (the primate caudate nucleus) striatum are not anatomically separated, making it difficult to distinguish their functions. By contrast, anatomical separation exists between the caudate nucleus and putamen in primates. Here, we successfully decreased dopamine D1 receptor (D1R) or D2R mRNA expression levels selectively in the marmoset caudate using shRNA knockdown techniques, as determined using positron emission tomography imaging with specific D1R and D2R ligands and postmortem in situ hybridization analysis. We then conducted a voxel-based correlation analysis between binding potential values of PET imaging and visual discrimination learning task performance in these genetically modified marmosets to find a critical role for the caudate D2R but no apparent role for the caudate D1R. This latter finding challenges the current understanding of the mechanisms underlying D1R activation in the caudate.
Collapse
|
44
|
Selegiline induces a wake promoting effect in rats which is related to formation of its active metabolites. Pharmacol Biochem Behav 2016; 150-151:147-152. [PMID: 27984094 DOI: 10.1016/j.pbb.2016.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/07/2016] [Accepted: 10/10/2016] [Indexed: 11/23/2022]
Abstract
The goal of the present work was to characterise the effects of selegiline on the rat sleep pattern. Furthermore, for comparative purposes, the pharmacokinetics of selegiline and its metabolites in brain and plasma were investigated, and microdialysis experiments were performed to examine the resulting effect on dopamine, noradrenaline and serotonin levels. Selegiline (1, 5, 10 and 30mg/kg) was found to dose-dependently increase the time spent awake following acute dosing. The pharmacokinetic assessment of selegiline showed that, following an oral dose of 5mg/kg, low circulating levels of the parent compound were found relative to those of biotransformed l-methamphetamine and l-amphetamine. The time course of selegiline-induced wakefulness was shown to follow the time course of l-methamphetamine and l-amphetamine in brain, suggesting that these metabolites are responsible for the modulation of sleep architecture. Furthermore, selegiline (5mg/kg) caused a significant increase of extracellular levels of DA (250%) and NA (200%), but not of 5-HT, in the rat prefrontal cortex. In summary, an integrated experimental approach was undertaken here to evaluate selegiline's effect on sleep architecture in rats in relation to its pharmacokinetics and changes in monoaminergic neurotransmitter levels in the brain. The effect of selegiline on sleep was likely mediated by an increase of dopamine and noradrenaline levels in the brain caused by the formed metabolites.
Collapse
|
45
|
Amosova NA, Smolentseva IG, Guseinova PM, Maslyuk OA, Gavrilov EL. [Sleep disorders in the early stage of Parkinson's disease in untreated patients]. Zh Nevrol Psikhiatr Im S S Korsakova 2016; 116:77-81. [PMID: 27723714 DOI: 10.17116/jnevro20161166277-81] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Disorders of sleep and wakefulness occur in about 60-98% of patients with Parkinson's disease (PD). The majority of research on the scale and nature of the disorders was performed in patients treated with antiparkinsonian drugs, and, therefore, the true picture of sleep disorders was interfering with side effects of therapy. The spectrum of these disorders in PD patients is broad and includes insomnia, parasomnia and hypersomnia. The main symptoms of insomnia are difficulty in maintaining sleep, associated with nocturia, rapid eye movement (REM) sleep behavior disorder (RBD), night cramps, akinesia and tremor. The frequency of hypersomnia in PD patients not receiving antiparkinsonian drugs was comparable to that in healthy people. In some studies, hypersomnia is considered as an independent phenomenon of PD, and not associated with the quality of night's sleep or concomitant therapy. Parasomnias in PD patients at the early stage are manifested primarily by REM-sleep behavior disorder (RBD), which has been proven to be a predictor of development of PD. According to our data, insomnia, hypersomnia and RBD were identified in patients who did not receive antiparkinsonian drugs. The polysomnographic study showed the development of RBD in 25% of patients. In the analysis of the anamnesis, it was noted that in 8 cases sleep disorders appeared several years before the first motor symptoms.
Collapse
Affiliation(s)
- N A Amosova
- 'Central state medical Academy' of Department for presidential Affairs of RF, Moscow, Russia
| | - I G Smolentseva
- 'Central state medical Academy' of Department for presidential Affairs of RF, Moscow, Russia
| | - P M Guseinova
- A. I. Burnazyans Federal medical biophysical center, Moscow, Russia
| | - O A Maslyuk
- 'Central state medical Academy' of Department for presidential Affairs of RF, Moscow, Russia
| | - E L Gavrilov
- 'Central state medical Academy' of Department for presidential Affairs of RF, Moscow, Russia
| |
Collapse
|
46
|
Kim JS, Park HE, Oh YS, Lee SH, Park JW, Son BC, Lee KS. Orthostatic hypotension and cardiac sympathetic denervation in Parkinson disease patients with REM sleep behavioral disorder. J Neurol Sci 2016; 362:59-63. [DOI: 10.1016/j.jns.2016.01.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/05/2015] [Accepted: 01/12/2016] [Indexed: 10/25/2022]
|
47
|
Pierantozzi M, Placidi F, Liguori C, Albanese M, Imbriani P, Marciani MG, Mercuri NB, Stanzione P, Stefani A. Rotigotine may improve sleep architecture in Parkinson's disease: a double-blind, randomized, placebo-controlled polysomnographic study. Sleep Med 2016; 21:140-4. [PMID: 27448485 DOI: 10.1016/j.sleep.2016.01.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 01/21/2016] [Indexed: 01/03/2023]
Abstract
BACKGROUND/OBJECTIVES Growing evidence demonstrates that in Parkinson's Disease (PD) sleep disturbances are frequent and difficult to treat. Since the efficacy of rotigotine on sleep is corroborated by studies lacking polysomnography (PSG), this study explores the possible rotigotine-mediated impact on PSG parameters in PD patients. METHODS This is a randomized, double-blind, placebo-controlled, parallel-group study to determine the efficacy of rotigotine vs placebo on PSG parameters in moderately advanced PD patients. An unusual protocol was utilized, since patches were maintained from 18:00 h to awakening, minimizing the possible diurnal impact on motor symptoms. All participants underwent sleep PSG recordings, subjective sleep questionnaires (Parkinson Disease Sleep Scale [PDSS], Pittsburgh Sleep Quality Index [PSQI]), and the assessment of early-morning motor disability. RESULTS We evaluated 42 PD patients (Hoehn & Yahr stages 2 and 3) with sleep impairment randomly assigned to active branch (N =21) or placebo (N = 21). Rotigotine significantly increased sleep efficiency and reduced both wakefulness after sleep onset and sleep latency compared to placebo. Moreover, the mean change in REM sleep quantity was significantly higher in the rotigotine than placebo group. The improvement of PSG parameters corresponded to the amelioration of PDSS and PSQI scores together with the improvement of patient morning motor symptoms. CONCLUSIONS This study demonstrated the significant effect of rotigotine on sleep quality and continuity in PD patients by promoting sleep stability and increasing REM. The effectiveness of rotigotine on sleep may be ascribed to its pharmacokinetic/pharmacodynamic profile directly on both D1 and D2 receptors.
Collapse
Affiliation(s)
- Mariangela Pierantozzi
- Movement Disorders Centre, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Fabio Placidi
- Sleep Disorders Centre, Neurophysiopathology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Claudio Liguori
- Sleep Disorders Centre, Neurophysiopathology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.
| | - Maria Albanese
- Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Paola Imbriani
- Movement Disorders Centre, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Maria Grazia Marciani
- Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Nicola Biagio Mercuri
- Sleep Disorders Centre, Neurophysiopathology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy; Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy
| | - Paolo Stanzione
- Movement Disorders Centre, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy; Sleep Disorders Centre, Neurophysiopathology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy
| | - Alessandro Stefani
- Movement Disorders Centre, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
48
|
Fifel K, Piggins H, Deboer T. Modeling sleep alterations in Parkinson's disease: How close are we to valid translational animal models? Sleep Med Rev 2016; 25:95-111. [DOI: 10.1016/j.smrv.2015.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 02/18/2015] [Accepted: 02/18/2015] [Indexed: 10/23/2022]
|
49
|
Ehrminger M, Leu-Semenescu S, Cormier F, Corvol JC, Vidailhet M, Debellemaniere E, Brice A, Arnulf I. Sleep aspects on video-polysomnography in LRRK2 mutation carriers. Mov Disord 2015; 30:1839-43. [PMID: 26468079 DOI: 10.1002/mds.26412] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 07/21/2015] [Accepted: 07/31/2015] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Rapid eye movement sleep behavior disorder and sleepiness precede or accompany idiopathic Parkinson's disease (PD), but their presence in subjects with leucine-rich repeat kinase 2 mutations is unknown. METHODS Ten patients with leucine-rich repeat kinase 2-associated PD, four healthy leucine-rich repeat kinase 2 mutation carriers, 20 patients with idiopathic PD, and 12 healthy controls underwent clinical assessments and a nighttime video-polysomnography. RESULTS No sleep changes, no rapid eye movement sleep behavior disorder, or rapid eye movement sleep without atonia was found in the 14 subjects with leucine-rich repeat kinase 2mutations compared with controls, whereas 41% of patients with idiopathic PD had rapid eye movement sleep behavior disorder. Eventually, 20% of patients with leucine-rich repeat kinase 2-associated PD had abnormal periodic leg movements, a frequency similar to the idiopathic PD group frequency. CONCLUSIONS The sleep phenotype in leucine-rich repeat kinase 2 mutations parallels that of idiopathic PD, except for absent rapid eye movement sleep behavior disorder here in the presymptomatic and symptomatic stages.
Collapse
Affiliation(s)
- Mickael Ehrminger
- Ecole Normale Supérieure, Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Sleep Disorders unit and Neurology Department, F-75013, Paris, France
| | - Smaranda Leu-Semenescu
- AP-HP, Hôpital Pitié-Salpêtrière, Sleep Disorders unit and Neurology Department, F-75013, Paris, France.,Sorbonne Universitiés, UPMC Univ Paris 06, UMR_S1127, ICM, F-75013, Paris, France.,INSERM, UMR_S1127 and CIC-1422, ICM, F-75013, Paris, France.,CNRS, UMR_7225, ICM, F-75005, Paris, France
| | - Florence Cormier
- AP-HP, Hôpital Pitié-Salpêtrière, Sleep Disorders unit and Neurology Department, F-75013, Paris, France.,Sorbonne Universitiés, UPMC Univ Paris 06, UMR_S1127, ICM, F-75013, Paris, France.,INSERM, UMR_S1127 and CIC-1422, ICM, F-75013, Paris, France.,CNRS, UMR_7225, ICM, F-75005, Paris, France
| | - Jean-Christophe Corvol
- AP-HP, Hôpital Pitié-Salpêtrière, Sleep Disorders unit and Neurology Department, F-75013, Paris, France.,Sorbonne Universitiés, UPMC Univ Paris 06, UMR_S1127, ICM, F-75013, Paris, France.,INSERM, UMR_S1127 and CIC-1422, ICM, F-75013, Paris, France.,CNRS, UMR_7225, ICM, F-75005, Paris, France
| | - Marie Vidailhet
- AP-HP, Hôpital Pitié-Salpêtrière, Sleep Disorders unit and Neurology Department, F-75013, Paris, France.,Sorbonne Universitiés, UPMC Univ Paris 06, UMR_S1127, ICM, F-75013, Paris, France.,INSERM, UMR_S1127 and CIC-1422, ICM, F-75013, Paris, France.,CNRS, UMR_7225, ICM, F-75005, Paris, France
| | - Eden Debellemaniere
- Sorbonne Universitiés, UPMC Univ Paris 06, UMR_S1127, ICM, F-75013, Paris, France.,INSERM, UMR_S1127 and CIC-1422, ICM, F-75013, Paris, France.,CNRS, UMR_7225, ICM, F-75005, Paris, France
| | - Alexis Brice
- AP-HP, Hôpital Pitié-Salpêtrière, Sleep Disorders unit and Neurology Department, F-75013, Paris, France.,Sorbonne Universitiés, UPMC Univ Paris 06, UMR_S1127, ICM, F-75013, Paris, France.,INSERM, UMR_S1127 and CIC-1422, ICM, F-75013, Paris, France.,CNRS, UMR_7225, ICM, F-75005, Paris, France
| | - Isabelle Arnulf
- AP-HP, Hôpital Pitié-Salpêtrière, Sleep Disorders unit and Neurology Department, F-75013, Paris, France.,Sorbonne Universitiés, UPMC Univ Paris 06, UMR_S1127, ICM, F-75013, Paris, France.,INSERM, UMR_S1127 and CIC-1422, ICM, F-75013, Paris, France.,CNRS, UMR_7225, ICM, F-75005, Paris, France
| |
Collapse
|
50
|
Arnulf I, Neutel D, Herlin B, Golmard JL, Leu-Semenescu S, Cochen de Cock V, Vidailhet M. Sleepiness in Idiopathic REM Sleep Behavior Disorder and Parkinson Disease. Sleep 2015; 38:1529-35. [PMID: 26085299 DOI: 10.5665/sleep.5040] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVE To determine whether patients with idiopathic and symptomatic RBD were sleepier than controls, and if sleepiness in idiopathic RBD predicted earlier conversion to Parkinson disease. METHODS The Epworth Sleepiness Scale (ESS) and its determinants were compared at the time of a video-polysomnography for an RBD diagnosis in patients with idiopathic RBD, in patients with Parkinson disease, and in controls. Whether sleepiness at time of RBD diagnosis predicted an earlier conversion to neurodegenerative diseases was retrospectively analyzed in the followed-up patients. RESULTS The 75 patients with idiopathic RBD were sleepier (ESS: 7.8 ± 4.6) at the time of RBD diagnosis than 74 age- and sex-matched controls (ESS: 5.0 ± 3.6, P < 0.0001). They reached the levels of 114 patients with Parkinson disease (ESS: 8.7 ± 4.8), whether they had (n = 78) or did not have (n = 36) concomitant RBD. The severity of sleepiness in idiopathic RBD correlated with younger age, but not with sleep measures. Among the 69 patients with idiopathic RBD who were followed up for a median 3 years (1-15 years), 16 (23.2%) developed parkinsonism (n = 6), dementia (n = 6), dementia plus parkinsonism (n = 2), and multiple system atrophy (n = 2). An ESS greater than 8 at time of RBD diagnosis predicted a shorter time to phenoconversion to parkinsonism and dementia, from RBD onset, and from RBD diagnosis (when adjusted for age and time between RBD onset and diagnosis). CONCLUSIONS Sleepiness is associated with idiopathic REM sleep behavior disorder and predicts more rapid conversion to parkinsonism and dementia, suggesting it is an early marker of neuronal loss in brainstem arousal systems.
Collapse
Affiliation(s)
- Isabelle Arnulf
- Sorbonne Universities, UPMC Univ Paris 06, Paris, France.,Brain Research Institute (CRICM - UPMC-Paris6), Paris, France.,Sleep Disorders Unit, Pitié-Salpêtrière University Hospital, Paris, France
| | - Dulce Neutel
- Sleep Disorders Unit, Pitié-Salpêtrière University Hospital, Paris, France
| | - Bastien Herlin
- Sleep Disorders Unit, Pitié-Salpêtrière University Hospital, Paris, France
| | - Jean-Louis Golmard
- Department of Biostatistics, Salpêtrière Hospital, ER4, Sorbonne Universites, UPMC Univ Paris 06, Paris, France
| | - Smaranda Leu-Semenescu
- Brain Research Institute (CRICM - UPMC-Paris6), Paris, France.,Sleep Disorders Unit, Pitié-Salpêtrière University Hospital, Paris, France
| | | | - Marie Vidailhet
- Sorbonne Universities, UPMC Univ Paris 06, Paris, France.,Brain Research Institute (CRICM - UPMC-Paris6), Paris, France
| |
Collapse
|