1
|
Arnaldo L, Mena J, Serradell M, Gaig C, Adamuz D, Vilas D, Samaniego D, Ispierto L, Montini A, Mayà G, Álvarez R, Pastor P, Iranzo A, Beyer K. Platelet miRNAs as early biomarkers for progression of idiopathic REM sleep behavior disorder to a synucleinopathy. Sci Rep 2025; 15:12136. [PMID: 40204936 PMCID: PMC11982324 DOI: 10.1038/s41598-025-96926-3] [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: 01/10/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
Individuals diagnosed with isolated REM sleep behavior disorder (IRBD) have a high risk of developing Lewy body disorders (LBD), mainly Parkinson's disease (PD) or dementia with Lewy bodies (DLB). As we have previously identified seven platelet-derived miRNAs as potential biomarkers for DLB, in this pilot study we aimed to investigate whether specific expression changes of these miRNAs are also present in IRBD. RNA was obtained from platelets of individuals with IRBD (n = 29) and controls (n = 34), and miRNA levels were determined with a miRCURY LNA miRNA Custom PCR Panel. miRNA interactomes of deregulated miRNAs were determined, and mRNA quantification of miRNA target genes was carried out using real-time PCR and the ΔΔCt method. We found that the expression of hsa-miR- 139 - 5p (p = 0.010) and hsa-miR- 142 - 3p (p = 0.017) was diminished, while hsa-miR- 191 - 5p (p = 0.023) was increased in platelets of IRBD patients compared with controls. Interactome analysis of these miRNAs showed that hsa-miR- 142 - 3p regulates genes related to the structure and maintenance of the cytoskeleton. Of the 15 genes expressed in platelets, the expression of WASL, a gene involved in actin filament organization, was increased in platelets of IRBD patients. Additionally, WASL expression correlated inversely with hsa-miR- 142 - 3p expression. Since the interactomes of hsa-miR- 139 - 5p and hsa-miR- 191 - 5p play a role in several cancer types, their expression was not addressed. Changes in hsa-miR- 142 - 3p, hsa-miR- 139 - 5p, and hsa-miR- 191 - 5p expression were found in IRBD platelets and might represent early biomarkers for LBD involving cytoskeleton dysfunction. Increased expression of WASL could indicate that altered platelet activation occurs early during the development of LBD.
Collapse
Affiliation(s)
- Laura Arnaldo
- Department of Neuroscience, Research Institute Germans Trias i Pujol, Badalona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jorge Mena
- Department of Neuroscience, Research Institute Germans Trias i Pujol, Badalona, Spain
| | - Mònica Serradell
- Department of Neurology, Sleep Unit, Hospital Clínic de Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain
| | - Carles Gaig
- Department of Neurology, Sleep Unit, Hospital Clínic de Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain
| | - David Adamuz
- Department of Neuroscience, Research Institute Germans Trias i Pujol, Badalona, Spain
| | - Dolores Vilas
- Unit of Neurodegenerative diseases, Department of Neurology, University Hospital Germans Trias I Pujol and the Germans Trias I Pujol Research Institute (IGTP) Badalona, Barcelona, Spain
| | - Daniela Samaniego
- Unit of Neurodegenerative diseases, Department of Neurology, University Hospital Germans Trias I Pujol and the Germans Trias I Pujol Research Institute (IGTP) Badalona, Barcelona, Spain
| | - Lourdes Ispierto
- Unit of Neurodegenerative diseases, Department of Neurology, University Hospital Germans Trias I Pujol and the Germans Trias I Pujol Research Institute (IGTP) Badalona, Barcelona, Spain
| | - Angelica Montini
- Department of Neurology, Sleep Unit, Hospital Clínic de Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain
| | - Gerard Mayà
- Department of Neurology, Sleep Unit, Hospital Clínic de Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain
| | - Ramiro Álvarez
- Unit of Neurodegenerative diseases, Department of Neurology, University Hospital Germans Trias I Pujol and the Germans Trias I Pujol Research Institute (IGTP) Badalona, Barcelona, Spain
| | - Pau Pastor
- Department of Neuroscience, Research Institute Germans Trias i Pujol, Badalona, Spain.
- Department of Neurology, Sleep Unit, Hospital Clínic de Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain.
- Unit of Neurodegenerative diseases, Department of Neurology, University Hospital Germans Trias I Pujol and the Germans Trias I Pujol Research Institute (IGTP) Badalona, Barcelona, Spain.
| | - Alex Iranzo
- Department of Neurology, Sleep Unit, Hospital Clínic de Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain.
- Neurology Service, Sleep Unit, Hospital Clínic de Barcelona, Barcelona, Spain.
| | - Katrin Beyer
- Department of Neuroscience, Research Institute Germans Trias i Pujol, Badalona, Spain.
- Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
2
|
Lucey BP. Sleep Alterations and Cognitive Decline. Semin Neurol 2025. [PMID: 40081821 DOI: 10.1055/a-2557-8422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Sleep disturbances and cognitive decline are intricately connected, and both are prevalent in aging populations and individuals with neurodegenerative disorders such as Alzheimer's disease (AD) and other dementias. Sleep is vital for cognitive functions including memory consolidation, executive function, and attention. Disruption in these processes is associated with cognitive decline, although causal evidence is mixed. This review delves into the bidirectional relationship between alterations in sleep and cognitive impairment, exploring key mechanisms such as amyloid-β accumulation, tau pathology, synaptic homeostasis, neurotransmitter dysregulation, oxidative stress, and vascular contributions. Evidence from both experimental research and population-based studies underscores the necessity of early interventions targeting sleep to mitigate risks of neurodegenerative diseases. A deeper understanding of the interplay between sleep and cognitive health may pave the way for innovative strategies to prevent or reduce cognitive decline through improved sleep management.
Collapse
Affiliation(s)
- Brendan P Lucey
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri
- Center On Biological Rhythms and Sleep, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
3
|
Pérez-Carbonell L, Iranzo A. REM sleep and neurodegeneration. J Sleep Res 2025; 34:e14263. [PMID: 38867555 DOI: 10.1111/jsr.14263] [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: 03/17/2024] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
Several brainstem, subcortical and cortical areas are involved in the generation of rapid eye movement (REM) sleep. The alteration of these structures as a result of a neurodegenerative process may therefore lead to REM sleep anomalies. REM sleep behaviour disorder is associated with nightmares, dream-enacting behaviours and increased electromyographic activity in REM sleep. Its isolated form is a harbinger of synucleinopathies such as Parkinson's disease or dementia with Lewy bodies, and neuroprotective interventions are advocated. This link might also be present in patients taking antidepressants, with post-traumatic stress disorder, or with a history of repeated traumatic head injury. REM sleep likely contributes to normal memory processes. Its alteration has also been proposed to be part of the neuropathological changes occurring in Alzheimer's disease.
Collapse
Affiliation(s)
- Laura Pérez-Carbonell
- Sleep Disorders Centre, Guy's and St Thomas' NHS Foundation Trust, King's College London, London, UK
| | - Alex Iranzo
- Neurology Service, Sleep Disorders Centre, Hospital Clínic de Barcelona, IDIBAPS, CIBERNED, University of Barcelona, Barcelona, Spain
| |
Collapse
|
4
|
Zitser J, Herman T, Giladi N, Hausdorff JM. Waking up to the role of sleep disturbances in freezing of gait among people with Parkinson's disease. Parkinsonism Relat Disord 2025; 133:107342. [PMID: 39988541 DOI: 10.1016/j.parkreldis.2025.107342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/03/2025] [Accepted: 02/15/2025] [Indexed: 02/25/2025]
Affiliation(s)
- Jennifer Zitser
- Department of Neurology, Sieratzki-Sagol Sleep Center, Tel Aviv Sourasky Medical Center, Affiliated with the Faculty of Medical & Health Sciences, Tel Aviv University, Israel; Department of Neurology, Movement Disorders Unit, Tel Aviv Sourasky Medical Center, Israel
| | - Talia Herman
- Center for the Study of Movement, Cognition and Mobility, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Nir Giladi
- Department of Neurology, Movement Disorders Unit, Tel Aviv Sourasky Medical Center, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Department of Neurology, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Jeffrey M Hausdorff
- Center for the Study of Movement, Cognition and Mobility, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Department of Physical Therapy, Faculty of Medical & Health Sciences, Tel Aviv, Israel; Rush Alzheimer's Disease Center and Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
5
|
Schenck CH. REM sleep behaviour disorder (RBD): Personal perspectives and research priorities. J Sleep Res 2025; 34:e14228. [PMID: 38782758 DOI: 10.1111/jsr.14228] [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: 02/12/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024]
Abstract
The formal identification and naming of rapid eye movement (REM) sleep behaviour disorder (RBD) in 1985-1987 is described; the historical background of RBD from 1966 to 1985 is briefly discussed; and RBD milestones are presented. Current knowledge on RBD is identified with reference to recent comprehensive reviews, allowing for a focus on research priorities for RBD: factors and predictors of neurodegenerative phenoconversion from isolated RBD and patient enrolment in neuroprotective trials; isolated RBD clinical research cohorts; epidemiology of RBD; traumatic brain injury, post-traumatic stress disorder, RBD and neurodegeneration; depression, RBD and synucleinopathy; evolution of prodromal RBD to neurodegeneration; gut microbiome dysbiosis and colonic synuclein histopathology in isolated RBD; other alpha-synuclein research in isolated RBD; narcolepsy-RBD; dreams and nightmares in RBD; phasic REM sleep in isolated RBD; RBD, periodic limb movements, periodic limb movement disorder pseudo-RBD; other neurophysiology research in RBD; cardiac scintigraphy (123I-MIBG) in isolated RBD; brain magnetic resonance imaging biomarkers in isolated RBD; microRNAs as biomarkers in isolated RBD; actigraphic, other automated digital monitoring and machine learning research in RBD; prognostic counselling and ethical considerations in isolated RBD; and REM sleep basic science research. RBD research is flourishing, and is strategically situated at an ever-expanding crossroads of clinical (sleep) medicine, neurology, psychiatry and neuroscience.
Collapse
Affiliation(s)
- Carlos H Schenck
- Minnesota Regional Sleep Disorders Center, Department of Psychiatry, Hennepin County Medical Center and University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
6
|
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
|
7
|
Sánchez-Benavides G, Iranzo A, Grau-Rivera O, Giraldo DM, Buongiorno M. Olfactory Dysfunction as a Clinical Marker of Early Glymphatic Failure in Neurodegenerative Diseases. Diagnostics (Basel) 2025; 15:719. [PMID: 40150062 PMCID: PMC11941644 DOI: 10.3390/diagnostics15060719] [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: 02/10/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
An abnormal accumulation of misfolded proteins is a common feature shared by most neurodegenerative disorders. Olfactory dysfunction (OD) is common in the elderly population and is present in 90% of patients with Alzheimer's or Parkinson's disease, usually preceding the cognitive and motor symptoms onset by several years. Early Aβ, tau, and α-synuclein protein aggregates deposit in brain structures involved in odor processing (olfactory bulb and tract, piriform cortex, amygdala, entorhinal cortex, and hippocampus) and seem to underly OD. The glymphatic system is a glial-associated fluid transport system that facilitates the movement of brain fluids and removes brain waste during specific sleep stages. Notably, the glymphatic system became less functional in aging and it is impaired in several conditions, including neurodegenerative diseases. As the nasal pathway has been recently described as the main outflow exit of cerebrospinal fluid and solutes, we hypothesized that OD may indeed be a clinical marker of early glymphatic dysfunction through abnormal accumulation of pathological proteins in olfactory structures. This effect may be more pronounced in peri- and postmenopausal women due to the well-documented impact of estrogen loss on the locus coeruleus, which may disrupt multiple mechanisms involved in glymphatic clearance. If this hypothesis is confirmed, olfactory dysfunction might be considered as a clinical proxy of glymphatic failure in neurodegenerative diseases.
Collapse
Affiliation(s)
- Gonzalo Sánchez-Benavides
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (G.S.-B.); (O.G.-R.)
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28029 Madrid, Spain
| | - Alex Iranzo
- Sleep Disorders Center, Neurology Service, Hospital Clínic Universitari de Barcelona, University of Barcelona, 08036 Barcelona, Spain;
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, 28031 Madrid, Spain
| | - Oriol Grau-Rivera
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; (G.S.-B.); (O.G.-R.)
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28029 Madrid, Spain
| | - Darly Milena Giraldo
- Neurology Department, Vall d’Hebron University Hospital, 08035 Barcelona, Spain;
- Neurovascular Diseases Research Group, Vall d’Hebron Research Institute, 08035 Barcelona, Spain
| | - Mariateresa Buongiorno
- Neurology Department, Vall d’Hebron University Hospital, 08035 Barcelona, Spain;
- Neurovascular Diseases Research Group, Vall d’Hebron Research Institute, 08035 Barcelona, Spain
| |
Collapse
|
8
|
van der Aar JF, van Gilst MM, van den Ende DA, van Gorp H, Anderer P, Pijpers A, Fonseca P, Peri E, Overeem S. Hypnogram and Hypnodensity Analysis of REM Sleep Behaviour Disorder Using Both EEG and HRV-Based Sleep Staging Models. J Sleep Res 2025:e70046. [PMID: 40077890 DOI: 10.1111/jsr.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/10/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
Rapid-eye-movement (REM) sleep behaviour disorder (RBD) is a primary sleep disorder strongly associated with Parkinson's disease. Assessing sleep structure in RBD is important for understanding the underlying pathophysiology and developing diagnostic methods. However, the performance of automated sleep stage classification (ASSC) models is considered suboptimal in RBD, for both models utilising neurological signals ("ExG": EEG, EOG, and chin EMG) and heart rate variability combined with body movements (HRVm). Here, we explore this underperformance through the categorical representation of sleep macrostructure (i.e., hypnogram) and a representation that leverages the underlying probability distribution of ASSCs (i.e., hypnodensity). By comparing the RBD population (n = 36) to a sex- and age-matched group of OSA patients chosen for their anticipated similarly decreased sleep stability, we confirm lower 4-stage classification performance in both ExG-based ASSC (RBD: κ = 0.74, OSA: κ = 0.80) and HRVm-based ASSC (RBD: κ = 0.50, OSA: κ = 0.63). Stages showing lower agreement in RBD, namely, N1 + N2 and REM sleep, exhibited elevated ambiguity in the hypnodensity, indicating more ambiguous classification distributions. Limited differences in bout durations between RBD and OSA suggested sleep instability is not necessarily driving lower agreement in RBD. However, stage transitions in OSA showed more abrupt changes in the underlying probability distribution, while RBD transitions had a more continuous profile, possibly complicating classification. Although both ExG-based and HRVm-based automated sleep staging in RBD remain challenging, hypnodensity analysis is informative for the characterisation of (RBD) sleep and can capture potential drivers of classification disagreement.
Collapse
Affiliation(s)
- Jaap F van der Aar
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Philips Sleep and Respiratory Care, Eindhoven, the Netherlands
| | - Merel M van Gilst
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Center for Sleep Medicine Kempenhaeghe, Heeze, the Netherlands
| | - Daan A van den Ende
- Philips Innovation & Strategy, Department of Innovation Engineering, Eindhoven, the Netherlands
| | - Hans van Gorp
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Philips Sleep and Respiratory Care, Eindhoven, the Netherlands
| | - Peter Anderer
- The Siesta Group Schlafanalyse Gmbh, Vienna, Austria
| | | | - Pedro Fonseca
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Philips Sleep and Respiratory Care, Eindhoven, the Netherlands
| | - Elisabetta Peri
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Sebastiaan Overeem
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Center for Sleep Medicine Kempenhaeghe, Heeze, the Netherlands
| |
Collapse
|
9
|
Xie C, Makwana A, Driver H, Shukla G. Association of early-onset psychiatric disorders with REM sleep behavior disorder - A retrospective study. Sleep Med 2025; 127:127-132. [PMID: 39952778 DOI: 10.1016/j.sleep.2025.01.015] [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: 09/20/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND REM sleep behaviour disorder (RBD) is a known predictor of the subsequent development of neurodegenerative diseases, particularly Parkinson's disease and other alpha synucleinopathies. RBD has also been found to be common among children with other psychiatric disorders such as anxiety, depression, and ADHD. OBJECTIVE This retrospective study aims to analyze the prevalence of early-onset psychiatric disorders among patients referred for RBD to our sleep laboratory. Our hypothesis is that early-onset psychiatric disorders are more common in patients with polysomnographically confirmed RBD. METHODS A retrospective chart review was performed through the Kingston Health Sciences Centre (KHSC) Sleep Laboratory. Data collection involved gathering information regarding the patient's sleep study, psychiatric diagnoses and/or symptoms, mental health medication history and any neurodegenerative conditions noted in hospital clinical notes. RESULTS Patients referred for and polysomnographically confirmed RBD were more likely to have presented with symptoms, or received a clinical diagnosis, of an early-onset psychiatric disorder at 32 % compared to the obstructive sleep apnea (OSA) control group at 3 %. CONCLUSIONS History of early-onset psychiatric disorders is more common among patients referred as RBD compared to a control group of patients with OSA. Future studies are required to confirm the validity and replicability of this finding.
Collapse
Affiliation(s)
- Connie Xie
- Center for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Aditii Makwana
- Center for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Helen Driver
- Department of Medicine, Queen's University, Kingston, ON, Canada; Sleep Disorders Lab and EEG/EMG Dept, Epilepsy Monitoring Unit (EMU) Kingston Health Sciences Centre, Kingston, ON, Canada
| | - Garima Shukla
- Center for Neuroscience Studies, Queen's University, Kingston, ON, Canada; Department of Medicine, Queen's University, Kingston, ON, Canada; Sleep Disorders Lab and EEG/EMG Dept, Epilepsy Monitoring Unit (EMU) Kingston Health Sciences Centre, Kingston, ON, Canada.
| |
Collapse
|
10
|
Ji KH, Yun CH. Brain Health in Sleep Disorders. Sleep Med Clin 2025; 20:57-72. [PMID: 39894599 DOI: 10.1016/j.jsmc.2024.10.004] [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] [Indexed: 02/04/2025]
Abstract
Sleep is a critical determinant of brain health, influencing cognitive, emotional, and physiologic functions. The complex bidirectional relationship between sleep and brain health underscores the importance of sleep in maintaining cognitive function, regulating brain homeostasis, and facilitating the clearance of metabolic waste through the glymphatic system. Chronic sleep deprivation and sleep disorders such as insomnia and obstructive sleep apnea have been shown to negatively impact brain structures and functions. This review discusses the impact of sleep disorders on brain health. It also explores the implications of impaired sleep on cardiovascular health, immune function, and neuroplasticity.
Collapse
Affiliation(s)
- Ki-Hwan Ji
- Department of Neurology, Inje University Busan Paik Hospital, College of Medicine, Inje University, 75 Bokji-ro, Busanjin-gu, Busan 47392, Republic of Korea
| | - Chang-Ho Yun
- Deparment of Neurology, Seoul National University Bundang Hospital and Seoul National University College of Medicine, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, Gyeonggi 13620, Republic of Korea.
| |
Collapse
|
11
|
Stotz S, Bes F, Kunz D. Ikelos-Rating Scale: Validation of a Behavioural Severity Marker in REM Sleep Behaviour Disorder. J Sleep Res 2025:e70019. [PMID: 39957343 DOI: 10.1111/jsr.70019] [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: 11/18/2024] [Revised: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 02/18/2025]
Abstract
The Ikelos-Rating Scale (Ikelos-RS) is a new, expert-interviewed and bedpartner-reported severity marker assessing frequency and expression of isolated REM sleep behaviour disorder (iRBD), a precursor of clinical α-synucleinopathies. This study aimed to validate the Ikelos-RS in 180 patients with three-night PSG-confirmed iRBD (68.4 ± 8.3 years; 139 m). Inter-rater (n = 45) and test-retest reliabilities (n = 25; 174 Ikelos-RS) were evaluated. For construct validity, correlation analyses were performed with: (1) Clinical Global Impressions-Severity (CGI-S; n = 151), (2) REM sleep without atonia (RWA) within videometry (n = 20), (3) RWA at initial diagnosis (n = 131) and changes over time (n = 36), (4) dopamine transporter scintigraphy (DaT-SPECT) at baseline and changes over time (n = 75). RWA- and DaT-SPECT-analyses were conducted for the whole sample ('all') and after excluding confounders ('cleaned'). Correlation analyses indicated high inter-rater (rs = 0.865, p < 0.001) and test-retest reliabilities (rs = 0.900, p < 0.001). Construct validity was supported by associations of Ikelos-RS with (1) CGI-S (rs = 0.845, p < 0.001), (2) RWA within videometric analysis (rs = 0.592, p = 0.006) and at baseline ('all': rs = 0.274, p = 0.002), (3) DaT-binding (z-scores) at baseline in right anterior putamen (AP) ('all': rs = -0.319, p = 0.005) and changes over time, most pronounced in right anterior putamen (AP) ('all': rs = -0.243, p = 0.035; 'cleaned': rs = -0.374, p = 0.008) and left posterior putamen (PP) ('all': rs = -0.259, p = 0.025; 'cleaned': rs = -0.319, p = 0.024). Given its high reliability and construct validity, demonstrated by associations with the best available severity markers DaT-binding ratios and RWA, Ikelos-RS appears to represent a reliable, valid and easy-to-use tool for measuring the severity of iRBD. Thus, Ikelos-RS may prove beneficial in research. Its suitability as a screening tool in older at-risk populations needs to be proven in future studies.
Collapse
Affiliation(s)
- Sophia Stotz
- Clinic for Sleep & Chronomedicine, St. Hedwig-Hospital, Berlin, Germany
- Sleep Research & Clinical Chronobiology, Institute of Physiology, Charité Universitätsmedizin, Berlin, Germany
| | - Frederik Bes
- Clinic for Sleep & Chronomedicine, St. Hedwig-Hospital, Berlin, Germany
- Sleep Research & Clinical Chronobiology, Institute of Physiology, Charité Universitätsmedizin, Berlin, Germany
| | - Dieter Kunz
- Clinic for Sleep & Chronomedicine, St. Hedwig-Hospital, Berlin, Germany
- Sleep Research & Clinical Chronobiology, Institute of Physiology, Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
12
|
Wang L, Cui Y, Han B, Du Y, Salewala KS, Wang S, Zhao W, Zhang H, Wang S, Xu X, Ma J, Zhu Y, Tuo H. Gut microbiota and Parkinson's disease. Chin Med J (Engl) 2025; 138:289-297. [PMID: 39501822 PMCID: PMC11771718 DOI: 10.1097/cm9.0000000000003318] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Indexed: 01/29/2025] Open
Abstract
ABSTRACT Emerging evidence suggests that dysbiosis of the gut microbiota is associated with the pathogenesis of Parkinson's disease (PD), a prevalent neurodegenerative disorder. The microbiota-gut-brain axis plays a crucial role in the development and progression of PD, and numerous studies have demonstrated the potential therapeutic benefits of modulations in the intestinal microbiota. This review provides insights into the characterization of the gut microbiota in patients with PD and highlights associations with clinical symptoms and underlying mechanisms. The discussion underscores the increased influence of the gut microbiota in the pathogenesis of PD. While the relationship is not fully elucidated, existing research demonstrates a strong correlation between changes in the composition of gut microbiota and disease development, and further investigation is warranted to explain the specific underlying mechanisms.
Collapse
Affiliation(s)
- Lin Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Ying Cui
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Bingyu Han
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yitong Du
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | | | - Shiya Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Wenlu Zhao
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Hongxin Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Sichen Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Xinran Xu
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jianpeng Ma
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yan Zhu
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Houzhen Tuo
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
13
|
Fu J, Tang Y, Pan L, Lv K, Cao X, Xu S, Geng D, Yu H, Zhang J. Neuromelanin-MRI identifies locus coeruleus and substantia nigra degeneration as key differentiators in isolated rapid eye movement sleep behavior disorder. Brain Imaging Behav 2025; 19:72-81. [PMID: 39476171 DOI: 10.1007/s11682-024-00926-2] [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] [Accepted: 09/05/2024] [Indexed: 02/23/2025]
Abstract
To explore the neuromelanin depigmentation of locus coeruleus (LC) and substantia nigra pars compacta (SNc) in the isolated rapid eye movement sleep behavior disorder (iRBD) using neuromelanin-sensitive MRI (NM-MRI), and to evaluate its utility for iRBD diagnosis. A total of 25 iRBD patients and 25 healthy controls were recruited and underwent NM-MRI. The contrast-to-noise ratio (CNR) of SNc and LC, and the volume of SNc were compared between groups and evaluated visually. The power of NM measures in discriminating iRBD patients from healthy controls were performed with receiver operating characteristic (ROC) curves and the area under curve (AUC) was calculated. The CNR of SNc and LC, the volume of SNc, the SNc/midbrain volume ratio as well as the visual scores of SNc and LC in iRBD patients were significantly decreased than those in controls (all P < 0.05). For quantitative analysis, the LC CNR acquired the highest accuracy in predicting iRBD (AUC 0.95, sensitivity 80%, specificity 100%), followed by SNc volume (AUC 0.93, sensitivity 88%, specificity 96%) and SNc CNR (AUC 0.74, sensitivity 92%, specificity 44%). For visual analysis, the accuracy of the visual score for SNc and LC were 78% (sensitivity 68%, specificity 88%) and 86% (sensitivity 88%, specificity 84%), respectively. The NM in the SNc and LC regions were significantly reduced in iRBD patients. NM measures showed good capability in discriminating iRBD from controls, suggesting that NM-MRI may be a valuable screening tool for iRBD.
Collapse
Affiliation(s)
- Junyan Fu
- Department of Radiology, Huashan Hospital, Fudan University, No.12 Wulumuqi Road (Middle), Shanghai, 200040, China
| | - Ye Tang
- Department of Radiology, Huashan Hospital, Fudan University, No.12 Wulumuqi Road (Middle), Shanghai, 200040, China
| | - Lei Pan
- Department of Radiology, Huashan Hospital, Fudan University, No.12 Wulumuqi Road (Middle), Shanghai, 200040, China
| | - Kun Lv
- Department of Radiology, Huashan Hospital, Fudan University, No.12 Wulumuqi Road (Middle), Shanghai, 200040, China
| | - Xin Cao
- Department of Radiology, Huashan Hospital, Fudan University, No.12 Wulumuqi Road (Middle), Shanghai, 200040, China
| | - Siting Xu
- Department of Radiology, Huashan Hospital, Fudan University, No.12 Wulumuqi Road (Middle), Shanghai, 200040, China
| | - Daoying Geng
- Department of Radiology, Huashan Hospital, Fudan University, No.12 Wulumuqi Road (Middle), Shanghai, 200040, China
| | - Huan Yu
- Department of Neurology, Huashan Hospital, Fudan University, No.12 Wulumuqi Road (Middle), Shanghai, 200040, China
| | - Jun Zhang
- Department of Radiology, Huashan Hospital, Fudan University, No.12 Wulumuqi Road (Middle), Shanghai, 200040, China.
- Department of Radiology, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai, 200040, China.
| |
Collapse
|
14
|
Trujillo P, O'Rourke KR, Roman OC, Song AK, Hett K, Cooper A, Black BK, Donahue MJ, Shibao CA, Biaggioni I, Claassen DO. Central Involvement in Pure Autonomic Failure: Insights from Neuromelanin-Sensitive Magnetic Resonance Imaging and 18F-Fluorodopa-Positron Emission Tomography. Mov Disord 2025. [PMID: 39825743 DOI: 10.1002/mds.30119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/16/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Central synucleinopathies, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA), involve alpha-synuclein accumulation and dopaminergic cell loss in the substantia nigra (SN) and locus coeruleus (LC). Pure autonomic failure (PAF), a peripheral synucleinopathy, often precedes central synucleinopathies. OBJECTIVES To assess early brain involvement in PAF using neuromelanin-sensitive magnetic resonance imaging (NM-MRI) and fluorodopa-positron emission tomography (FDOPA-PET), and to determine whether PAF patients with a high likelihood ratio (LR) for conversion to a central synucleinopathy exhibit reduced NM-MRI contrast in the LC and SN compared with controls and low-LR patients. METHODS Participants with PAF (n = 23) were categorized as high-LR (n = 13) or low-LR (n = 10) for conversion to central synucleinopathy. Additional participants included PD (n = 22), DLB (n = 8), and age- and sex-matched healthy controls (n = 23). NM-MRI at 3 T was used to quantify contrast ratios in the LC and SN, while FDOPA-PET measured presynaptic dopamine synthesis. Linear regression analyses, adjusted for age and sex, were used to compare NM-MRI contrast across groups. RESULTS High-LR PAF patients showed reduced contrast in the LC and SN compared with controls and low-LR PAF patients, with values similar to PD and DLB. The NM-MRI contrast in the SN correlated with dopamine uptake in the striatum. Longitudinal imaging in PAF patients (n = 6) demonstrated reduced NM-MRI and PET values in individuals who developed central synucleinopathies. CONCLUSIONS NM-MRI and FDOPA-PET may serve as potential biomarkers for early brain involvement and predicting progression to central synucleinopathies in PAF and could help identify patients for early intervention. © 2025 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Paula Trujillo
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kaitlyn R O'Rourke
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Olivia C Roman
- Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alexander K Song
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kilian Hett
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Amy Cooper
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bonnie K Black
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Manus J Donahue
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Cyndya A Shibao
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Italo Biaggioni
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Daniel O Claassen
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
15
|
Mariño N, Serradell M, Gaig C, Mayà G, Montini A, Matos N, Pont-Sunyer C, Uscamaita K, Marrero-González P, Buongiorno M, Iranzo A. Audiovisual analysis of the diagnostic video polysomnography in patients with isolated REM sleep behavior disorder. J Neurol 2025; 272:146. [PMID: 39812859 DOI: 10.1007/s00415-024-12761-y] [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: 07/15/2024] [Revised: 09/15/2024] [Accepted: 10/29/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND The diagnosis of isolated REM sleep behavior disorder (IRBD) requires video polysomnography (V-PSG) showing increased muscle activity and abnormal behaviors in REM sleep. OBJECTIVE To describe in IRBD the behavioral manifestations occurring during REM sleep in the diagnostic V-PSG. METHODS This is a systematic audiovisual V-PSG analysis of consecutive IRBD patients. According to the International RBD Study Group recommendations, REM sleep movements and vocalizations were classified into categories and severity. RESULTS We analyzed the V-PSG of 62 IRBD patients with a mean age of 67.6 ± 8.1 years. Of 6,330 30-s epochs of REM sleep, 55.1% epochs exhibited motor events, 5.5% contained vocalizations and 39.4% were silent. Among the epochs with motor manifestations, 66.1% contained simple minor movements, 25.0% simple major and 8.9% complex movements. Motor severity of the epochs was mild in 82.2%, moderate in 13.2% and severe in 4.6%. Most movements were bilateral (62.4%) and located in the upper limbs (42.5%). Of the epochs with vocalizations, 61.5% were simple minor, 20.7% complex and 17.8% simple major of mild (72.7%), moderate (23.0%) and severe (4.3%) severity. Complex movements occurred in 87.1% of the patients and complex vocalizations in 38.7%. CONCLUSIONS In IRBD, the most common manifestations in REM sleep are simple minor movements and vocalizations of mild intensity. Complex movements are observed during REM sleep in most patients but are much less frequent than simple minor and major motor events. These findings should be considered for the routine diagnosis of IRBD when reviewing the V-PSG studies.
Collapse
Affiliation(s)
- Nathalie Mariño
- Sleep Unit, Neurology Service, Hospital Clínic Barcelona, IDIBAPS, CIBERNED, Universitat de Barcelona, Villarroel 170, 08036, Barcelona, Spain
| | - Mónica Serradell
- Sleep Unit, Neurology Service, Hospital Clínic Barcelona, IDIBAPS, CIBERNED, Universitat de Barcelona, Villarroel 170, 08036, Barcelona, Spain
| | - Carles Gaig
- Sleep Unit, Neurology Service, Hospital Clínic Barcelona, IDIBAPS, CIBERNED, Universitat de Barcelona, Villarroel 170, 08036, Barcelona, Spain
| | - Gerard Mayà
- Sleep Unit, Neurology Service, Hospital Clínic Barcelona, IDIBAPS, CIBERNED, Universitat de Barcelona, Villarroel 170, 08036, Barcelona, Spain
| | - Angelica Montini
- Sleep Unit, Neurology Service, Hospital Clínic Barcelona, IDIBAPS, CIBERNED, Universitat de Barcelona, Villarroel 170, 08036, Barcelona, Spain
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Nuria Matos
- Althaia Xarxa Assistencial Universitària de Manresa, Barcelona, Spain
- Facultat de Medicina, Universitat de Vic-Central de Catalunya (UVIC-UCC), Barcelona, Spain
| | - Claustre Pont-Sunyer
- Movement Disorders Unit, Neurology Service, Hospital General de Granollers, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Karol Uscamaita
- Neurology Service, Sleep Disorders Unit, Hospital Universitari Sagrat Cor, Grupo Quirónsalud, Barcelona, Spain
| | - Paula Marrero-González
- Neurology Department, Hospital de Sant Joan Despí Moisès Broggi, Consorci Sanitari Integral Barcelona Spain, Barcelona, Spain
| | - Mariateresa Buongiorno
- Neurodegenerative Unit, Neurology Service, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Iranzo
- Sleep Unit, Neurology Service, Hospital Clínic Barcelona, IDIBAPS, CIBERNED, Universitat de Barcelona, Villarroel 170, 08036, Barcelona, Spain.
| |
Collapse
|
16
|
Yuan Y, Li Y, Zhang H, Zang Y, Liu X, Hou Y, Zhan S, Cai Y, Mao W, Chan P. Factors associated with phenoconversion of idiopathic rapid eye movement sleep behavior disorder: a prospective study. NPJ Parkinsons Dis 2025; 11:10. [PMID: 39762239 PMCID: PMC11704329 DOI: 10.1038/s41531-024-00856-2] [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: 04/09/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
This study explores the effect of risk factors on the progression of idiopathic rapid eye movement (REM) sleep behavior disorder (iRBD) to α-synucleinopathies in a Chinese cohort. Patients with iRBD were enrolled and assessed for environmental factors and lifestyle using standardized structured questionnaires at baseline. All patients were prospectively followed for phenoconversion monitoring. The cumulative incidence was estimated using survival analysis. Of 155 iRBD enrolled in the cohort, follow-up information was available in 141 patients. The phenoconversion rate was 16.3% after 3 years, 27.6% after 5 years, and 57.2% after 10 years. Eighteen participants converted within 3 years, 27 converted within 5 years, and 36 converted within 10 years. IRBD with positive family history of parkinsonism had an increased risk of being converted to α-synucleinopathies, while tea drinking was associated with a decreased phenoconversion risk. Our findings shed light on a potential application of tea drinking in modifying iRBD progression.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yuan Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Hui Zhang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yajie Zang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xiaonan Liu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yue Hou
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Shuqin Zhan
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yanning Cai
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
- Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing Key Laboratory on Parkinson's Disease, Parkinson's Disease Center for Beijing Institute on Brain Disorders, Clinical and Research Center for Parkinson's Disease of Capital Medical University, Beijing, China
| | - Wei Mao
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.
| | - Piu Chan
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China.
- Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing Key Laboratory on Parkinson's Disease, Parkinson's Disease Center for Beijing Institute on Brain Disorders, Clinical and Research Center for Parkinson's Disease of Capital Medical University, Beijing, China.
- National Clinical Research Center for Geriatric Disorders, Beijing, China.
| |
Collapse
|
17
|
Pinoli M, Terzaghi M, Marino F, Comi C, Versino M, Cosentino M. CD4+ T-cell transcription factors predict phenoconversion in idiopathic rapid eye movement sleep behavior disorder. Future Sci OA 2024; 10:2418821. [PMID: 39539158 PMCID: PMC11572078 DOI: 10.1080/20565623.2024.2418821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Aim: Early biomarkers of phenoconversion to neurodegeneration are crucial to identify individuals at high risk. In patients with idiopathic REM sleep behavior disorder (iRBD), the strongest risk factor for neurodegeneration, CD4+ T cells exhibit a peculiar transcription factor pattern.Objective: To assess transcription factor mRNA levels in CD4+ T cells as predictive biomarkers of phenoconversion in iRBD patients.Methods: iRBD patients were followed prospectively. ROC curve analysis and Kaplan-Meier curves were used to assess the discrimination between converters and non-converters.Results: CD4+ T cells from converters had higher STAT1, and lower GATA3 and FOXP3 mRNA levels. Hazard ratio was 58.3 (95% CI: 6.2-547.1) for higher STAT1, 101.2 (95% CI: 16.8-609.4) for lower GATA3 and 15.7 (2.7-91.4) for lower FOXP3.Conclusion: STAT1, GATA3 and FOXP3 mRNA levels in CD4+ T cells are promising predictive biomarkers of phenoconversion in iRBD patients.
Collapse
Affiliation(s)
- Monica Pinoli
- Center of Research in Medical Pharmacology, University of Insubria, Varese (I), Italy
| | - Michele Terzaghi
- Unit of Sleep Medicine & Epilepsy, IRCCS Mondino Foundation, Pavia, Italy
| | - Franca Marino
- Center of Research in Medical Pharmacology, University of Insubria, Varese (I), Italy
| | - Cristoforo Comi
- Center of Research in Medical Pharmacology, University of Insubria, Varese (I), Italy
- Movement Disorders Centre, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Maurizio Versino
- Department of Medicine & Surgery, University of Insubria, Varese, Italy
| | - Marco Cosentino
- Center of Research in Medical Pharmacology, University of Insubria, Varese (I), Italy
| |
Collapse
|
18
|
Hernandez J, Lina JM, Dubé J, Lafrenière A, Gagnon JF, Montplaisir JY, Postuma RB, Carrier J. Electroencephalogram rhythmic and arrhythmic spectral components and functional connectivity at resting state may predict the development of synucleinopathies in idiopathic rapid eye movement sleep behavior disorder. Sleep 2024; 47:zsae074. [PMID: 38497896 PMCID: PMC11632188 DOI: 10.1093/sleep/zsae074] [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: 10/30/2023] [Revised: 01/25/2024] [Indexed: 03/19/2024] Open
Abstract
STUDY OBJECTIVES Idiopathic/isolated rapid eye movement-sleep behavior disorder (iRBD) often precedes the onset of synucleinopathies. Here, we investigated whether baseline resting-state EEG advanced spectral power and functional connectivity differed between iRBD patients who converted towards a synucleinopathy at follow-up and those who did not. METHODS Eighty-one participants with iRBD (66.89 ± 6.91 years) underwent a baseline resting-state EEG recording, a neuropsychological assessment, and a neurological examination. We estimated EEG power spectral density using standard analyses and derived spectral estimates of rhythmic and arrhythmic components. Global and pairwise EEG functional connectivity analyses were computed using the weighted phase-lag index (wPLI). Pixel-based permutation tests were used to compare groups. RESULTS After a mean follow-up of 5.01 ± 2.76 years, 34 patients were diagnosed with a synucleinopathy (67.81 ± 7.34 years) and 47 remained disease-free (65.53 ± 7.09 years). Among patients who converted, 22 were diagnosed with Parkinson's disease and 12 with dementia with Lewy bodies. As compared to patients who did not convert, patients who converted exhibited at baseline higher relative theta standard power, steeper slopes of the arrhythmic component and higher theta rhythmic power mostly in occipital regions. Furthermore, patients who converted showed higher beta global wPLI but lower alpha wPLI between left temporal and occipital regions. CONCLUSIONS Analyses of resting-state EEG rhythmic and arrhythmic components and functional connectivity suggest an imbalanced excitatory-to-inhibitory activity within large-scale networks, which is associated with later development of a synucleinopathy in patients with iRBD.
Collapse
Affiliation(s)
- Jimmy Hernandez
- Center for Advanced Research in Sleep Medicine, Research center, CIUSSS du Nord de l’Île-de-Montréal, Montreal, QC, Canada
- Department of Neuroscience, Université de Montréal, Montreal, QC, Canada
| | - Jean-Marc Lina
- Center for Advanced Research in Sleep Medicine, Research center, CIUSSS du Nord de l’Île-de-Montréal, Montreal, QC, Canada
- Department of electrical engineering, École de technologie supérieure, Montreal, QC, Canada
| | - Jonathan Dubé
- Center for Advanced Research in Sleep Medicine, Research center, CIUSSS du Nord de l’Île-de-Montréal, Montreal, QC, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
| | - Alexandre Lafrenière
- Center for Advanced Research in Sleep Medicine, Research center, CIUSSS du Nord de l’Île-de-Montréal, Montreal, QC, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
| | - Jean-François Gagnon
- Center for Advanced Research in Sleep Medicine, Research center, CIUSSS du Nord de l’Île-de-Montréal, Montreal, QC, Canada
- Department of Psychology, Université du Québec à Montréal, Montreal, QC, Canada
| | - Jacques-Yves Montplaisir
- Center for Advanced Research in Sleep Medicine, Research center, CIUSSS du Nord de l’Île-de-Montréal, Montreal, QC, Canada
- Department of psychiatry, Université de Montréal, Montreal, QC, Canada
| | - Ronald B Postuma
- Center for Advanced Research in Sleep Medicine, Research center, CIUSSS du Nord de l’Île-de-Montréal, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, QC, Canada
| | - Julie Carrier
- Center for Advanced Research in Sleep Medicine, Research center, CIUSSS du Nord de l’Île-de-Montréal, Montreal, QC, Canada
- Department of Psychology, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
19
|
Tener SJ, Kim CE, Lee J, Oraedu K, Gatto JA, Chang TY, Lam C, Schanta R, Jankowski MS, Park SJ, Hurley JM, Ulgherait M, Canman JC, Ja WW, Collins DB, Shirasu-Hiza M. Investigating the consequences of chronic short sleep for metabolism and survival of oxidative stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.01.626207. [PMID: 39677628 PMCID: PMC11642809 DOI: 10.1101/2024.12.01.626207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
In previous work, we found that short sleep caused sensitivity to oxidative stress; here we set out to characterize the physiological state of a diverse group of chronically short-sleeping mutants during hyperoxia as an acute oxidative stress. Using RNA-sequencing analysis, we found that short-sleeping mutants had a normal transcriptional oxidative stress response relative to controls. In both short-sleeping mutants and controls, hyperoxia led to downregulation of glycolytic genes and upregulation of genes involved in fatty acid metabolism, reminiscent of metabolic shifts during sleep. We hypothesized that short-sleeping mutants may be sensitive to hyperoxia because of defects in metabolism. Consistent with this, short-sleeping mutants were sensitive to starvation. Using metabolomics, we identified a pattern of low levels of long chain fatty acids and lysophospholipids in short-sleeping mutants relative to controls during hyperoxia, suggesting a defect in lipid metabolism. Though short-sleeping mutants did not have common defects in many aspects of lipid metabolism (basal fat stores, usage kinetics during hyperoxia, respiration rates, and cuticular hydrocarbon profiles), they were all sensitive to dehydration, suggesting a general defect in cuticular hydrocarbons, which protect against dehydration. To test the bi-directionality of sleep and lipid metabolism, we tested flies with both diet-induced obesity and genetic obesity. Flies with diet-induced obesity had no sleep or oxidative stress phenotype; in contrast, the lipid metabolic mutant, brummer , slept significantly more than controls but was sensitive to oxidative stress. Previously, all short sleepers tested were sensitive and all long sleepers resistant to oxidative stress. brummer mutants, the first exceptions to this rule, lack a key enzyme required to mobilize fat stores, suggesting that a defect in accessing lipid stores can cause sensitivity to oxidative stress. Taken together, we found that short-sleeping mutants have many phenotypes in common: sensitivity to oxidative stress, starvation, dehydration, and defects in lipid metabolites. These results argue against a specific role for sleep as an antioxidant and suggest the possibility that lipid metabolic defects underlie the sensitivity to oxidative stress of short-sleeping mutants.
Collapse
|
20
|
Kakazu VA, Poyares D, Tufik S, Ferraz HB, Pires GN. History of symptoms consistent with REM sleep behavior disorder in a population with Parkinson's Disease. Sleep Med 2024; 124:308-314. [PMID: 39366086 DOI: 10.1016/j.sleep.2024.09.042] [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: 12/11/2023] [Revised: 03/02/2024] [Accepted: 09/28/2024] [Indexed: 10/06/2024]
Abstract
REM Sleep Behavioral Disorder (RBD) is a parasomnia marked by the maintenance of muscle tone during REM sleep. Evidence has placed RBD as one of the possible prodromal stages of Parkinson's Disease (PD), but data on the proportion of people with PD who have had symptoms of RBD are limited. This study aimed to investigate the history of symptoms compatible with RBD in a population with PD. The sample was composed by 73 patients with clinically diagnosed PD being followed up at a reference outpatient setting, compared to 73 age- and sex-matched individuals with no PD. The evaluation of symptoms compatible with RBD was performed using the Brazilian version of the RBD Screening Questionnaire (RBDSQ). The prevalence of symptoms compatible with RBD was 65 % for PD and 10.09 % for controls. The RBDSQ score was significantly higher in the PD group (6.03 ± 0.35) in comparison to the control group (2.38 ± 0.23). The odds ratio for presenting previous RBD-compatible symptoms was 12.09 in favor of positive PD cases. PD diagnosis has the following diagnostic properties in relation to presenting RBD symptoms: sensitivity of 0.65, specificity of 0.86, positive predictive value of 0.82 and negative predictive value of 0.71. In conclusion, the proportion of PD patients showing RBD symptoms is high, corroborating the expected neuroprogression of the disease on a case-control design comprising outpatient PD cases. Clinical practitioners should include evaluations of RBD-compatible symptoms during the PD assessment and, if positive, forward to a sleep specialist.
Collapse
Affiliation(s)
- Viviane Akemi Kakazu
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Dalva Poyares
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil; Sleep Institute, São Paulo, Brazil
| | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil; Sleep Institute, São Paulo, Brazil
| | | | - Gabriel Natan Pires
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil; Sleep Institute, São Paulo, Brazil.
| |
Collapse
|
21
|
Alexandres CA, McCarter SJ, Tabatabai GM, LeClair-Visonneau L, Feemster JC, Gossard TR, Strainis EP, Jagielski JT, Kelleher MR, Finstuen T, Ali F, Botha H, Graff-Radford J, Olson EJ, Sandness DJ, Morgenthaler TJ, Kantarci K, Savica R, Singer W, Covassin N, Somers VK, Kirkland JL, Junna M, Lipford M, Matarese CA, Moore JL, Tippmann-Peikert M, Carvalho DZ, Boeve BF, Silber MH, St Louis EK. Phenoconversion in Women and Men With Isolated REM Sleep Behavior Disorder: A Retrospective Cohort Study. Neurology 2024; 103:e209993. [PMID: 39454123 PMCID: PMC11515114 DOI: 10.1212/wnl.0000000000209993] [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: 11/09/2023] [Accepted: 09/16/2024] [Indexed: 10/27/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Isolated REM sleep behavior disorder (iRBD) is strongly associated with synucleinopathies. Previous iRBD cohort studies have primarily focused on older (>50 years), male-predominant cohorts. Risk of phenoconversion in women and younger adults remains unclear. The study aimed to determine clinical features associated with conversion to a defined neurodegenerative disorder in women and men with iRBD. METHODS One hundred eighty-six women and 186 men with iRBD were matched by polysomnography month. Baseline clinical variables and subsequent neurodegenerative outcomes were abstracted by chart review. Kaplan-Meier curves assessed conversion rates. Cox proportional hazards modeling evaluated factors associated with phenoconversion risk. RESULTS Age at iRBD diagnosis was younger in women compared with men (54.9 vs 62.5 years, p < 0.01). Forty-eight patients (12.9%), including 18 women (9.7%) and 30 men (16.1%), phenoconverted during a median follow-up of 6.0 years. Conversion rates were lower in antidepressant users and patients with chronic pain or psychiatric comorbidity while rates were higher in those with vascular comorbidity. Only age at diagnosis (HR 1.09, 95% CI 1.06-1.13) was associated with phenoconversion after adjusting for RBD symptom duration; sex; antidepressant use; and psychiatric, chronic pain, and vascular comorbidities. DISCUSSION Age at diagnosis was independently associated with phenoconversion risk in women and men with iRBD.
Collapse
Affiliation(s)
- Christina A Alexandres
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Stuart J McCarter
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Grace M Tabatabai
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Laurene LeClair-Visonneau
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - John C Feemster
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Thomas R Gossard
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Emma P Strainis
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Jack T Jagielski
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Makayla R Kelleher
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Thomas Finstuen
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Farwa Ali
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Hugo Botha
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Jonathan Graff-Radford
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Eric J Olson
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - David J Sandness
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Timothy J Morgenthaler
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Kejal Kantarci
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Rodolfo Savica
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Wolfgang Singer
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Naima Covassin
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Virend K Somers
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - James L Kirkland
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Mithri Junna
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Melissa Lipford
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Christine A Matarese
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - James L Moore
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Maja Tippmann-Peikert
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Diego Z Carvalho
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Bradley F Boeve
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Michael H Silber
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Erik K St Louis
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| |
Collapse
|
22
|
Zhang RY, Li JY, Liu YN, Zhang ZX, Zhao J, Li FJ. The causal relationship between type 2 diabetes mellitus and isolated REM sleep behavior disorder: results from multivariable and network Mendelian randomization analysis. Front Endocrinol (Lausanne) 2024; 15:1408053. [PMID: 39655344 PMCID: PMC11625559 DOI: 10.3389/fendo.2024.1408053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/25/2024] [Indexed: 12/12/2024] Open
Abstract
Objectives To investigate the causal relationship between type 2 diabetes mellitus (T2DM, exposure) and isolated REM sleep behavior disorder (iRBD, outcome). Methods Genome-wide association study (GWAS) data for iRBD comprised 9,447 samples, including 1,061 iRBD cases from the International RBD Study Group. Initially, we performed linkage disequilibrium score regression (LDSC) to explore the genetic correlation between T2DM and iRBD. Then the two-sample univariate MR (UVMR) analysis was conducted to examine the effects of T2DM and blood sugar metabolism-related factors on iRBD. Subsequently, we applied multivariable MR (MVMR) methods to further adjust for confounders. Lastly, we executed a network MR analysis, with cytokines and immune cell characteristics as potential mediators, aiming to investigate indirect effect of T2DM on iRBD. Results Results from LDSC suggest a genetic correlation between T2DM and iRBD (rg=0.306, P=0.029). UVMR analysis indicates that both T2DM (Odds Ratio [95% Confidence Interval] = 1.19 [1.03, 1.37], P = 0.017) and high blood glucose levels (1.55 [1.04, 2.30], P = 0.032) are risk factors for iRBD. Even after adjusting for confounders in MVMR, the association between T2DM and iRBD remains robust. Finally, results from network MR analysis suggest that T2DM may indirectly promote the development of iRBD by reducing levels of Stromal Cell-Derived Factor 2 in circulation and by increasing BAFF-receptor expression in IgD- CD38- B cells. Conclusions T2DM may promote the onset of iRBD by influencing immune-inflammatory responses. Our findings provide valuable insights and directions for understanding the pathogenesis of iRBD, identifying high-risk groups, and discovering new therapeutic targets.
Collapse
Affiliation(s)
- Ru-Yu Zhang
- Department of Pulmonary and Critical Care Medicine, First People's Hospital of Zigong, Zigong, Sichuan, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jin-Yu Li
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu-Ning Liu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zi-Xuan Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jie Zhao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fu-Jia Li
- Department of Pulmonary and Critical Care Medicine, First People's Hospital of Zigong, Zigong, Sichuan, China
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| |
Collapse
|
23
|
Miranda NC, Oliveira LM, Moreira TS, Ramirez JM, Kalume F, Takakura AC. Sleep-related respiratory disruptions and laterodorsal tegmental nucleus in a mouse model of Parkinson's disease. iScience 2024; 27:111251. [PMID: 39563887 PMCID: PMC11574806 DOI: 10.1016/j.isci.2024.111251] [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: 06/07/2024] [Revised: 08/27/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative disorder affecting the motor system, with non-classic symptoms such as sleep disturbances and respiratory dysfunctions. These issues reflect a complex pathophysiological interaction that severely impacts quality of life. Using a 6-hydroxydopamine (6-OHDA) mouse model of PD, we investigated these connections by analyzing sleep patterns and respiratory parameters during non-rapid eye movement (NREM) and rapid eye movement (REM) sleep. Our findings revealed altered breathing, including reduced respiratory frequency and increased apneas during both NREM and REM. To address these abnormalities, we employed chemogenetic stimulation of cholinergic neurons in the laterodorsal tegmental nucleus (LDTg), a key region for sleep-wake regulation and respiratory modulation. This intervention normalized respiratory function. These results highlight the critical role of LDTg cholinergic neurons in the coordinating sleep and breathing, suggesting that targeting these neurons could offer a therapeutic strategy for managing PD-related respiratory complications.
Collapse
Affiliation(s)
- Nicole C Miranda
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, São Paulo 05508-000, SP, Brazil
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Avenue, Seattle, WA 98101, USA
| | - Luiz M Oliveira
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Avenue, Seattle, WA 98101, USA
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, São Paulo 05508-000, SP, Brazil
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Avenue, Seattle, WA 98101, USA
- Department of Neurological Surgery, University of Washington, 1900 9th Avenue, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington, 1900 9th Avenue, Seattle, WA 98101, USA
| | - Franck Kalume
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Avenue, Seattle, WA 98101, USA
- Department of Neurological Surgery, University of Washington, 1900 9th Avenue, Seattle, WA 98101, USA
| | - Ana C Takakura
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, São Paulo 05508-000, SP, Brazil
| |
Collapse
|
24
|
Wang X, Zheng Y, Cai H, Kou W, Yang C, Li S, Zhu B, Wu J, Zhang N, Feng T, Li X, Xiao F, Yu Z. α-Synuclein species in plasma neuron-derived extracellular vesicles as biomarkers for iRBD. Ann Clin Transl Neurol 2024; 11:2891-2903. [PMID: 39291779 PMCID: PMC11572749 DOI: 10.1002/acn3.52200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/05/2024] [Accepted: 08/25/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVE Isolated REM sleep behavior disorder (iRBD) is considered as the strongest predictor of Parkinson's disease (PD). Reliable and accurate biomarkers for iRBD detection and the prediction of phenoconversion are in urgent need. This study aimed to investigate whether α-Synuclein (α-Syn) species in plasma neuron-derived extracellular vesicles (NDEVs) could differentiate between iRBD patients and healthy controls (HCs). METHODS Nanoscale flow cytometry was used to detect α-Syn-containing NDEVs in plasma. RESULTS A total of 54 iRBD patients and 53 HCs were recruited. The concentrations of total α-Syn, α-Syn aggregates, and phosphorylated α-Syn at Ser129 (pS129)-containing NDEVs in plasma of iRBD individuals were significantly higher than those in HCs (p < 0.0001 for all). In distinguishing between iRBD and HCs, the area under the receiver operating characteristic (ROC) curve (AUC) for an integrative model incorporating the levels of α-Syn, pS129, and α-Syn aggregate-containing NDEVs in plasma was 0.965. This model achieved a sensitivity of 94.3% and a specificity of 88.9%. In iRBD group, the concentrations of α-Syn aggregate-containing NDEVs exhibited a negative correlation with Sniffin' Sticks olfactory scores (r = -0.351, p = 0.039). Smokers with iRBD exhibited lower levels of α-Syn aggregates and pS129-containing NDEVs in plasma compared to nonsmokers (pα-Syn aggregates = 0.014; ppS129 = 0.003). INTERPRETATION The current study demonstrated that the levels of total α-Syn, α-Syn aggregates, and pS129-containing NDEVs in the plasma of individuals with iRBD were significantly higher compared to HCs. The levels of α-Syn species-containing NDEVs in plasma may serve as biomarkers for iRBD.
Collapse
Affiliation(s)
- Xuemei Wang
- Center for Movement Disorders, Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Yuanchu Zheng
- Center for Movement Disorders, Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Huihui Cai
- Center for Movement Disorders, Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Wenyi Kou
- Center for Movement Disorders, Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Chen Yang
- Center for Movement Disorders, Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Siming Li
- Center for Movement Disorders, Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Bingxu Zhu
- Center for Movement Disorders, Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Jiayi Wu
- Center for Movement Disorders, Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Ning Zhang
- Department of Neuropsychiatry and Behavioral Neurology and Clinical PsychologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Tao Feng
- Center for Movement Disorders, Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Xiaohong Li
- Department of NeurologyAffiliated Dalian Municipal Friendship Hospital of Dalian Medical UniversityDalianChina
| | - Fulong Xiao
- Division of Sleep MedicinePeking University People's HospitalBeijingChina
| | - Zhenwei Yu
- Department of PathophysiologyBeijing Neurosurgical Institute, Capital Medical UniversityBeijingChina
| |
Collapse
|
25
|
Illner V, Novotný M, Kouba T, Tykalová T, Šimek M, Sovka P, Švihlík J, Růžička E, Šonka K, Dušek P, Rusz J. Smartphone Voice Calls Provide Early Biomarkers of Parkinsonism in Rapid Eye Movement Sleep Behavior Disorder. Mov Disord 2024; 39:1752-1762. [PMID: 39001636 DOI: 10.1002/mds.29921] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/03/2024] [Accepted: 06/21/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Speech dysfunction represents one of the initial motor manifestations to develop in Parkinson's disease (PD) and is measurable through smartphone. OBJECTIVE The aim was to develop a fully automated and noise-resistant smartphone-based system that can unobtrusively screen for prodromal parkinsonian speech disorder in subjects with isolated rapid eye movement sleep behavior disorder (iRBD) in a real-world scenario. METHODS This cross-sectional study assessed regular, everyday voice call data from individuals with iRBD compared to early PD patients and healthy controls via a developed smartphone application. The participants also performed an active, regular reading of a short passage on their smartphone. Smartphone data were continuously collected for up to 3 months after the standard in-person assessments at the clinic. RESULTS A total of 3525 calls that led to 5990 minutes of preprocessed speech were extracted from 72 participants, comprising 21 iRBD patients, 26 PD patients, and 25 controls. With a high area under the curve of 0.85 between iRBD patients and controls, the combination of passive and active smartphone data provided a comparable or even more sensitive evaluation than laboratory examination using a high-quality microphone. The most sensitive features to induce prodromal neurodegeneration in iRBD included imprecise vowel articulation during phone calls (P = 0.03) and monopitch in reading (P = 0.05). Eighteen minutes of speech corresponding to approximately nine calls was sufficient to obtain the best sensitivity for the screening. CONCLUSION We consider the developed tool widely applicable to deep longitudinal digital phenotyping data with future applications in neuroprotective trials, deep brain stimulation optimization, neuropsychiatry, speech therapy, population screening, and beyond. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Vojtěch Illner
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Michal Novotný
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Tomáš Kouba
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Tereza Tykalová
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Michal Šimek
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Pavel Sovka
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Jan Švihlík
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
- Department of Mathematics, Informatics and Cybernetics, Faculty of Chemical Engineering, University of Chemistry and Technology, Prague, Czech Republic
| | - Evžen Růžička
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Karel Šonka
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Petr Dušek
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jan Rusz
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Department of Neurology and ARTORG Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
26
|
Angerbauer R, Stefani A, Zitser J, Ibrahim A, Anselmi V, Süzgün MA, Egger K, Brandauer E, Högl B, Cesari M. Temporal progression of sleep electroencephalography features in isolated rapid eye movement sleep behaviour disorder. J Sleep Res 2024:e14351. [PMID: 39322419 DOI: 10.1111/jsr.14351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/16/2024] [Accepted: 09/04/2024] [Indexed: 09/27/2024]
Abstract
Previous studies indicated that patients with isolated rapid eye movement (REM) sleep behaviour disorder (iRBD) exhibit alterations in spectral electroencephalographic (EEG), spindle, and slow-wave features. As it is currently unknown how these EEG features evolve over time, this study aimed to evaluate their temporal progression in patients with iRBD in comparison to controls. We included 23 patients with iRBD and 23 controls. Two polysomnographies (baseline and follow-up) were recorded with a mean (standard deviation) interval of 4.0 (2.5) years and were automatically analysed for sleep stages, spectral bandpower, spindles, and slow waves. We used linear models to evaluate differences at each time point, and linear mixed-effects models to analyse differences in temporal progression between the groups. At baseline, patients with iRBD presented EEG slowing both in REM (expressed as significantly reduced α-bandpower and increased δ-bandpower in frontal channels) and in non-REM (NREM) sleep (significantly increased slow-to-fast ratio in central channels). These differences vanished at follow-up. In both REM and NREM sleep, γ-bandpower was increased at follow-up in patients with iRBD, resulting in significantly different temporal progression between groups (in occipital channels during REM sleep and frontal channels during NREM sleep). Relative power of sleep spindles was significantly higher at baseline in patients with iRBD in frontal channels, but we observed a significant reduction over time in central channels. Finally, slow waves were significantly shorter in patients with iRBD at both time-points. Our results underscore the need of considering longitudinal data when analysing sleep EEG features in patients with iRBD. The observed temporal changes as markers of progression of neurodegeneration require further investigations.
Collapse
Affiliation(s)
- Raphael Angerbauer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ambra Stefani
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jennifer Zitser
- Sleep Center and Movement Disorders Unit, Neurology Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Abubaker Ibrahim
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Victoria Anselmi
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Merve Aktan Süzgün
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kristin Egger
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Elisabeth Brandauer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Birgit Högl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Matteo Cesari
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
27
|
Li FJ, Zhang ZX, Li YDY, Li JY, Liu YN, Liu XJ, Zhang RY, Liu X, Zhang W, Xu CY, Cui GY. High bioavailable testosterone levels increase the incidence of isolated REM sleep behavior disorder: Results from multivariable and network Mendelian randomization analysis. Sleep Med 2024; 121:102-110. [PMID: 38959716 DOI: 10.1016/j.sleep.2024.06.024] [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: 02/15/2024] [Revised: 06/09/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
OBJECTIVES To explore the causal relationships between sex hormone levels and incidence of isolated REM sleep behavior disorder (iRBD). METHODS In our study, we utilized Genome-Wide Association Studies (GWAS) data for iRBD, including 9447 samples with 1061 cases of iRBD provided by the International RBD Study Group. Initially, we conducted a two-sample univariate MR analysis to explore the impact of sex hormone-related indicators on iRBD. This was followed by the application of multivariable MR methods to adjust for other hormone levels and potential confounders. Finally, we undertook a network MR analysis, employing brain structure Magnetic Resonance Imaging (MRI) characteristics as potential mediators, to examine whether sex hormones could indirectly influence the incidence of iRBD by affecting brain structure. RESULTS Bioavailable testosterone (BioT) is an independent risk factor for iRBD (Odds Ratio [95 % Confidence Interval] = 2.437 [1.308, 4.539], P = 0.005, corrected-P = 0.020), a finding that remained consistent even after adjusting for other sex hormone levels and potential confounders. Additionally, BioT appears to indirectly increase the risk of iRBD by reducing axial diffusivity and increasing the orientation dispersion index in the left cingulum and cingulate gyrus. CONCLUSIONS Our research reveals that elevated levels of BioT contribute to the development of iRBD. However, the specific impact of BioT on different sexes remains unclear. Furthermore, high BioT may indirectly lead to iRBD by impairing normal pathways in the left cingulum and cingulate gyrus and fostering abnormal pathway formation.
Collapse
Affiliation(s)
- Fu-Jia Li
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, People's Republic of China
| | - Zi-Xuan Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, People's Republic of China
| | - Yang-Dan-Yu Li
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, People's Republic of China
| | - Jin-Yu Li
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, People's Republic of China
| | - Yu-Ning Liu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, People's Republic of China
| | - Xuan-Jing Liu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, People's Republic of China
| | - Ru-Yu Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, People's Republic of China
| | - Xu Liu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, People's Republic of China
| | - Wei Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, People's Republic of China
| | - Chuan-Ying Xu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, People's Republic of China
| | - Gui-Yun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, People's Republic of China.
| |
Collapse
|
28
|
Zheng Y, Yu Z, Cai H, Kou W, Yang C, Li S, Zhang N, Feng T. Detection of α-Synuclein in Oral Mucosa by Seed Amplification Assay in Synucleinopathies and Isolated REM Sleep Behavior Disorder. Mov Disord 2024; 39:1300-1309. [PMID: 38715177 DOI: 10.1002/mds.29828] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/01/2024] [Accepted: 04/18/2024] [Indexed: 08/23/2024] Open
Abstract
OBJECTIVE Evidence of abnormal α-synuclein (α-Syn) deposition in the brain is required for definitive diagnosis of synucleinopathies, which remains challenging. The seed amplification assay (SAA) is an innovative technique that can detect the seeding activity of misfolded α-Syn, enabling the amplification and detection of minute quantities of pathogenic α-Syn aggregates. This study aimed to evaluate oral mucosa α-Syn SAA as possible diagnostic and prodromal biomarkers for synucleinopathies. METHODS A total of 107 Parkinson's disease (PD) patients, 99 multiple system atrophy (MSA) patients, 33 patients with isolated rapid eye movement sleep behavior disorder (iRBD) and 103 healthy controls (HC) were included. The SAA was applied to detect the seeding activity of α-Syn from oral mucosa. A combination of morphological, biochemical, and biophysical methods was also used to analyze the fibrils generated from the oral mucosa α-Syn SAA. RESULTS Structured illumination microscopy images revealed the increased α-Syn species in oral mucosa of PD, MSA, and iRBD patients than in HCs. Oral mucosa α-Syn SAA distinguished patients with PD from HC with 67.3% sensitivity and 90.3% specificity. Oral mucosa was α-Syn SAA positive in 53.5% MSA patients and 63.6% iRBD patients. Furthermore, the α-Syn fibrils generated from MSA demonstrated greater resistance to proteinase K digestion and exhibited stronger cytotoxicity compared to those from PD patients. CONCLUSION Oral mucosa α-Syn seeding activity may serve as novel non-invasive diagnostic and prodromal biomarkers for synucleinopathies. The α-Syn aggregates amplified from the oral mucosa of PD and MSA exhibited distinct biochemical and biophysical properties. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Yuanchu Zheng
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhenwei Yu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Huihui Cai
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenyi Kou
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chen Yang
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Siming Li
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ning Zhang
- Department of Neuropsychiatry and Behavioral Neurology and Clinical Psychology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Feng
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
29
|
Li FJ, Zhang RY, Li JY, Liu YN, Zhang ZX, Du L, Li YDY, Liu X, Zhang W, Cui GY, Xu CY. Pain, obesity, adenosine salvage disruption, and smoking behavior mediate the effect of gut microbiota on sleep disorders: results from network Mendelian randomization and 16S rDNA sequencing. Front Microbiol 2024; 15:1413218. [PMID: 39144232 PMCID: PMC11322093 DOI: 10.3389/fmicb.2024.1413218] [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: 05/02/2024] [Accepted: 07/12/2024] [Indexed: 08/16/2024] Open
Abstract
Objectives The objective of this study is to investigate the indirect causalities between gut microbiota and sleep disorders. Methods In stage 1, we utilized 196 gut microbiota as the exposure factor and conducted a two-sample univariable Mendelian randomization (MR) analysis on five sleep disorders: insomnia, excessive daytime sleepiness (EDS), sleep-wake rhythm disorders (SWRD), obstructive sleep apnea (OSA), and isolated REM sleep behavior disorder (iRBD). In stage 2, we validated the MR findings by comparing fecal microbiota abundance between patients and healthy controls through 16S rDNA sequencing. In stage 3, we explored the indirect pathways by which the microbiota affects sleep, using 205 gut microbiota metabolic pathways and 9 common risk factors for sleep disorders as candidate mediators in a network MR analysis. Results In stage 1, the univariable MR analysis identified 14 microbiota potentially influencing five different sleep disorders. In stage 2, the results from our observational study validated four of these associations. In stage 3, the network MR analysis revealed that the Negativicutes class and Selenomonadales order might worsen insomnia by increasing pain [mediation: 12.43% (95% CI: 0.47, 24.39%)]. Oxalobacter could raise EDS by disrupting adenosine reuptake [25.39% (1.84, 48.95%)]. Allisonella may elevate OSA risk via obesity promotion [36.88% (17.23, 56.54%)], while the Eubacterium xylanophilum group may lower OSA risk by decreasing smoking behavior [7.70% (0.66, 14.74%)]. Conclusion Triangulation of evidence from the MR and observational study revealed indirect causal relationships between the microbiota and sleep disorders, offering fresh perspectives on how gut microbiota modulate sleep.
Collapse
Affiliation(s)
- Fu-Jia Li
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ru-Yu Zhang
- Department of Pulmonary and Critical Care Medicine, First People’s Hospital of Zigong, Zigong, Sichuan, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jin-Yu Li
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yu-Ning Liu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zi-Xuan Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Li Du
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yang-Dan-Yu Li
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xu Liu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wei Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Gui-Yun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chuan-Ying Xu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
30
|
Zheng Y, Li Y, Cai H, Kou W, Yang C, Li S, Wang J, Zhang N, Feng T. Alterations of Peripheral Lymphocyte Subsets in Isolated Rapid Eye Movement Sleep Behavior Disorder. Mov Disord 2024; 39:1179-1189. [PMID: 38529776 DOI: 10.1002/mds.29798] [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: 01/04/2024] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Adaptive immune dysfunction may play a crucial role in Parkinson's disease (PD) development. Isolated rapid eye movement sleep behavior disorder (iRBD) represents the prodromal stage of synucleinopathies, including PD. Elucidating the peripheral adaptive immune system is crucial in iRBD, but current knowledge remains limited. OBJECTIVE This study aimed to characterize peripheral lymphocyte profiles in iRBD patients compared with healthy control subjects (HCs). METHODS This cross-sectional study recruited polysomnography-confirmed iRBD patients and age- and sex-matched HCs. Venous blood was collected from each participant. Flow cytometry was used to evaluate surface markers and intracellular cytokine production in peripheral blood mononuclear cells. RESULTS Forty-four iRBD patients and 36 HCs were included. Compared with HCs, patients with iRBD exhibited significant decreases in absolute counts of total lymphocytes and CD3+ T cells. In terms of T cell subsets, iRBD patients showed higher frequencies and counts of proinflammatory T helper 1 cells and INF-γ+ CD8+ T cells, along with lower frequencies and counts of anti-inflammatory T helper 2 cells. A significant increase in the frequency of central memory T cells in CD8+ T cells was also observed in iRBD. Regarding B cells, iRBD patients demonstrated reduced frequencies and counts of double-negative memory B cells compared with control subjects. CONCLUSIONS This study demonstrated alterations in the peripheral adaptive immune system in iRBD, specifically in CD4+ and INF-γ+ CD8+ T cell subsets. An overall shift toward a proinflammatory state of adaptive immunity was already evident in iRBD. These observations might provide insights into the optimal timing for initiating immune interventions in PD. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Yuanchu Zheng
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yatong Li
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Huihui Cai
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenyi Kou
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chen Yang
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Siming Li
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiawei Wang
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ning Zhang
- Department of Neuropsychiatry and Behavioral Neurology and Clinical Psychology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Feng
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
31
|
Marecek S, Krajca T, Krupicka R, Sojka P, Nepozitek J, Varga Z, Mala C, Keller J, Waugh JL, Zogala D, Trnka J, Sonka K, Ruzicka E, Dusek P. Analysis of striatal connectivity corresponding to striosomes and matrix in de novo Parkinson's disease and isolated REM behavior disorder. NPJ Parkinsons Dis 2024; 10:124. [PMID: 38918417 PMCID: PMC11199557 DOI: 10.1038/s41531-024-00736-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Striosomes and matrix are two compartments that comprise the striatum, each having its own distinct immunohistochemical properties, function, and connectivity. It is currently not clear whether prodromal or early manifest Parkinson's disease (PD) is associated with any striatal matrix or striosomal abnormality. Recently, a method of striatal parcellation using probabilistic tractography has been described and validated, using the distinct connectivity of these two compartments to identify voxels with striosome- and matrix-like connectivity. The goal of this study was to use this approach in tandem with DAT-SPECT, a method used to quantify the level of nigrostriatal denervation, to analyze the striatum in populations of de novo diagnosed, treatment-naïve patients with PD, isolated REM behavioral disorder (iRBD) patients, and healthy controls. We discovered a shift in striatal connectivity, which showed correlation with nigrostriatal denervation. Patients with PD exhibited a significantly higher matrix-like volume and associated connectivity than healthy controls and higher matrix-associated connectivity than iRBD patients. In contrast, the side with less pronounced nigrostriatal denervation in PD and iRBD patients showed a decrease in striosome-like volume and associated connectivity indices. These findings could point to a compensatory neuroplastic mechanism in the context of nigrostriatal denervation and open a new avenue in the investigation of the pathophysiology of Parkinson's disease.
Collapse
Affiliation(s)
- S Marecek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
| | - T Krajca
- Czech Technical University in Prague, Faculty of Biomedical Engineering, Kladno, Czech Republic
| | - R Krupicka
- Czech Technical University in Prague, Faculty of Biomedical Engineering, Kladno, Czech Republic
| | - P Sojka
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - J Nepozitek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Z Varga
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - C Mala
- Czech Technical University in Prague, Faculty of Biomedical Engineering, Kladno, Czech Republic
| | - J Keller
- Department of Radiodiagnostics, Na Homolce Hospital, Prague, Czech Republic
| | - J L Waugh
- Division of Pediatric Neurology, Department of Pediatrics, University of Texas Southwestern, Dallas, TX, USA
| | - D Zogala
- Institute of Nuclear Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - J Trnka
- Institute of Nuclear Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - K Sonka
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - E Ruzicka
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - P Dusek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
32
|
Avila A, Zhang SL. A circadian clock regulates the blood-brain barrier across phylogeny. VITAMINS AND HORMONES 2024; 126:241-287. [PMID: 39029975 DOI: 10.1016/bs.vh.2024.04.004] [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] [Indexed: 07/21/2024]
Abstract
As the central regulatory system of an organism, the brain is responsible for overseeing a wide variety of physiological processes essential for an organism's survival. To maintain the environment necessary for neurons to function, the brain requires highly selective uptake and elimination of specific molecules through the blood-brain barrier (BBB). As an organism's activities vary throughout the day, how does the BBB adapt to meet the changing needs of the brain? A mechanism is through temporal regulation of BBB permeability via its circadian clock, which will be the focal point of this chapter. To comprehend the circadian clock's role within the BBB, we will first examine the anatomy of the BBB and the transport mechanisms enabling it to fulfill its role as a restrictive barrier. Next, we will define the circadian clock, and the discussion will encompass an introduction to circadian rhythms, the Transcription-Translation Feedback Loop (TTFL) as the mechanistic basis of circadian timekeeping, and the organization of tissue clocks found in organisms. Then, we will cover the role of the circadian rhythms in regulating the cellular mechanisms and functions of the BBB. We discuss the implications of this regulation in influencing sleep behavior, the progression of neurodegenerative diseases, and finally drug delivery for treatment of neurological diseases.
Collapse
Affiliation(s)
- Ashley Avila
- Cell Biology Department, Emory University, Atlanta, GA, United States
| | - Shirley L Zhang
- Cell Biology Department, Emory University, Atlanta, GA, United States.
| |
Collapse
|
33
|
Bramich S, Noyce AJ, King AE, Naismith SL, Kuruvilla MV, Lewis SJG, Roccati E, Bindoff AD, Barnham KJ, Beauchamp LC, Vickers JC, Pérez-Carbonell L, Alty J. Isolated rapid eye movement sleep behaviour disorder (iRBD) in the Island Study Linking Ageing and Neurodegenerative Disease (ISLAND) Sleep Study: protocol and baseline characteristics. J Sleep Res 2024; 33:e14109. [PMID: 38014898 DOI: 10.1111/jsr.14109] [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: 07/04/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/29/2023]
Abstract
Isolated rapid eye movement (REM) sleep behaviour disorder (iRBD) is a sleep disorder that is characterised by dream enactment episodes during REM sleep. It is the strongest known predictor of α-synuclein-related neurodegenerative disease (αNDD), such that >80% of people with iRBD will eventually develop Parkinson's disease, dementia with Lewy bodies, or multiple system atrophy in later life. More research is needed to understand the trajectory of phenoconversion to each αNDD. Only five 'gold standard' prevalence studies of iRBD in older adults have been undertaken previously, with estimates ranging from 0.74% to 2.01%. The diagnostic recommendations for video-polysomnography (vPSG) to confirm iRBD makes prevalence studies challenging, as vPSG is often unavailable to large cohorts. In Australia, there have been no iRBD prevalence studies, and little is known about the cognitive and motor profiles of Australian people with iRBD. The Island Study Linking Ageing and Neurodegenerative Disease (ISLAND) Sleep Study will investigate the prevalence of iRBD in Tasmania, an island state of Australia, using validated questionnaires and home-based vPSG. It will also explore several cognitive, motor, olfactory, autonomic, visual, tactile, and sleep profiles in people with iRBD to better understand which characteristics influence the progression of iRBD to αNDD. This paper details the ISLAND Sleep Study protocol and presents preliminary baseline results.
Collapse
Affiliation(s)
- Samantha Bramich
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Australia
| | - Alastair J Noyce
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University, London, UK
| | - Anna E King
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Australia
| | - Sharon L Naismith
- Brain and Mind Centre, The University of Sydney, Camperdown, Australia
| | | | - Simon J G Lewis
- Brain and Mind Centre, The University of Sydney, Camperdown, Australia
| | - Eddy Roccati
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Australia
| | - Aidan D Bindoff
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Australia
| | - Kevin J Barnham
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Leah C Beauchamp
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - James C Vickers
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Australia
| | - Laura Pérez-Carbonell
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University, London, UK
- Sleep Disorders Centre, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Jane Alty
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Australia
- School of Medicine, University of Tasmania, Hobart, Australia
- Department of Neurology, Royal Hobart Hospital, Hobart, Australia
| |
Collapse
|
34
|
Park G, Jo H, Chai Y, Park HR, Lee H, Joo EY, Kim H. Static and dynamic brain morphological changes in isolated REM sleep behavior disorder compared to normal aging. Front Neurosci 2024; 18:1365307. [PMID: 38751861 PMCID: PMC11094219 DOI: 10.3389/fnins.2024.1365307] [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: 01/04/2024] [Accepted: 04/16/2024] [Indexed: 05/18/2024] Open
Abstract
Objective/background To assess whether cerebral structural alterations in isolated rapid eye movement sleep behavior disorder (iRBD) are progressive and differ from those of normal aging and whether they are related to clinical symptoms. Patients/methods In a longitudinal study of 18 patients with iRBD (age, 66.1 ± 5.7 years; 13 males; follow-up, 1.6 ± 0.6 years) and 24 age-matched healthy controls (age, 67.0 ± 4.9 years; 12 males; follow-up, 2.0 ± 0.9 years), all participants underwent multiple extensive clinical examinations, neuropsychological tests, and magnetic resonance imaging at baseline and follow-up. Surface-based cortical reconstruction and automated subcortical structural segmentation were performed on T1-weighted images. We used mixed-effects models to examine the differences between the groups and the differences in anatomical changes over time. Results None of the patients with iRBD demonstrated phenoconversion during the follow-up. Patients with iRBD had thinner cortices in the frontal, occipital, and temporal regions, and more caudate atrophy, compared to that in controls. In similar regions, group-by-age interaction analysis revealed that patients with iRBD demonstrated significantly slower decreases in cortical thickness and caudate volume with aging than that observed in controls. Patients with iRBD had lower scores on the Korean version of the Mini-Mental Status Examination (p = 0.037) and frontal and executive functions (p = 0.049) at baseline than those in controls; however, no significant group-by-age interaction was identified. Conclusion Patients with iRBD show brain atrophy in the regions that are overlapped with the areas that have been documented to be affected in early stages of Parkinson's disease. Such atrophy in iRBD may not be progressive but may be slower than that in normal aging. Cognitive impairment in iRBD is not progressive.
Collapse
Affiliation(s)
- Gilsoon Park
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| | - Hyunjin Jo
- Department of Neurology, Neuroscience Center, Samsung Medical Center, Samsung Biomedical Research Institute, School of Medicine, Sungkyunkwan University, Seoul, Republic of Korea
- Medical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Yaqiong Chai
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| | - Hea Ree Park
- Department of Neurology, Inje University College of Medicine, Ilsan Paik Hospital, Goyang, Republic of Korea
| | - Hanul Lee
- Department of Neurology, Neuroscience Center, Samsung Medical Center, Samsung Biomedical Research Institute, School of Medicine, Sungkyunkwan University, Seoul, Republic of Korea
| | - Eun Yeon Joo
- Department of Neurology, Neuroscience Center, Samsung Medical Center, Samsung Biomedical Research Institute, School of Medicine, Sungkyunkwan University, Seoul, Republic of Korea
| | - Hosung Kim
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
35
|
Wang R, Lian T, He M, Guo P, Yu S, Zuo L, Hu Y, Zhang W. Clinical features and neurobiochemical mechanisms of olfactory dysfunction in patients with Parkinson disease. J Neurol 2024; 271:1959-1972. [PMID: 38151574 DOI: 10.1007/s00415-023-12122-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 12/29/2023]
Abstract
This study aimed to investigate clinical features, influencing factors and neurobiochemical mechanisms of olfactory dysfunction (OD) in Parkinson disease (PD). Total 39 patients were divided into the PD with OD (PD-OD) and PD with no OD (PD-nOD) groups according to overall olfactory function, including threshold, discrimination and identification, assessed by Sniffin' Sticks test. Motor function and non-motor symptoms were rated by multiple scales. Dopamine, acetylcholine, norepinephrine and 5-hydroxytryptamine levels in cerebrospinal fluid (CSF) were measured. We found that the PD-OD group showed significantly lower score of Montreal Cognitive Assessment Scale, higher scores of rapid eye movement sleep behavior disorder (RBD) Screening Questionnaire and Epworth Sleepiness Scale than the PD-nOD group (p < 0.05). RBD Screening Questionnaire score was independently associated with the scores of overall olfactory function and discrimination (p < 0.05). Dopamine and acetylcholine levels in CSF from the PD-OD group was significantly lower than that from the PD-nOD group (p < 0.05). Dopamine and acetylcholine levels in CSF were significantly and positively correlated with the scores of overall olfactory function, threshold, discrimination and identification in PD patients (p < 0.05). RBD Screening Questionnaire score was significantly and negatively correlated with acetylcholine level in CSF in PD patients with poor olfactory detection (p < 0.05). This investigation reveals that PD-OD is associated with cognitive impairment, probable RBD and excessive daytime sleepiness. PD-OD is correlated with the decreased levels of dopamine and acetylcholine in CSF. RBD is an independent influencing factor of overall olfactory function and discrimination, and the decreased acetylcholine level in CSF may be the common neurobiochemical basis of RBD and OD in PD patients.
Collapse
Grants
- 2016YFC1306000 National Key Research and Development Program of China
- 2016YFC1306300 National Key Research and Development Program of China
- 81970992 National Natural Science Foundation of China
- 81571229 National Natural Science Foundation of China
- 81071015 National Natural Science Foundation of China
- 30770745 National Natural Science Foundation of China
- 82201639 National Natural Science Foundation of China
- 2022-2-2048 Capital's Funds for Health Improvement and Research (CFH)
- kz201610025030 Key Technology R&D Program of Beijing Municipal Education Commission
- 4161004 Key Project of Natural Science Foundation of Beijing, China
- 7082032 Natural Science Foundation of Beijing, China
- JJ2018-48 Project of Scientific and Technological Development of Traditional Chinese Medicine in Beijing
- Z121107001012161 Capital Clinical Characteristic Application Research
- 2009-3-26 High Level Technical Personnel Training Project of Beijing Health System, China
- BIBD-PXM2013_014226_07_000084 Project of Beijing Institute for Brain Disorders
- 20071D0300400076 Excellent Personnel Training Project of Beijing, China
- IDHT20140514 Project of Construction of Innovative Teams and Teacher Career Development for Universities and Colleges Under Beijing Municipality
- JING-15-2 Beijing Healthcare Research Project, China
- 2015-JL-PT-X04 Basic-Clinical Research Cooperation Funding of Capital Medical University, China
- 10JL49 Basic-Clinical Research Cooperation Funding of Capital Medical University, China
- 14JL15 Basic-Clinical Research Cooperation Funding of Capital Medical University, China
- PYZ2018077 Natural Science Foundation of Capital Medical University, Beijing, China
- 2019-028 Science and Technology Development Fund of Beijing Rehabilitation Hospital, Capital Medical University
Collapse
Affiliation(s)
- Ruidan Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Tenghong Lian
- Center for Cognitive Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Mingyue He
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Peng Guo
- Center for Cognitive Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Shuyang Yu
- Center for Cognitive Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Lijun Zuo
- Center for Cognitive Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Yang Hu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Wei Zhang
- Center for Cognitive Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China.
- Beijing Key Laboratory on Parkinson Disease, Beijing Institute for Brain Disorders, Beijing, 10053, China.
| |
Collapse
|
36
|
Tassan Mazzocco M, Serra M, Maspero M, Coliva A, Presotto L, Casu MA, Morelli M, Moresco RM, Belloli S, Pinna A. Positive relation between dopamine neuron degeneration and metabolic connectivity disruption in the MPTP plus probenecid mouse model of Parkinson's disease. Exp Neurol 2024; 374:114704. [PMID: 38281587 DOI: 10.1016/j.expneurol.2024.114704] [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: 10/11/2023] [Revised: 01/15/2024] [Accepted: 01/25/2024] [Indexed: 01/30/2024]
Abstract
The clinical manifestation of Parkinson's disease (PD) appears when neurodegeneration is already advanced, compromising the efficacy of disease-modifying treatment approaches. Biomarkers to identify the early stages of PD are therefore of paramount importance for the advancement of the therapy of PD. In the present study, by using a mouse model of PD obtained by subchronic treatment with the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and the clearance inhibitor probenecid (MPTPp), we identified prodromal markers of PD by combining in vivo positron emission tomography (PET) imaging and ex vivo immunohistochemistry. Longitudinal PET imaging of the dopamine transporter (DAT) by [18F]-N-(3-fluoropropyl)-2β-carboxymethoxy-3β-(4-iodophenyl) nortropane ([18F]-FP-CIT), and brain glucose metabolism by 2-deoxy-2-[18F]-fluoroglucose ([18F]-FDG) were performed before MPTPp treatment and after 1, 3, and 10 MPTPp administrations, in order to assess relation between dopamine neuron integrity and brain connectivity. The results show that in vivo [18F]-FP-CIT in the dorsal striatum was not modified after the first administration of MPTPp, tended to decrease after 3 administrations, and significantly decreased after 10 MPTPp administrations. Post-mortem immunohistochemical analyses of DAT and tyrosine hydroxylase (TH) in the striatum showed a positive correlation with [18F]-FP-CIT, confirming the validity of repeated MPTPp-treated mice as a model that can reproduce the progressive pathological changes in the early phases of PD. Analysis of [18F]-FDG uptake in several brain areas connected to the striatum showed that metabolic connectivity was progressively disrupted, starting from the first MPTPp administration, and that significant connections between cortical and subcortical regions were lost after 10 MPTPp administrations, suggesting an association between dopamine neuron degeneration and connectivity disruption in this PD model. The results of this study provide a relevant model, where new drugs that can alleviate neurodegeneration in PD could be evaluated preclinically.
Collapse
Affiliation(s)
- Margherita Tassan Mazzocco
- PhD Program in Neuroscience, Medicine and Surgery Department, University of Milano-Bicocca, Monza, Italy; Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Marcello Serra
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Marco Maspero
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; National Research Council of Italy, Institute of Molecular Bioimaging and Physiology, UOS of Segrate, Italy
| | - Angela Coliva
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Luca Presotto
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; Department of Physics "G. Occhialini", University of Milano - Bicocca, Milan, Italy
| | - Maria Antonietta Casu
- National Research Council of Italy, Institute of Translational Pharmacology, UOS of Cagliari, Scientific and Technological Park of Sardinia POLARIS, Pula, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy; National Research Council of Italy, Neuroscience Institute, UOS of Cagliari, Italy
| | - Rosa Maria Moresco
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; National Research Council of Italy, Institute of Molecular Bioimaging and Physiology, UOS of Segrate, Italy; School of Medicine and Surgery, University of Milano - Bicocca, Monza, Italy.
| | - Sara Belloli
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy; National Research Council of Italy, Institute of Molecular Bioimaging and Physiology, UOS of Segrate, Italy
| | - Annalisa Pinna
- National Research Council of Italy, Neuroscience Institute, UOS of Cagliari, Italy
| |
Collapse
|
37
|
Ojo OO, Bandres-Ciga S, Makarious MB, Crea PW, Hernandez DG, Houlden H, Rizig M, Singleton AB, Noyce AJ, Nalls MA, Blauwendraat C, Okubadejo NU. GBA1 rs3115534 Is Associated with REM Sleep Behavior Disorder in Parkinson's Disease in Nigerians. Mov Disord 2024; 39:728-733. [PMID: 38390630 DOI: 10.1002/mds.29753] [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: 11/23/2023] [Revised: 01/25/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Rapid eye movement (REM) sleep behavior disorder (RBD) is an early feature of Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Damaging coding variants in Glucocerebrosidase (GBA1) are a genetic risk factor for RBD. Recently, a population-specific non-coding risk variant (rs3115534) was found to be associated with PD risk and earlier onset in individuals of African ancestry. OBJECTIVES We aimed to investigate whether the GBA1 rs3115534 PD risk variant is associated with RBD in persons with PD. METHODS We studied 709 persons with PD and 776 neurologically healthy controls from Nigeria. All DNA samples were genotyped and imputed, and the GBA1 rs3115534 risk variant was extracted. The RBD screening questionnaire (RBDSQ) was used to assess symptoms of possible RBD. RESULTS RBD was present in 200 PD (28.2%) and 51 (6.6%) controls. We identified that the non-coding GBA1 rs3115534 risk variant is associated with possible RBD in individuals of Nigerian origin (β, 0.3640; standard error [SE], 0.103, P = 4.093e-04), as well as in all samples after adjusting for PD status (β, 0.2542; SE, 0.108; P = 0.019) suggesting that although non-coding, this variant may have the same downstream consequences as GBA1 coding variants. CONCLUSIONS Our results indicate that the non-coding GBA1 rs3115534 risk variant is associated with an increasing number of RBD symptoms in persons with PD of Nigerian origin. Further research is needed to assess if this variant is also associated with polysomnography-defined RBD and with RBD symptoms in DLB. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Oluwadamilola Omolara Ojo
- College of Medicine, University of Lagos, Idi-Araba, Lagos State, Nigeria
- Lagos University Teaching Hospital, Idi-Araba, Lagos State, Nigeria
| | - Sara Bandres-Ciga
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Mary B Makarious
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- UCL Movement Disorders Centre, University College London, London, United Kingdom
| | - Peter Wild Crea
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- UCL Movement Disorders Centre, University College London, London, United Kingdom
| | - Dena G Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Mie Rizig
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Andrew B Singleton
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Alastair J Noyce
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University London, London, United Kingdom
| | - Mike A Nalls
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
- DataTecnica LLC, Washington, District of Columbia, USA
| | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Njideka Ulunma Okubadejo
- College of Medicine, University of Lagos, Idi-Araba, Lagos State, Nigeria
- Lagos University Teaching Hospital, Idi-Araba, Lagos State, Nigeria
| |
Collapse
|
38
|
Gabb VG, Blackman J, Morrison HD, Biswas B, Li H, Turner N, Russell GM, Greenwood R, Jolly A, Trender W, Hampshire A, Whone A, Coulthard E. Remote Evaluation of Sleep and Circadian Rhythms in Older Adults With Mild Cognitive Impairment and Dementia: Protocol for a Feasibility and Acceptability Mixed Methods Study. JMIR Res Protoc 2024; 13:e52652. [PMID: 38517469 PMCID: PMC10998181 DOI: 10.2196/52652] [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: 09/19/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Sleep disturbances are a potentially modifiable risk factor for neurodegenerative dementia secondary to Alzheimer disease (AD) and Lewy body disease (LBD). Therefore, we need to identify the best methods to study sleep in this population. OBJECTIVE This study will assess the feasibility and acceptability of various wearable devices, smart devices, and remote study tasks in sleep and cognition research for people with AD and LBD. METHODS We will deliver a feasibility and acceptability study alongside a prospective observational cohort study assessing sleep and cognition longitudinally in the home environment. Adults aged older than 50 years who were diagnosed with mild to moderate dementia or mild cognitive impairment (MCI) due to probable AD or LBD and age-matched controls will be eligible. Exclusion criteria include lack of capacity to consent to research, other causes of MCI or dementia, and clinically significant sleep disorders. Participants will complete a cognitive assessment and questionnaires with a researcher and receive training and instructions for at-home study tasks across 8 weeks. At-home study tasks include remote sleep assessments using wearable devices (electroencephalography headband and actigraphy watch), app-based sleep diaries, online cognitive assessments, and saliva samples for melatonin- and cortisol-derived circadian markers. Feasibility outcomes will be assessed relating to recruitment and retention, data completeness, data quality, and support required. Feedback on acceptability and usability will be collected throughout the study period and end-of-study interviews will be analyzed using thematic analysis. RESULTS Recruitment started in February 2022. Data collection is ongoing, with final data expected in February 2024 and data analysis and publication of findings scheduled for the summer of 2024. CONCLUSIONS This study will allow us to assess if remote testing using smart devices and wearable technology is a viable alternative to traditional sleep measurements, such as polysomnography and questionnaires, in older adults with and without MCI or dementia due to AD or LBD. Understanding participant experience and the barriers and facilitators to technology use for research purposes and remote research in this population will assist with the development of, recruitment to, and retention within future research projects studying sleep and cognition outside of the clinic or laboratory. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/52652.
Collapse
Affiliation(s)
- Victoria Grace Gabb
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Neurology Department, Bristol Brain Centre, North Bristol NHS Trust, Bristol, United Kingdom
| | - Jonathan Blackman
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Neurology Department, Bristol Brain Centre, North Bristol NHS Trust, Bristol, United Kingdom
| | - Hamish Duncan Morrison
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Neurology Department, Bristol Brain Centre, North Bristol NHS Trust, Bristol, United Kingdom
| | - Bijetri Biswas
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Haoxuan Li
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Neurology Department, Bristol Brain Centre, North Bristol NHS Trust, Bristol, United Kingdom
- King's College Hospital, King's College Hospital NHS Foundation Trust, London, United Kingdom
- Bristol Royal Infirmary, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Nicholas Turner
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | | | - Rosemary Greenwood
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Research & Innovation, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Amy Jolly
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
- UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - William Trender
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Adam Hampshire
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Alan Whone
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Neurology Department, Bristol Brain Centre, North Bristol NHS Trust, Bristol, United Kingdom
| | - Elizabeth Coulthard
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Neurology Department, Bristol Brain Centre, North Bristol NHS Trust, Bristol, United Kingdom
| |
Collapse
|
39
|
Šubert M, Novotný M, Tykalová T, Hlavnička J, Dušek P, Růžička E, Škrabal D, Pelletier A, Postuma RB, Montplaisir J, Gagnon JF, Galbiati A, Ferini-Strambi L, Marelli S, St Louis EK, Timm PC, Teigen LN, Janzen A, Oertel W, Heim B, Holzknecht E, Stefani A, Högl B, Dauvilliers Y, Evangelista E, Šonka K, Rusz J. Spoken Language Alterations can Predict Phenoconversion in Isolated Rapid Eye Movement Sleep Behavior Disorder: A Multicenter Study. Ann Neurol 2024; 95:530-543. [PMID: 37997483 DOI: 10.1002/ana.26835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 11/25/2023]
Abstract
OBJECTIVE This study assessed the relationship between speech and language impairment and outcome in a multicenter cohort of isolated/idiopathic rapid eye movement (REM) sleep behavior disorder (iRBD). METHODS Patients with iRBD from 7 centers speaking Czech, English, German, French, and Italian languages underwent a detailed speech assessment at baseline. Story-tale narratives were transcribed and linguistically annotated using fully automated methods based on automatic speech recognition and natural language processing algorithms, leading to the 3 distinctive linguistic and 2 acoustic patterns of language deterioration and associated composite indexes of their overall severity. Patients were then prospectively followed and received assessments for parkinsonism or dementia during follow-up. The Cox proportional hazard was performed to evaluate the predictive value of language patterns for phenoconversion over a follow-up period of 5 years. RESULTS Of 180 patients free of parkinsonism or dementia, 156 provided follow-up information. After a mean follow-up of 2.7 years, 42 (26.9%) patients developed neurodegenerative disease. Patients with higher severity of linguistic abnormalities (hazard ratio [HR = 2.35]) and acoustic abnormalities (HR = 1.92) were more likely to develop a defined neurodegenerative disease, with converters having lower content richness (HR = 1.74), slower articulation rate (HR = 1.58), and prolonged pauses (HR = 1.46). Dementia-first (n = 16) and parkinsonism-first with mild cognitive impairment (n = 9) converters had higher severity of linguistic abnormalities than parkinsonism-first with normal cognition converters (n = 17). INTERPRETATION Automated language analysis might provide a predictor of phenoconversion from iRBD into synucleinopathy subtypes with cognitive impairment, and thus can be used to stratify patients for neuroprotective trials. ANN NEUROL 2024;95:530-543.
Collapse
Affiliation(s)
- Martin Šubert
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Michal Novotný
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Tereza Tykalová
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Jan Hlavnička
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Petr Dušek
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Evžen Růžička
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Dominik Škrabal
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Amelie Pelletier
- Department of Neurology, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec, Canada
- Center for Advanced Research in Sleep Medicine, CIUSSS-NÎM - Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada
| | - Ronald B Postuma
- Department of Neurology, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec, Canada
- Center for Advanced Research in Sleep Medicine, CIUSSS-NÎM - Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada
| | - Jacques Montplaisir
- Center for Advanced Research in Sleep Medicine, CIUSSS-NÎM - Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada
| | - Jean-François Gagnon
- Center for Advanced Research in Sleep Medicine, CIUSSS-NÎM - Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada
| | - Andrea Galbiati
- Sleep Disorders Center, Division of Neuroscience, Ospedale San Raffaele, Università Vita-Salute, Milan, Italy
- Department of Psychology, "Vita-Salute" San Raffaele University, Milan, Italy
| | - Luigi Ferini-Strambi
- Sleep Disorders Center, Division of Neuroscience, Ospedale San Raffaele, Università Vita-Salute, Milan, Italy
- Department of Psychology, "Vita-Salute" San Raffaele University, Milan, Italy
| | - Sara Marelli
- Sleep Disorders Center, Division of Neuroscience, Ospedale San Raffaele, Università Vita-Salute, Milan, Italy
| | - Erik K St Louis
- Mayo Center for Sleep Medicine, and Sleep Behavior and Neurophysiology Research Laboratory, Departments of Neurology and Medicine, Division of Pulmonary and Critical Care Medicine Mayo Clinic College of Medicine and Science Rochester, Rochester, MN, USA
- Mayo Clinic Health System Southwest Wisconsin, La Crosse, WI, USA
| | - Paul C Timm
- Mayo Center for Sleep Medicine, and Sleep Behavior and Neurophysiology Research Laboratory, Departments of Neurology and Medicine, Division of Pulmonary and Critical Care Medicine Mayo Clinic College of Medicine and Science Rochester, Rochester, MN, USA
| | - Luke N Teigen
- Mayo Center for Sleep Medicine, and Sleep Behavior and Neurophysiology Research Laboratory, Departments of Neurology and Medicine, Division of Pulmonary and Critical Care Medicine Mayo Clinic College of Medicine and Science Rochester, Rochester, MN, USA
| | - Annette Janzen
- Department of Neurology, Philipps University Marburg, Marburg, Germany
| | - Wolfgang Oertel
- Department of Neurology, Philipps University Marburg, Marburg, Germany
| | - Beatrice Heim
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Evi Holzknecht
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ambra Stefani
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Birgit Högl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Yves Dauvilliers
- National Reference Network for Narcolepsy, Sleep-Wake Disorder Unit, Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier, INSERM, University of Montpellier, Montpellier, France
| | - Elisa Evangelista
- National Reference Network for Narcolepsy, Sleep-Wake Disorder Unit, Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier, INSERM, University of Montpellier, Montpellier, France
| | - Karel Šonka
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jan Rusz
- Department of Circuit Theory, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
- Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Department of Neurology & ARTORG Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
40
|
Jones BM, McCarter SJ. Rapid Eye Movement Sleep Behavior Disorder: Clinical Presentation and Diagnostic Criteria. Sleep Med Clin 2024; 19:71-81. [PMID: 38368071 DOI: 10.1016/j.jsmc.2023.10.004] [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] [Indexed: 02/19/2024]
Abstract
Rapid eye movement (REM) sleep behavior disorder (RBD) classically presents with repetitive complex motor behavior during sleep with associated dream mentation. The diagnosis requires a history of repetitive complex motor behaviors and polysomnographic demonstration of REM sleep without atonia (RSWA) or capturing dream enactment behaviors. RSWA is best evaluated in the chin or flexor digitorum superficialis muscles. The anterior tibialis muscle is insufficiently accurate to be relied upon solely for RBD diagnosis. RBD may present with parkinsonism or cognitive impairment or may present in isolation. Patients should be monitored for parkinsonism, autonomic failure, or cognitive impairment.
Collapse
Affiliation(s)
- Brandon M Jones
- Department of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | - Stuart J McCarter
- Department of Neurology; Center for Sleep Medicine, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA.
| |
Collapse
|
41
|
Matsushima T, Yoshinaga K, Wakasugi N, Togo H, Hanakawa T. Functional connectivity-based classification of rapid eye movement sleep behavior disorder. Sleep Med 2024; 115:5-13. [PMID: 38295625 DOI: 10.1016/j.sleep.2024.01.019] [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: 04/23/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Isolated rapid eye movement sleep behavior disorder (iRBD) is a clinically important parasomnia syndrome preceding α-synucleinopathies, thereby prompting us to develop methods for evaluating latent brain states in iRBD. Resting-state functional magnetic resonance imaging combined with a machine learning-based classification technology may help us achieve this purpose. METHODS We developed a machine learning-based classifier using functional connectivity to classify 55 patients with iRBD and 97 healthy elderly controls (HC). Selecting 55 HCs randomly from the HC dataset 100 times, we conducted a classification of iRBD and HC for each sampling, using functional connectivity. Random forest ranked the importance of functional connectivity, which was subsequently used for classification with logistic regression and a support vector machine. We also conducted correlation analysis of the selected functional connectivity with subclinical variations in motor and non-motor functions in the iRBDs. RESULTS Mean classification performance using logistic regression was 0.649 for accuracy, 0.659 for precision, 0.662 for recall, 0.645 for f1 score, and 0.707 for the area under the receiver operating characteristic curve (p < 0.001 for all). The result was similar in the support vector machine. The classifier used functional connectivity information from nine connectivities across the motor and somatosensory areas, parietal cortex, temporal cortex, thalamus, and cerebellum. Inter-individual variations in functional connectivity were correlated with the subclinical motor and non-motor symptoms of iRBD patients. CONCLUSIONS Machine learning-based classifiers using functional connectivity may be useful to evaluate latent brain states in iRBD.
Collapse
Affiliation(s)
- Toma Matsushima
- Department of Advanced Neuroimaging, Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8501, Japan; Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Koganei, Tokyo, 184-8588, Japan
| | - Kenji Yoshinaga
- Department of Integrated Neuroanatomy and Neuroimaging, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Noritaka Wakasugi
- Department of Advanced Neuroimaging, Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8501, Japan
| | - Hiroki Togo
- Department of Advanced Neuroimaging, Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8501, Japan; Department of Integrated Neuroanatomy and Neuroimaging, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Takashi Hanakawa
- Department of Advanced Neuroimaging, Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8501, Japan; Department of Integrated Neuroanatomy and Neuroimaging, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan.
| |
Collapse
|
42
|
Zhang RY, Li FJ, Zhang Q, Xin LH, Huang JY, Zhao J. Causal associations between modifiable risk factors and isolated REM sleep behavior disorder: a mendelian randomization study. Front Neurol 2024; 15:1321216. [PMID: 38385030 PMCID: PMC10880103 DOI: 10.3389/fneur.2024.1321216] [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: 10/17/2023] [Accepted: 01/11/2024] [Indexed: 02/23/2024] Open
Abstract
Objectives This Mendelian randomization (MR) study identified modifiable risk factors for isolated rapid eye movement sleep behavior disorder (iRBD). Methods Genome-wide association study (GWAS) datasets for 29 modifiable risk factors for iRBD in discovery and replication stages were used. GWAS data for iRBD cases were obtained from the International RBD Study Group. The inverse variance weighted (IVW) method was primarily employed to explore causality, with supplementary analyses used to verify the robustness of IVW findings. Co-localization analysis further substantiated causal associations identified via MR. Genetic correlations between mental illness and iRBD were identified using trait covariance, linkage disequilibrium score regression, and co-localization analyses. Results Our study revealed causal associations between sun exposure-related factors and iRBD. Utilizing sun protection (odds ratio [OR] = 0.31 [0.14, 0.69], p = 0.004), ease of sunburn (OR = 0.70 [0.57, 0.87], p = 0.001), childhood sunburn occasions (OR = 0.58 [0.39, 0.87], p = 0.008), and phototoxic dermatitis (OR = 0.78 [0.66, 0.92], p = 0.003) decreased iRBD risk. Conversely, a deep skin color increased risk (OR = 1.42 [1.04, 1.93], p = 0.026). Smoking, alcohol consumption, low education levels, and mental illness were not risk factors for iRBD. Anxiety disorders and iRBD were genetically correlated. Conclusion Our study does not corroborate previous findings that identified smoking, alcohol use, low education, and mental illness as risk factors for iRBD. Moreover, we found that excessive sun exposure elevates iRBD risk. These findings offer new insights for screening high-risk populations and devising preventive measures.
Collapse
Affiliation(s)
- Ru-Yu Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Fu-Jia Li
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qian Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Li-Hong Xin
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jing-Ying Huang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jie Zhao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
43
|
Wang S, An N, Wang Y, Li Y, Li H, Bai Y. Knowledge mapping of prodromal Parkinson's disease: A bibliometric review and analysis (2000-2023). Medicine (Baltimore) 2024; 103:e36985. [PMID: 38306521 PMCID: PMC10843421 DOI: 10.1097/md.0000000000036985] [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: 11/22/2023] [Accepted: 12/22/2023] [Indexed: 02/04/2024] Open
Abstract
The prodromal period of Parkinson's disease (PD) is currently a hot topic in PD research. However, no bibliometric analysis has been conducted in this research field. This study aimed to provide a comprehensive overview of the status, hotspots, and trends in the prodromal period of PD using bibliometrics. CiteSpace and visualization of similarities viewer were used to analyze articles and reviews on the prodromal period of PD in the Web of Science Core Collection (WoSCC) database. We analyzed the data on countries, institutions, journals, authors, keywords, and cited references. In total, 909 articles from 65 countries, including the United States (n = 265, 29.15%) and Germany (n = 174, 19.14%), were included. The number of articles and reviews related to the prodromal period of PD has increased yearly. The University of Tubingen (n = 45, 4.95%), McGill University (n = 33, 3.63%), and University of London (n = 33, 3.63%) were the research institutions with the most published studies. Movement Disorders is the journal with the largest number of published papers (n = 98, 10.8%) and the most cited publications (co-citation = 7035). These publications are from 4681 authors, with Berg (n = 49, 5.39%) and Postuma (n = 40, 4.40%) publishing the most publications, and Postuma's study (n = 1206) having the most citations. Studying the nonmotor symptoms of PD precursors is a major topic in this research field. This is the first bibliometric study to comprehensively summarize the research trends and developments in the prodromal period of PD. This information identifies recent research frontiers and hotspots and provides a reference for scholars studying the prodromal period of PD.
Collapse
Affiliation(s)
- Shun Wang
- Department of Acupuncture and Moxibustion, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Ning An
- Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Heilongjiang, China
| | - Yulin Wang
- Department of Science and Technology, Heilongjiang University of Traditional Chinese Medicine, Heilongjiang, China
| | - Yuan Li
- Department of Acupuncture and Moxibustion, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, China
| | - Hailong Li
- Department of Acupuncture and Moxibustion, Heilongjiang Academy of Traditional Chinese Medicine, Heilongjiang, China
| | - Yan Bai
- Department of Acupuncture and Moxibustion, Heilongjiang Academy of Traditional Chinese Medicine, Institute of Acupuncture and Moxibustion, Heilongjiang, China
| |
Collapse
|
44
|
Kim J, Lee HJ, Lee DA, Park KM. Sarcopenia in patients with isolated rapid eye movement sleep behavior disorder. Sleep Med 2024; 114:189-193. [PMID: 38215670 DOI: 10.1016/j.sleep.2024.01.004] [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: 10/10/2023] [Revised: 12/17/2023] [Accepted: 01/03/2024] [Indexed: 01/14/2024]
Abstract
OBJECTIVES Evaluating of sarcopenia is important for promoting healthy aging, preventing functional decline, reducing the risk of falls and fractures, and improving overall quality of life. This study aimed to investigate sarcopenia in patients with isolated rapid eye movement sleep behavior disorder (RBD) using temporal muscle thickness (TMT) measurement. METHODS This investigation was retrospectively conducted at a single tertiary hospital. We recruited patients diagnosed with isolated RBD confirmed by polysomnography and clinical history and healthy participants as controls. Patients with isolated RBD and healthy controls underwent brain MRI scans, including three-dimensional T1-weighted imaging. We measured TMT, a radiographic marker of sarcopenia, based on the T1-weighted imaging. We compared the TMT between the groups and performed receiver operating characteristic (ROC) curve analysis to evaluate how well the TMT differentiated patients with isolated RBD from healthy controls. We also conducted a correlation analysis between the TMT and clinical factors. RESULTS Our study included 28 patients with isolated RBD and 30 healthy controls. There was a significant difference in the TMT of both groups. The TMT was reduced in patients with isolated RBD than in healthy controls (11.843 vs. 10.420 mm, p = 0.002). In the ROC curve analysis, the TMT exhibited good performance in differentiating patients with isolated RBD from healthy controls, with an area under the curve of 0.708. Furthermore, age was negatively correlated with TMT in patients with isolated RBD (r = -0.453, p = 0.015). CONCLUSION We demonstrate that TMT is reduced in patients with isolated RBD compared with healthy controls, confirming sarcopenia in patients with isolated RBD. The result suggests an association between neurodegeneration and sarcopenia. TMT can be used to evaluate sarcopenia in sleep disorders.
Collapse
Affiliation(s)
- Jinseung Kim
- Department of Family Medicine, Busan Paik Hospital, Inje University College of Medicine, Republic of Korea
| | - Ho-Joon Lee
- Department of Radiology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Dong Ah Lee
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Kang Min Park
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea.
| |
Collapse
|
45
|
Leclair-Visonneau L, Feemster JC, Bibi N, Gossard TR, Jagielski JT, Strainis EP, Carvalho DZ, Timm PC, Bliwise DL, Boeve BF, Silber MH, McCarter SJ, St. Louis EK. Contemporary diagnostic visual and automated polysomnographic REM sleep without atonia thresholds in isolated REM sleep behavior disorder. J Clin Sleep Med 2024; 20:279-291. [PMID: 37823585 PMCID: PMC10835777 DOI: 10.5664/jcsm.10862] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
STUDY OBJECTIVES Accurate diagnosis of isolated rapid eye movement (REM) sleep behavior disorder (iRBD) is crucial due to its injury potential and neurological prognosis. We aimed to analyze visual and automated REM sleep without atonia (RSWA) diagnostic thresholds applicable in varying clinical presentations in a contemporary cohort of patients with iRBD using submentalis (SM) and individual bilateral flexor digitorum superficialis (FDS) and anterior tibialis electromyography limb recordings during polysomnography. METHODS We analyzed RSWA in 20 patients with iRBD and 20 age-, REM-, apnea-hypopnea index-matched controls between 2017 and 2022 for phasic burst durations, density of phasic, tonic, and "any" muscle activity (number of 3-second mini-epochs containing phasic or tonic muscle activity divided by the total number of REM sleep 3-second mini-epochs), and automated Ferri REM atonia index (RAI). Group RSWA metrics were comparatively analyzed. Receiver operating characteristic curves determined optimized area under the curve (AUC) and maximized specificity and sensitivity diagnostic iRBD RSWA thresholds. RESULTS All mean RSWA metrics were higher in patients with iRBD than in controls (P < .05), except for selected anterior tibialis measures. Optimized, maximal specificity AUC diagnostic cutoffs for coprimary outcomes were: SM "any" 6.5%, 14.0% (AUC = 92.5%) and combined SM+FDS "any" 15.1%, 27.4% (AUC = 95.8%), while SM burst durations were 0.72, and 0.72 seconds (AUC 90.2%) and FDS RAI = 0.930, 0.888 (AUC 92.8%). CONCLUSIONS This study provides evidence for current quantitative RSWA diagnostic thresholds in chin and individual 4 limb muscles applicable in different iRBD clinical settings and confirms the key value of SM or SM+FDS to assure accurate iRBD diagnosis. Evolving iRBD recognition underscores the necessity of continuous assessment with future large, prospective, well-harmonized, multicenter polysomnographic analyses. CITATION Leclair-Visonneau L, Feemster JC, Bibi N, et al. Contemporary diagnostic visual and automated polysomnographic REM sleep without atonia thresholds in isolated REM sleep behavior disorder. J Clin Sleep Med. 2024;20(2):279-291.
Collapse
Affiliation(s)
- Laurène Leclair-Visonneau
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Rochester, Minnesota
- Mayo Center for Sleep Medicine, Rochester, Minnesota
- Department of Neurology, Mayo Clinic and Foundation, Rochester, Minnesota
- Department of Clinical Neurophysiology, CHU de Nantes, Nantes, France
- Nantes Université, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, Nantes, France
| | - John C. Feemster
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Rochester, Minnesota
- Mayo Center for Sleep Medicine, Rochester, Minnesota
- Department of Neurology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Noor Bibi
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Rochester, Minnesota
- Mayo Center for Sleep Medicine, Rochester, Minnesota
- Department of Neurology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Thomas R. Gossard
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Rochester, Minnesota
- Mayo Center for Sleep Medicine, Rochester, Minnesota
- Department of Neurology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Jack T. Jagielski
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Rochester, Minnesota
- Mayo Center for Sleep Medicine, Rochester, Minnesota
- Department of Neurology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Emma P. Strainis
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Rochester, Minnesota
- Mayo Center for Sleep Medicine, Rochester, Minnesota
- Department of Neurology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Diego Z. Carvalho
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Rochester, Minnesota
- Mayo Center for Sleep Medicine, Rochester, Minnesota
- Department of Neurology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Paul C. Timm
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Rochester, Minnesota
- Mayo Center for Sleep Medicine, Rochester, Minnesota
- Department of Neurology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Donald L. Bliwise
- Emory Sleep Center and Department of Neurology, Emory University, Atlanta, Georgia
| | - Bradley F. Boeve
- Mayo Center for Sleep Medicine, Rochester, Minnesota
- Department of Neurology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Michael H. Silber
- Mayo Center for Sleep Medicine, Rochester, Minnesota
- Department of Neurology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Stuart J. McCarter
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Rochester, Minnesota
- Mayo Center for Sleep Medicine, Rochester, Minnesota
- Department of Neurology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Erik K. St. Louis
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Rochester, Minnesota
- Mayo Center for Sleep Medicine, Rochester, Minnesota
- Department of Neurology, Mayo Clinic and Foundation, Rochester, Minnesota
- Department of Medicine, Mayo Clinic and Foundation, Rochester, Minnesota
- Department of Clinical and Translational Science, Mayo Clinic Health System Southwest Wisconsin, La Crosse, Wisconsin
| |
Collapse
|
46
|
Donzuso G, Cicero CE, Giuliano L, Squillaci R, Luca A, Palmucci S, Basile A, Lanza G, Ferri R, Zappia M, Nicoletti A. Neuroanatomical findings in isolated REM sleep behavior disorder and early Parkinson's disease: a Voxel-based morphometry study. Brain Imaging Behav 2024; 18:83-91. [PMID: 37897654 PMCID: PMC10844466 DOI: 10.1007/s11682-023-00815-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2023] [Indexed: 10/30/2023]
Abstract
Isolated rapid eye movement (REM) sleep behavior disorder (iRBD) is a parasomnia characterized by loss of physiological atonia of skeletal muscles with abnormal behaviors arising during REM sleep. RBD is often the early manifestation of neurodegenerative diseases, particularly alpha-synucleinopathies, such as Parkinson's disease (PD). Both structural and functional neuroimaging studies suggest that iRBD might share, or even precede, some of the features commonly found in PD, although without a definitive conclusion. Aim of the study is to evaluate the presence of structural abnormalities involving cortical and subcortical areas in PD patients with RBD and iRBD. Patients with video-polysomnographic (VPSG)-confirmed iRBD, and patients with a diagnosis of PD were recruited. In all PD patients, the presence of probable RBD was assessed during the follow-up visits (PD/pRBD). A group of healthy controls (HC) subjects was also recruited. Each subject underwent a structural brain MRI using a 3-D T1-weighted spoiled gradient echo sequence. Twenty-three patients with iRBD, 24 PD/pRBD, and 26 HC were enrolled. Voxel-based morphometry-AnCOVA analysis revealed clusters of grey matter changes in iRBD and PD/pRBD compared to HC in several regions, involving mainly the frontal and temporal regions. The involvement of cortical brain structures associated to the control of sleep cycle and REM stage both in PD/pRBD and iRBD might suggest the presence of a common structural platform linking iRBD and PD, although this pattern may not underlie exclusively RBD-related features. Further longitudinal studies are needed to clarify the patterns of changes occurring at different time points of RBD-related neurodegeneration.
Collapse
Affiliation(s)
- Giulia Donzuso
- Department of Medical, Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, Via Santa Sofia 78, 95123, Catania, Italy
| | - Calogero E Cicero
- Department of Medical, Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, Via Santa Sofia 78, 95123, Catania, Italy
| | - Loretta Giuliano
- Department of Medical, Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, Via Santa Sofia 78, 95123, Catania, Italy
| | - Raffaele Squillaci
- Department of Medical, Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, Via Santa Sofia 78, 95123, Catania, Italy
| | - Antonina Luca
- Department of Medical, Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, Via Santa Sofia 78, 95123, Catania, Italy
| | - Stefano Palmucci
- Radiodiagnostic and Radiotherapy Unit, University Hospital "Policlinico-San Marco", Via Santa Sofia 78, 95123, Catania, Italy
| | - Antonello Basile
- Radiodiagnostic and Radiotherapy Unit, University Hospital "Policlinico-San Marco", Via Santa Sofia 78, 95123, Catania, Italy
| | - Giuseppe Lanza
- Sleep Research Center, Department of Neurology IC, Oasi Research Institute - IRCCS, Troina, Italy
- Department of Surgery and Medical-Surgical Specialties, University of Catania, 95123, Catania, Italy
| | - Raffaele Ferri
- Sleep Research Center, Department of Neurology IC, Oasi Research Institute - IRCCS, Troina, Italy
| | - Mario Zappia
- Department of Medical, Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, Via Santa Sofia 78, 95123, Catania, Italy
| | - Alessandra Nicoletti
- Department of Medical, Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, Via Santa Sofia 78, 95123, Catania, Italy.
| |
Collapse
|
47
|
Naveed M, Chao OY, Hill JW, Yang YM, Huston JP, Cao R. Circadian neurogenetics and its implications in neurophysiology, behavior, and chronomedicine. Neurosci Biobehav Rev 2024; 157:105523. [PMID: 38142983 PMCID: PMC10872425 DOI: 10.1016/j.neubiorev.2023.105523] [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: 09/05/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 12/26/2023]
Abstract
The circadian rhythm affects multiple physiological processes, and disruption of the circadian system can be involved in a range of disease-related pathways. The genetic underpinnings of the circadian rhythm have been well-studied in model organisms. Significant progress has been made in understanding how clock genes affect the physiological functions of the nervous system. In addition, circadian timing is becoming a key factor in improving drug efficacy and reducing drug toxicity. The circadian biology of the target cell determines how the organ responds to the drug at a specific time of day, thus regulating pharmacodynamics. The current review brings together recent advances that have begun to unravel the molecular mechanisms of how the circadian clock affects neurophysiological and behavioral processes associated with human brain diseases. We start with a brief description of how the ubiquitous circadian rhythms are regulated at the genetic, cellular, and neural circuit levels, based on knowledge derived from extensive research on model organisms. We then summarize the latest findings from genetic studies of human brain disorders, focusing on the role of human clock gene variants in these diseases. Lastly, we discuss the impact of common dietary factors and medications on human circadian rhythms and advocate for a broader application of the concept of chronomedicine.
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Owen Y Chao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Joseph P Huston
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Ruifeng Cao
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA; Department of Neurology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
48
|
Huang J, Li W. Molecular crosstalk between circadian clock and NLRP3 inflammasome signaling in Parkinson's disease. Heliyon 2024; 10:e24752. [PMID: 38268831 PMCID: PMC10803942 DOI: 10.1016/j.heliyon.2024.e24752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 12/12/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. Research has recently found that both animal models and patients with PD have circadian dysfunction, accompanied by abnormal expression of circadian genes and proteins, which implies that the circadian clock plays a crucial role in PD etiopathogenesis. In addition, a strong relationship between NLRP3 inflammasome signaling and PD has been observed. Meanwhile, the activation of the NLRP3 inflammasome is highly relevant to dysfunctions of the molecular clock. Therefore, alleviating the neuroinflammation caused by NLRP3 inflammasome signaling by adjusting the abnormal molecular clock may be a potential strategy for preventing and treating PD. In this article, we have reviewed the potential or direct relationship between abnormalities of the circadian clock and NLRP3 inflammasome signaling in PD.
Collapse
Affiliation(s)
- Jiahua Huang
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, 201500, Shanghai, China
- Institute of Neurology, Institutes of Integrative Medicine, Fudan University, 201500, Shanghai, China
| | - Wenwei Li
- Laboratory of Neuropathology and Neuropharmacology, Department of Neurology, Shanghai Public Health Clinical Center, Fudan University, 201500, Shanghai, China
- Institute of Neurology, Institutes of Integrative Medicine, Fudan University, 201500, Shanghai, China
| |
Collapse
|
49
|
Fernandes M, Maio S, Eusebi P, Placidi F, Izzi F, Spanetta M, De Masi C, Lupo C, Calvello C, Nuccetelli M, Bernardini S, Mercuri NB, Liguori C. Cerebrospinal-fluid biomarkers for predicting phenoconversion in patients with isolated rapid-eye movement sleep behavior disorder. Sleep 2024; 47:zsad198. [PMID: 37542734 DOI: 10.1093/sleep/zsad198] [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: 02/24/2023] [Revised: 05/22/2023] [Indexed: 08/07/2023] Open
Abstract
STUDY OBJECTIVES Patients with isolated rapid-eye-movement sleep behavior disorder (iRBD) have an increased risk of developing neurodegenerative diseases. This study assessed cerebrospinal-fluid (CSF) biomarkers of neurodegeneration and blood-brain barrier (BBB) alteration in patients with iRBD compared to controls and ascertain whether these biomarkers may predict phenoconversion to alpha-synucleinopathies (Parkinson's Disease (PD), Dementia with Lewy bodies (DLB), Multiple System Atrophy (MSA)). METHODS Patients and controls underwent between 2012 and 2016 a neurological assessment, a lumbar puncture for CSF biomarker analysis (β-amyloid42 - Aβ42; total-tau, and phosphorylated tau), and BBB alteration (CSF/serum albumin ratio). All patients with iRBD were followed until 2021 and then classified into patients who converted to alpha-synucleinopathies (iRBD converters, cRBD) or not (iRBD non-converters, ncRBD). RESULTS Thirty-four patients with iRBD (mean age 67.12 ± 8.14) and 33 controls (mean age 64.97 ± 8.91) were included. At follow-up (7.63 ± 3.40 years), eight patients were ncRBD and 33 patients were cRBD: eleven converted to PD, 10 to DLB, and two to MSA. Patients with iRBD showed lower CSF Aβ42 levels and higher CSF/serum albumin ratio than controls. Cox regression analysis showed that the phenoconversion rate increases with higher motor impairment (hazard ratio [HR] = 1.23, p = 0.032). CSF Aβ42 levels predicted phenoconversion to DLB (HR = 0.67, p = 0.038) and BBB alteration predicted phenoconversion to PD (HR = 1.20, p = 0.038). DISCUSSION This study showed that low CSF Aβ42 levels and high BBB alteration may predict the phenoconversion to DLB and PD in patients with iRBD, respectively. These findings highlight the possibility to discriminate phenoconversion in iRBD patients through CSF biomarkers; however, further studies are needed.
Collapse
Affiliation(s)
- Mariana Fernandes
- Department of Systems Medicine, University of Rome 'Tor Vergata", Rome, Italy
| | - Silvia Maio
- Department of Systems Medicine, University of Rome 'Tor Vergata", Rome, Italy
- Sleep Medicine Centre, Neurology Unit, University Hospital "Tor Vergata", Rome, Italy
| | - Paolo Eusebi
- Department of Medicine, Neurology Clinic, University Hospital of Perugia, Italy
| | - Fabio Placidi
- Department of Systems Medicine, University of Rome 'Tor Vergata", Rome, Italy
- Sleep Medicine Centre, Neurology Unit, University Hospital "Tor Vergata", Rome, Italy
| | - Francesca Izzi
- Sleep Medicine Centre, Neurology Unit, University Hospital "Tor Vergata", Rome, Italy
| | - Matteo Spanetta
- Department of Systems Medicine, University of Rome 'Tor Vergata", Rome, Italy
| | - Claudia De Masi
- Sleep Medicine Centre, Neurology Unit, University Hospital "Tor Vergata", Rome, Italy
| | - Clementina Lupo
- Department of Systems Medicine, University of Rome 'Tor Vergata", Rome, Italy
| | - Carmen Calvello
- Department of Systems Medicine, University of Rome 'Tor Vergata", Rome, Italy
| | - Marzia Nuccetelli
- Department of Clinical Biochemistry and Molecular Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Sergio Bernardini
- Department of Clinical Biochemistry and Molecular Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome 'Tor Vergata", Rome, Italy
- Sleep Medicine Centre, Neurology Unit, University Hospital "Tor Vergata", Rome, Italy
| | - Claudio Liguori
- Department of Systems Medicine, University of Rome 'Tor Vergata", Rome, Italy
- Sleep Medicine Centre, Neurology Unit, University Hospital "Tor Vergata", Rome, Italy
| |
Collapse
|
50
|
Ren L, Yin X, Wang HY, Hao X, Wang D, Jin F, Zhang T, Li T, Zhou T, Liang Z. Correlation and underlying brain mechanisms between rapid eye movement sleep behavior disorder and executive functions in Parkinson's disease: an fNIRS study. Front Aging Neurosci 2024; 15:1290108. [PMID: 38274985 PMCID: PMC10809391 DOI: 10.3389/fnagi.2023.1290108] [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: 09/07/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
Purpose Rapid eye movement sleep behavior disorder (RBD) affects 30%-40% of patients with Parkinson's disease (PD) and has been linked to a higher risk of cognitive impairment, especially executive dysfunction. The aim of this study was to investigate the brain activation patterns in PD patients with RBD (PD-RBD+) compared to those without RBD (PD-RBD-) and healthy controls (HCs), and to analyze the correlation between changes in cerebral cortex activity and the severity of RBD. Methods We recruited 50 PD patients, including 30 PD-RBD+, 20 PD-RBD-, and 20 HCs. We used functional near infrared spectroscopy during a verbal fluency task (VFT-fNIRS) and clinical neuropsychological assessment to explore the correlation between PD-RBD+ and executive function and changes in neural activity. Results The VFT-fNIRS analysis revealed a significant reduction in activation among PD-RBD+ patients across multiple channels when compared to both the PD-RBD- and HC groups. Specifically, PD-RBD+ patients exhibited diminished activation in the bilateral dorsolateral prefrontal cortex (DLPFC) and the right ventrolateral prefrontal cortex (VLPFC) relative to their PD-RBD- counterparts. Furthermore, compared to the HC group, PD-RBD+ patients displayed reduced activation specifically in the right DLPFC. Significantly, a noteworthy negative correlation was identified between the average change in oxygenated hemoglobin concentration (ΔHbO2) in the right DLPFC of PD-RBD+ patients and the severity of their RBD. Conclusion Our study offers compelling evidence that RBD exacerbates cognitive impairment in PD, manifested as executive dysfunction, primarily attributed to reduced prefrontal activation. These aberrations in brain activation may potentially correlate with the severity of RBD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Tingting Zhou
- Department of Neurology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhanhua Liang
- Department of Neurology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|