1
|
Mylonas D, Patel R, Larson O, Zhu L, Vangel M, Baxter B, Manoach DS. Does fragmented sleep mediate the relationship between deficits in sleep spindles and memory consolidation in schizophrenia? SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2024; 6:zpae090. [PMID: 39811395 PMCID: PMC11725649 DOI: 10.1093/sleepadvances/zpae090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/28/2024] [Indexed: 01/16/2025]
Abstract
Study Objectives Sleep spindles, defining electroencephalographic oscillations of nonrapid eye movement (NREM) stage 2 sleep (N2), mediate sleep-dependent memory consolidation (SDMC). Spindles are also thought to protect sleep continuity by suppressing thalamocortical sensory relay. Schizophrenia is characterized by spindle deficits and a correlated reduction of SDMC. We investigated whether this relationship is mediated by sleep fragmentation. Methods We detected spindles (12-15 Hz) during N2 at central electrodes in overnight polysomnography records from 56 participants with chronic schizophrenia and 59 healthy controls. Our primary measures of sleep continuity were the sleep fragmentation index and, in a subset of the data, visually scored arousals. SDMC was measured as overnight improvement on the finger-tapping motor sequence task. Results Participants with schizophrenia showed reductions of both spindle density (#/min) and SDMC in the context of normal sleep continuity and architecture. Spindle density predicted SDMC in both groups. In contrast, neither increased sleep fragmentation nor arousals predicted lower spindle density or worse SDMC in either group. Conclusions Our findings fail to support the hypothesis that sleep fragmentation accounts for spindle deficits, impaired SDMC, or their relationship in individuals with chronic schizophrenia. Instead, our findings are consistent with the hypothesis that spindle deficits directly impair memory consolidation in schizophrenia. Since sleep continuity and architecture are intact in this population, research aimed at developing interventions should instead focus on understanding dysfunction within the thalamocortical-hippocampal circuitry that both generates spindles and synchronizes them with other NREM oscillations to mediate SDMC.
Collapse
Affiliation(s)
- Dimitrios Mylonas
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Rudra Patel
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Olivia Larson
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
| | - Lin Zhu
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark Vangel
- Department of Biostatistics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Bryan Baxter
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Dara S Manoach
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| |
Collapse
|
2
|
Dudysová D, Janků K, Piorecký M, Hantáková V, Orendáčová M, Piorecká V, Štrobl J, Kliková M, Ngo HV, Kopřivová J. Closed-loop auditory stimulation of slow-wave sleep in chronic insomnia: a pilot study. J Sleep Res 2024; 33:e14179. [PMID: 38467353 PMCID: PMC11597015 DOI: 10.1111/jsr.14179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/12/2023] [Accepted: 02/07/2024] [Indexed: 03/13/2024]
Abstract
Insomnia is a prevalent and disabling condition whose treatment is not always effective. This pilot study explores the feasibility and effects of closed-loop auditory stimulation (CLAS) as a potential non-invasive intervention to improve sleep, its subjective quality, and memory consolidation in patients with insomnia. A total of 27 patients with chronic insomnia underwent a crossover, sham-controlled study with 2 nights of either CLAS or sham stimulation. Polysomnography was used to record sleep parameters, while questionnaires and a word-pair memory task were administered to assess subjective sleep quality and memory consolidation. The initial analyses included 17 patients who completed the study, met the inclusion criteria, and received CLAS. From those, 10 (58%) received only a small number of stimuli. In the remaining seven (41%) patients with sufficient CLAS, we evaluated the acute and whole-night effect on sleep. CLAS led to a significant immediate increase in slow oscillation (0.5-1 Hz) amplitude and activity, and reduced delta (1-4 Hz) and sigma/sleep spindle (12-15 Hz) activity during slow-wave sleep across the whole night. All these fundamental sleep rhythms are implicated in sleep-dependent memory consolidation. Yet, CLAS did not change sleep-dependent memory consolidation or sleep macrostructure characteristics, number of arousals, or subjective perception of sleep quality. Results showed CLAS to be feasible in patients with insomnia. However, a high variance in the efficacy of our automated stimulation approach suggests that further research is needed to optimise stimulation protocols to better unlock potential CLAS benefits for sleep structure and subjective sleep quality in such clinical settings.
Collapse
Affiliation(s)
- Daniela Dudysová
- National Institute of Mental HealthKlecanyCzech Republic
- Third Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Karolina Janků
- National Institute of Mental HealthKlecanyCzech Republic
| | - Marek Piorecký
- National Institute of Mental HealthKlecanyCzech Republic
- Faculty of Biomedical EngineeringCzech Technical University in PraguePragueCzech Republic
| | - Veronika Hantáková
- National Institute of Mental HealthKlecanyCzech Republic
- School of Medicine, Medical Sciences and NutritionUniversity of AberdeenAberdeenScotland
| | - Mária Orendáčová
- National Institute of Mental HealthKlecanyCzech Republic
- Third Faculty of MedicineCharles UniversityPragueCzech Republic
- Department of Physiology, Faculty of ScienceCharles University in PraguePragueCzech Republic
| | - Václava Piorecká
- National Institute of Mental HealthKlecanyCzech Republic
- Faculty of Biomedical EngineeringCzech Technical University in PraguePragueCzech Republic
| | - Jan Štrobl
- National Institute of Mental HealthKlecanyCzech Republic
- Faculty of Biomedical EngineeringCzech Technical University in PraguePragueCzech Republic
| | - Monika Kliková
- National Institute of Mental HealthKlecanyCzech Republic
| | - Hong‐Viet V. Ngo
- Center for Brain, Behaviour and MetabolismUniversity of LübeckLübeckGermany
- Department of PsychologyUniversity of LübeckLübeckGermany
- Department of PsychologyUniversity of EssexColchesterUK
| | - Jana Kopřivová
- National Institute of Mental HealthKlecanyCzech Republic
- Third Faculty of MedicineCharles UniversityPragueCzech Republic
| |
Collapse
|
3
|
Zeller CJ, Wunderlin M, Wicki K, Teunissen CE, Nissen C, Züst MA, Klöppel S. Multi-night acoustic stimulation is associated with better sleep, amyloid dynamics, and memory in older adults with cognitive impairment. GeroScience 2024; 46:6157-6172. [PMID: 38744792 PMCID: PMC11493878 DOI: 10.1007/s11357-024-01195-z] [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: 02/19/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Sleep is a potential early, modifiable risk factor for cognitive decline and dementia. Impaired slow wave sleep (SWS) is pronounced in individuals with cognitive impairment (CI). Cognitive decline and impairments of SWS are bi-directionally linked in a vicious cycle. SWS can be enhanced non-invasively using phase-locked acoustic stimulation (PLAS), potentially breaking this vicious cycle. Eighteen healthy older adults (HC, agemean±sd, 68.3 ± 5.1) and 16 older adults (agemean±sd, 71.9 ± 3.9) with CI (Montreal Cognitive Assessment ≤ 25) underwent one baseline (sham-PLAS) night and three consecutive stimulation nights (real-PLAS). EEG responses and blood-plasma amyloid beta Aβ42/Aβ40 ratio were measured pre- and post-intervention, as was episodic memory. The latter was again evaluated 1 week and 3 months after the intervention. In both groups, PLAS induced a significant electrophysiological response in both voltage- and time-frequency analyses, and memory performance improved in association with the magnitude of this response. In the CI group, both electrophysiological and associated memory effects were delayed compared to the healthy group. After 3 intervention nights, electrophysiological response to PLAS was no longer different between CI and HC groups. Only in the CI sample, stronger electrophysiological responses were significantly associated with improving post-intervention Aβ42/Aβ40 ratios. PLAS seems to improve SWS electrophysiology, memory, and amyloid dynamics in older adults with CI. However, effects on memory require more time to unfold compared to healthy older adults. This indicates that PLAS may become a potential tool to ameliorate cognitive decline, but longer interventions are necessary to compensate for declining brain integrity. This study was pre-registered (clinicaltrials.gov: NCT04277104).
Collapse
Affiliation(s)
- Céline J Zeller
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000, Bern 60, Switzerland
- Graduate School for Health Sciences, University of Bern, 3012, Bern, Switzerland
| | - Marina Wunderlin
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000, Bern 60, Switzerland
| | - Korian Wicki
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000, Bern 60, Switzerland
- Graduate School for Health Sciences, University of Bern, 3012, Bern, Switzerland
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, Netherlands
| | - Christoph Nissen
- Division of Psychiatric Specialties, Department of Psychiatry, Geneva University Hospitals (HUG), 1201, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, 1201, Geneva, Switzerland
| | - Marc A Züst
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000, Bern 60, Switzerland.
| | - Stefan Klöppel
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000, Bern 60, Switzerland
| |
Collapse
|
4
|
Carro-Domínguez M, Huwiler S, Stich FM, Sala R, Aziri F, Trippel A, Heimhofer C, Huber R, Meissner SN, Wenderoth N, Lustenberger C. Overnight changes in performance fatigability and their relationship to modulated deep sleep oscillations via auditory stimulation. J Sleep Res 2024:e14371. [PMID: 39420437 DOI: 10.1111/jsr.14371] [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: 07/01/2024] [Revised: 09/11/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Deep sleep oscillations are proposed to be central in restoring brain function and to affect different aspects of motor performance such as facilitating the consolidation of motor sequences resulting in faster and more accurate sequence tapping. Yet, whether deep sleep modulates performance fatigability during fatiguing tasks remains unexplored. We investigated overnight changes in tapping speed and resistance against performance fatigability via a finger tapping task. During fast tapping, fatigability manifests as a reduction in speed (or "motor slowing") which affects all tapping tasks, including motor sequences used to study motor memory formation. We further tested whether overnight changes in performance fatigability are influenced by enhancing deep sleep oscillations using auditory stimulation. We found an overnight increase in tapping speed alongside a reduction in performance fatigability and perceived workload. Auditory stimulation led to a global enhancement of slow waves and both slow and fast spindles during the stimulation window and a local increase in slow spindles in motor areas across the night. However, overnight performance improvements were not significantly modulated by auditory stimulation and changes in tapping speed or performance fatigability were not predicted by individual changes in deep sleep oscillations. Our findings demonstrate overnight changes in fatigability but revealed no evidence suggesting that this effect is causally linked to temporary augmentation of slow waves or sleep spindles. Our results are important for future studies using tapping tasks to test the relationship between sleep and motor memory consolidation, as overnight changes in objectively measured and subjectively perceived fatigue likely impact behavioural outcomes.
Collapse
Affiliation(s)
- Manuel Carro-Domínguez
- Neural Control of Movement Laboratory, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Stephanie Huwiler
- Neural Control of Movement Laboratory, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Fabia M Stich
- Neural Control of Movement Laboratory, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Rossella Sala
- Neural Control of Movement Laboratory, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Florent Aziri
- Neural Control of Movement Laboratory, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Anna Trippel
- Neural Control of Movement Laboratory, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Caroline Heimhofer
- Neural Control of Movement Laboratory, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Reto Huber
- Centre of Competence Sleep & Health Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich (ZNZ), University of Zurich, ETH Zurich, Zurich, Switzerland
- Child Development Centre, University Children's Hospital, University of Zurich, Zurich, Switzerland
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sarah Nadine Meissner
- Neural Control of Movement Laboratory, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Nicole Wenderoth
- Neural Control of Movement Laboratory, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich (ZNZ), University of Zurich, ETH Zurich, Zurich, Switzerland
- Future Health Technologies, Singapore-ETH Center, Campus for Research Excellence and Technological Enterprise (CREATE), Singapore
| | - Caroline Lustenberger
- Neural Control of Movement Laboratory, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Centre of Competence Sleep & Health Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich (ZNZ), University of Zurich, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Carvalho DZ, Kremen V, Mivalt F, St. Louis EK, McCarter SJ, Bukartyk J, Przybelski SA, Kamykowski MG, Spychalla AJ, Machulda MM, Boeve BF, Petersen RC, Jack CR, Lowe VJ, Graff-Radford J, Worrell GA, Somers VK, Varga AW, Vemuri P. Non-rapid eye movement sleep slow-wave activity features are associated with amyloid accumulation in older adults with obstructive sleep apnoea. Brain Commun 2024; 6:fcae354. [PMID: 39429245 PMCID: PMC11487750 DOI: 10.1093/braincomms/fcae354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/12/2024] [Accepted: 10/04/2024] [Indexed: 10/22/2024] Open
Abstract
Obstructive sleep apnoea (OSA) is associated with an increased risk for cognitive impairment and dementia, which likely involves Alzheimer's disease pathology. Non-rapid eye movement slow-wave activity (SWA) has been implicated in amyloid clearance, but it has not been studied in the context of longitudinal amyloid accumulation in OSA. This longitudinal retrospective study aims to investigate the relationship between polysomnographic and electrophysiological SWA features and amyloid accumulation. From the Mayo Clinic Study of Aging cohort, we identified 71 participants ≥60 years old with OSA (mean baseline age = 72.9 ± 7.5 years, 60.6% male, 93% cognitively unimpaired) who had at least 2 consecutive Amyloid Pittsburgh Compound B (PiB)-PET scans and a polysomnographic study within 5 years of the baseline scan and before the second scan. Annualized PiB-PET accumulation [global ΔPiB(log)/year] was estimated by the difference between the second and first log-transformed global PiB-PET uptake estimations divided by the interval between scans (years). Sixty-four participants were included in SWA analysis. SWA was characterized by the mean relative spectral power density (%) in slow oscillation (SO: 0.5-0.9 Hz) and delta (1-3.9 Hz) frequency bands and by their downslopes (SO-slope and delta-slope, respectively) during the diagnostic portion of polysomnography. We fit linear regression models to test for associations among global ΔPiB(log)/year, SWA features (mean SO% and delta% or mean SO-slope and delta-slope), and OSA severity markers, after adjusting for age at baseline PiB-PET, APOE ɛ4 and baseline amyloid positivity. For 1 SD increase in SO% and SO-slope, global ΔPiB(log)/year increased by 0.0033 (95% CI: 0.0001; 0.0064, P = 0.042) and 0.0069 (95% CI: 0.0009; 0.0129, P = 0.026), which were comparable to 32% and 59% of the effect size associated with baseline amyloid positivity, respectively. Delta-slope was associated with a reduction in global ΔPiB(log)/year by -0.0082 (95% CI: -0.0143; -0.0021, P = 0.009). Sleep apnoea severity was not associated with amyloid accumulation. Regional associations were stronger in the pre-frontal region. Both slow-wave slopes had more significant and widespread regional associations. Annualized PiB-PET accumulation was positively associated with SO and SO-slope, which may reflect altered sleep homeostasis due to increased homeostatic pressure in the setting of unmet sleep needs, increased synaptic strength, and/or hyper-excitability in OSA. Delta-slope was inversely associated with PiB-PET accumulation, suggesting it may represent residual physiological activity. Further investigation of SWA dynamics in the presence of sleep disorders before and after treatment is necessary for understanding the relationship between amyloid accumulation and SWA physiology.
Collapse
Affiliation(s)
- Diego Z Carvalho
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Center for Sleep Medicine, Rochester, MN 55905, USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Vaclav Kremen
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Filip Mivalt
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Erik K St. Louis
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Center for Sleep Medicine, Rochester, MN 55905, USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Stuart J McCarter
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Center for Sleep Medicine, Rochester, MN 55905, USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jan Bukartyk
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Bradley F Boeve
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Center for Sleep Medicine, Rochester, MN 55905, USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Virend K Somers
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Andrew W Varga
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | |
Collapse
|
6
|
Beauchemin N, Charland P, Karran A, Boasen J, Tadson B, Sénécal S, Léger PM. Enhancing learning experiences: EEG-based passive BCI system adapts learning speed to cognitive load in real-time, with motivation as catalyst. Front Hum Neurosci 2024; 18:1416683. [PMID: 39435350 PMCID: PMC11491376 DOI: 10.3389/fnhum.2024.1416683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/26/2024] [Indexed: 10/23/2024] Open
Abstract
Computer-based learning has gained popularity in recent years, providing learners greater flexibility and freedom. However, these learning environments do not consider the learner's mental state in real-time, resulting in less optimized learning experiences. This research aimed to explore the effect on the learning experience of a novel EEG-based Brain-Computer Interface (BCI) that adjusts the speed of information presentation in real-time during a learning task according to the learner's cognitive load. We also explored how motivation moderated these effects. In accordance with three experimental groups (non-adaptive, adaptive, and adaptive with motivation), participants performed a calibration task (n-back), followed by a memory-based learning task concerning astrological constellations. Learning gains were assessed based on performance on the learning task. Self-perceived mental workload, cognitive absorption and satisfaction were assessed using a post-test questionnaire. Between-group analyses using Mann-Whitney tests suggested that combining BCI and motivational factors led to more significant learning gains and an improved learning experience. No significant difference existed between the BCI without motivational factor and regular non-adaptive interface for overall learning gains, self-perceived mental workload, and cognitive absorption. However, participants who undertook the experiment with an imposed learning pace reported higher overall satisfaction with their learning experience and a higher level of temporal stress. Our findings suggest BCI's potential applicability and feasibility in improving memorization-based learning experiences. Further work should seek to optimize the BCI adaptive index and explore generalizability to other learning contexts.
Collapse
Affiliation(s)
- Noémie Beauchemin
- Tech3Lab, HEC Montréal, Information Technology Department, Montreal, QC, Canada
| | - Patrick Charland
- Didactics Department, Université du Québec à Montréal, Montreal, QC, Canada
| | - Alexander Karran
- Tech3Lab, HEC Montréal, Information Technology Department, Montreal, QC, Canada
| | - Jared Boasen
- Tech3Lab, HEC Montréal, Information Technology Department, Montreal, QC, Canada
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Bella Tadson
- Tech3Lab, HEC Montréal, Information Technology Department, Montreal, QC, Canada
| | - Sylvain Sénécal
- Tech3Lab, HEC Montréal, Information Technology Department, Montreal, QC, Canada
| | | |
Collapse
|
7
|
Dias I, Kollarik S, Siegel M, Baumann CR, Moreira CG, Noain D. Novel murine closed-loop auditory stimulation paradigm elicits macrostructural sleep benefits in neurodegeneration. J Sleep Res 2024:e14316. [PMID: 39223830 DOI: 10.1111/jsr.14316] [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: 02/09/2024] [Revised: 07/05/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Boosting slow-wave activity (SWA) by modulating slow waves through closed-loop auditory stimulation (CLAS) might provide a powerful non-pharmacological tool to investigate the link between sleep and neurodegeneration. Here, we established mouse CLAS (mCLAS)-mediated SWA enhancement and explored its effects on sleep deficits in neurodegeneration, by targeting the up-phase of slow waves in mouse models of Alzheimer's disease (AD, Tg2576) and Parkinson's disease (PD, M83). We found that tracking a 2 Hz component of slow waves leads to highest precision of non-rapid eye movement (NREM) sleep detection in mice, and that its combination with a 30° up-phase target produces a significant 15-30% SWA increase from baseline in wild-type (WTAD) and transgenic (TGAD) mice versus a mock stimulation group. Conversely, combining 2 Hz with a 40° phase target yields a significant increase ranging 30-35% in WTPD and TGPD mice. Interestingly, these phase-target-triggered SWA increases are not genotype dependent but strain specific. Sleep alterations that may contribute to disease progression and burden were described in AD and PD lines. Notably, pathological sleep traits were rescued by mCLAS, which elicited a 14% decrease of pathologically heightened NREM sleep fragmentation in TGAD mice, accompanied by a steep decrease in microarousal events during both light and dark periods. Overall, our results indicate that model-tailored phase targeting is key to modulate SWA through mCLAS, prompting the acute alleviation of key neurodegeneration-associated sleep phenotypes and potentiating sleep regulation and consolidation. Further experiments assessing the long-term effect of mCLAS in neurodegeneration may majorly impact the establishment of sleep-based therapies.
Collapse
Affiliation(s)
- Inês Dias
- Department of Neurology, University Hospital Zurich (USZ), Schlieren, Switzerland
- Department of Health Sciences and Technology (D-HEST), ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich (UZH), Zurich, Switzerland
| | - Sedef Kollarik
- Department of Neurology, University Hospital Zurich (USZ), Schlieren, Switzerland
| | - Michelle Siegel
- Department of Neurology, University Hospital Zurich (USZ), Schlieren, Switzerland
| | - Christian R Baumann
- Department of Neurology, University Hospital Zurich (USZ), Schlieren, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich (UZH), Zurich, Switzerland
- Center of Competence Sleep and Health, University of Zurich (UZH), Zurich, Switzerland
| | - Carlos G Moreira
- Department of Neurology, University Hospital Zurich (USZ), Schlieren, Switzerland
| | - Daniela Noain
- Department of Neurology, University Hospital Zurich (USZ), Schlieren, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich (UZH), Zurich, Switzerland
- Center of Competence Sleep and Health, University of Zurich (UZH), Zurich, Switzerland
| |
Collapse
|
8
|
Leach S, Krugliakova E, Sousouri G, Snipes S, Skorucak J, Schühle S, Müller M, Ferster ML, Da Poian G, Karlen W, Huber R. Acoustically evoked K-complexes together with sleep spindles boost verbal declarative memory consolidation in healthy adults. Sci Rep 2024; 14:19184. [PMID: 39160150 PMCID: PMC11333484 DOI: 10.1038/s41598-024-67701-7] [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: 10/13/2023] [Accepted: 07/15/2024] [Indexed: 08/21/2024] Open
Abstract
Over the past decade, phase-targeted auditory stimulation (PTAS), a neuromodulation approach which presents auditory stimuli locked to the ongoing phase of slow waves during sleep, has shown potential to enhance specific aspects of sleep functions. However, the complexity of PTAS responses complicates the establishment of causality between specific electroencephalographic events and observed benefits. Here, we used down-PTAS during sleep to specifically evoke the early, K-complex (KC)-like response following PTAS without leading to a sustained increase in slow-wave activity throughout the stimulation window. Over the course of two nights, one with down-PTAS, the other without, high-density electroencephalography (hd-EEG) was recorded from 14 young healthy adults. The early response exhibited striking similarities to evoked KCs and was associated with improved verbal memory consolidation via stimulus-evoked spindle events nested into the up-phase of ongoing 1 Hz waves in a central region. These findings suggest that the early, KC-like response is sufficient to boost memory, potentially by orchestrating aspects of the hippocampal-neocortical dialogue.
Collapse
Affiliation(s)
- Sven Leach
- Child Development Centre and Children's Research Centre, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Elena Krugliakova
- Child Development Centre and Children's Research Centre, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | - Georgia Sousouri
- Child Development Centre and Children's Research Centre, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
- Mobile Health Systems Lab, Department of Health Sciences and Technology, Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich, Switzerland
- Institute of Pharmacology & Toxicology, University of Zurich, Zurich, Switzerland
| | - Sophia Snipes
- Child Development Centre and Children's Research Centre, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jelena Skorucak
- Child Development Centre and Children's Research Centre, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Selina Schühle
- Child Development Centre and Children's Research Centre, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Manuel Müller
- Child Development Centre and Children's Research Centre, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Maria Laura Ferster
- Mobile Health Systems Lab, Department of Health Sciences and Technology, Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich, Switzerland
| | - Giulia Da Poian
- Sensory-Motor Systems Lab, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Walter Karlen
- Mobile Health Systems Lab, Department of Health Sciences and Technology, Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich, Switzerland
- Institute of Biomedical Engineering, Faculty of Engineering, Computer Science and Psychology, Ulm University, Ulm, Germany
| | - Reto Huber
- Child Development Centre and Children's Research Centre, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland.
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric Hospital, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
9
|
van der Heijden AC, van der Werf YD, van den Heuvel OA, Talamini LM, van Marle HJF. Targeted memory reactivation to augment treatment in post-traumatic stress disorder. Curr Biol 2024; 34:3735-3746.e5. [PMID: 39116885 DOI: 10.1016/j.cub.2024.07.019] [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: 06/20/2022] [Revised: 01/30/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024]
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric disorder with traumatic memories at its core. Post-treatment sleep may offer a unique time window to increase therapeutic efficacy through consolidation of therapeutically modified traumatic memories. Targeted memory reactivation (TMR) enhances memory consolidation by presenting reminder cues (e.g., sounds associated with a memory) during sleep. Here, we applied TMR in PTSD patients to strengthen therapeutic memories during sleep after one treatment session with eye movement desensitization and reprocessing (EMDR). PTSD patients received either slow oscillation (SO) phase-targeted TMR, using modeling-based closed-loop neurostimulation (M-CLNS) with EMDR clicks as a reactivation cue (n = 17), or sham stimulation (n = 16). Effects of TMR on sleep were assessed through high-density polysomnography. Effects on treatment outcome were assessed through subjective, autonomic, and fMRI responses to script-driven imagery (SDI) of the targeted traumatic memory and overall PTSD symptom level. Compared to sham stimulation, TMR led to stimulus-locked increases in SO and spindle dynamics, which correlated positively with PTSD symptom reduction in the TMR group. Given the role of SOs and spindles in memory consolidation, these findings suggest that TMR may have strengthened the consolidation of the EMDR-treatment memory. Clinically, TMR vs. sham stimulation resulted in a larger reduction of avoidance level during SDI. TMR did not disturb sleep or trigger nightmares. Together, these data provide first proof of principle that TMR may be a safe and viable future treatment augmentation strategy for PTSD. The required follow-up studies may implement multi-night TMR or TMR during REM sleep to further establish the clinical effect of TMR for traumatic memories.
Collapse
Affiliation(s)
- Anna C van der Heijden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department Anatomy & Neuroscience, Boelelaan 1081 HV Amsterdam, the Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Psychiatry, Oldenaller 1081 HJ Amsterdam, the Netherlands; Amsterdam Neuroscience, Mood Anxiety Psychosis Stress Sleep, Boelelaan 1081 HV Amsterdam, the Netherlands; University of Amsterdam, Department of Psychology, Brain & Cognition, Nieuwe Achtergracht 1018 WS Amsterdam, the Netherlands
| | - Ysbrand D van der Werf
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department Anatomy & Neuroscience, Boelelaan 1081 HV Amsterdam, the Netherlands; Amsterdam Neuroscience, Compulsivity Impulsivity and Attention, Boelelaan 1081 HV Amsterdam, the Netherlands
| | - Odile A van den Heuvel
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department Anatomy & Neuroscience, Boelelaan 1081 HV Amsterdam, the Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Psychiatry, Oldenaller 1081 HJ Amsterdam, the Netherlands; Amsterdam Neuroscience, Compulsivity Impulsivity and Attention, Boelelaan 1081 HV Amsterdam, the Netherlands
| | - Lucia M Talamini
- University of Amsterdam, Department of Psychology, Brain & Cognition, Nieuwe Achtergracht 1018 WS Amsterdam, the Netherlands; University of Amsterdam, Amsterdam Brain and Cognition, Nieuwe Achtergracht 1001 NK Amsterdam, the Netherlands
| | - Hein J F van Marle
- Amsterdam UMC, Vrije Universiteit Amsterdam, Psychiatry, Oldenaller 1081 HJ Amsterdam, the Netherlands; Amsterdam Neuroscience, Mood Anxiety Psychosis Stress Sleep, Boelelaan 1081 HV Amsterdam, the Netherlands; GGZ inGeest Mental Health Care, Oldenaller 1081 HJ Amsterdam, the Netherlands; ARQ National Psychotrauma Center, Nienoord 1112 XE Diemen, the Netherlands.
| |
Collapse
|
10
|
Balsamo F, Berretta E, Meneo D, Baglioni C, Gelfo F. The Complex Relationship between Sleep and Cognitive Reserve: A Narrative Review Based on Human Studies. Brain Sci 2024; 14:654. [PMID: 39061395 PMCID: PMC11274941 DOI: 10.3390/brainsci14070654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Sleep and brain/cognitive/neural reserve significantly impact well-being and cognition throughout life. This review aims to explore the intricate relationship between such factors, with reference to their effects on human cognitive functions. The specific goal is to understand the bidirectional influence that sleep and reserve exert on each other. Up to 6 February 2024, a methodical search of the literature was conducted using the PubMed database with terms related to brain, cognitive or neural reserve, and healthy or disturbed sleep. Based on the inclusion criteria, 11 articles were selected and analyzed for this review. The articles focus almost exclusively on cognitive reserve, with no explicit connection between sleep and brain or neural reserve. The results evidence sleep's role as a builder of cognitive reserve and cognitive reserve's role as a moderator in the effects of physiological and pathological sleep on cognitive functions. In conclusion, the findings of the present review support the notion that both sleep and cognitive reserve are critical factors in cognitive functioning. Deepening comprehension of the interactions between them is essential for devising strategies to enhance brain health and resilience against age- and pathology-related conditions.
Collapse
Affiliation(s)
- Francesca Balsamo
- Department of Human Sciences, Guglielmo Marconi University, 00193 Rome, Italy
- IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | | | - Debora Meneo
- Department of Human Sciences, Guglielmo Marconi University, 00193 Rome, Italy
| | - Chiara Baglioni
- Department of Human Sciences, Guglielmo Marconi University, 00193 Rome, Italy
- Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Francesca Gelfo
- Department of Human Sciences, Guglielmo Marconi University, 00193 Rome, Italy
- IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| |
Collapse
|
11
|
Bressler S, Neely R, Yost RM, Wang D. A randomized controlled trial of alpha phase-locked auditory stimulation to treat symptoms of sleep onset insomnia. Sci Rep 2024; 14:13039. [PMID: 38844793 PMCID: PMC11156862 DOI: 10.1038/s41598-024-63385-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Sleep onset insomnia is a pervasive problem that contributes significantly to the poor health outcomes associated with insufficient sleep. Auditory stimuli phase-locked to slow-wave sleep oscillations have been shown to augment deep sleep, but it is unknown whether a similar approach can be used to accelerate sleep onset. The present randomized controlled crossover trial enrolled adults with objectively verified sleep onset latencies (SOLs) greater than 30 min to test the effect of auditory stimuli delivered at specific phases of participants' alpha oscillations prior to sleep onset. During the intervention week, participants wore an electroencephalogram (EEG)-enabled headband that delivered acoustic pulses timed to arrive anti-phase with alpha for 30 min (Stimulation). During the Sham week, the headband silently recorded EEG. The primary outcome was SOL determined by blinded scoring of EEG records. For the 21 subjects included in the analyses, stimulation had a significant effect on SOL according to a linear mixed effects model (p = 0.0019), and weekly average SOL decreased by 10.5 ± 15.9 min (29.3 ± 44.4%). These data suggest that phase-locked acoustic stimulation can be a viable alternative to pharmaceuticals to accelerate sleep onset in individuals with prolonged sleep onset latencies. Trial Registration: This trial was first registered on clinicaltrials.gov on 24/02/2023 under the name Sounds Locked to ElectroEncephalogram Phase For the Acceleration of Sleep Onset Time (SLEEPFAST), and assigned registry number NCT05743114.
Collapse
Affiliation(s)
- Scott Bressler
- Elemind Technologies, Inc., Cambridge, MA, USA
- Science and Research, Elemind Technologies, Inc., Cambridge, MA, 02139, USA
| | - Ryan Neely
- Elemind Technologies, Inc., Cambridge, MA, USA.
- Science and Research, Elemind Technologies, Inc., Cambridge, MA, 02139, USA.
| | - Ryan M Yost
- Elemind Technologies, Inc., Cambridge, MA, USA
- Science and Research, Elemind Technologies, Inc., Cambridge, MA, 02139, USA
| | - David Wang
- Elemind Technologies, Inc., Cambridge, MA, USA
| |
Collapse
|
12
|
Smith SK, Kafashan M, Rios RL, Brown EN, Landsness EC, Guay CS, Palanca BJA. Daytime dexmedetomidine sedation with closed-loop acoustic stimulation alters slow wave sleep homeostasis in healthy adults. BJA OPEN 2024; 10:100276. [PMID: 38571816 PMCID: PMC10990715 DOI: 10.1016/j.bjao.2024.100276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/29/2024] [Indexed: 04/05/2024]
Abstract
Background The alpha-2 adrenergic agonist dexmedetomidine induces EEG patterns resembling those of non-rapid eye movement (NREM) sleep. Fulfilment of slow wave sleep (SWS) homeostatic needs would address the assumption that dexmedetomidine induces functional biomimetic sleep states. Methods In-home sleep EEG recordings were obtained from 13 healthy participants before and after dexmedetomidine sedation. Dexmedetomidine target-controlled infusions and closed-loop acoustic stimulation were implemented to induce and enhance EEG slow waves, respectively. EEG recordings during sedation and sleep were staged using modified American Academy of Sleep Medicine criteria. Slow wave activity (EEG power from 0.5 to 4 Hz) was computed for NREM stage 2 (N2) and NREM stage 3 (N3/SWS) epochs, with the aggregate partitioned into quintiles by time. The first slow wave activity quintile served as a surrogate for slow wave pressure, and the difference between the first and fifth quintiles as a measure of slow wave pressure dissipation. Results Compared with pre-sedation sleep, post-sedation sleep showed reduced N3 duration (mean difference of -17.1 min, 95% confidence interval -30.0 to -8.2, P=0.015). Dissipation of slow wave pressure was reduced (P=0.02). Changes in combined durations of N2 and N3 between pre- and post-sedation sleep correlated with total dexmedetomidine dose, (r=-0.61, P=0.03). Conclusions Daytime dexmedetomidine sedation and closed-loop acoustic stimulation targeting EEG slow waves reduced N3/SWS duration and measures of slow wave pressure dissipation on the post-sedation night in healthy young adults. Thus, the paired intervention induces sleep-like states that fulfil certain homeostatic NREM sleep needs in healthy young adults. Clinical trial registration ClinicalTrials.gov NCT04206059.
Collapse
Affiliation(s)
- S. Kendall Smith
- Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Center on Biological Rhythms and Sleep, Washington University in St. Louis, St. Louis, MO, USA
| | - MohammadMehdi Kafashan
- Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Center on Biological Rhythms and Sleep, Washington University in St. Louis, St. Louis, MO, USA
| | - Rachel L. Rios
- Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Center on Biological Rhythms and Sleep, Washington University in St. Louis, St. Louis, MO, USA
| | - Emery N. Brown
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eric C. Landsness
- Center on Biological Rhythms and Sleep, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neurology, Division of Sleep Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Christian S. Guay
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ben Julian A. Palanca
- Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Center on Biological Rhythms and Sleep, Washington University in St. Louis, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
13
|
Moguilner SG, Berezuk C, Bender AC, Pellerin KR, Gomperts SN, Cash SS, Sarkis RA, Lam AD. Sleep functional connectivity, hyperexcitability, and cognition in Alzheimer's disease. Alzheimers Dement 2024; 20:4234-4249. [PMID: 38764252 PMCID: PMC11180941 DOI: 10.1002/alz.13861] [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: 02/12/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 05/21/2024]
Abstract
INTRODUCTION Sleep disturbances are common in Alzheimer's disease (AD) and may reflect pathologic changes in brain networks. To date, no studies have examined changes in sleep functional connectivity (FC) in AD or their relationship with network hyperexcitability and cognition. METHODS We assessed electroencephalogram (EEG) sleep FC in 33 healthy controls, 36 individuals with AD without epilepsy, and 14 individuals with AD and epilepsy. RESULTS AD participants showed increased gamma connectivity in stage 2 sleep (N2), which was associated with longitudinal cognitive decline. Network hyperexcitability in AD was associated with a distinct sleep connectivity signature, characterized by decreased N2 delta connectivity and reversal of several connectivity changes associated with AD. Machine learning algorithms using sleep connectivity features accurately distinguished diagnostic groups and identified "fast cognitive decliners" among study participants who had AD. DISCUSSION Our findings reveal changes in sleep functional networks associated with cognitive decline in AD and may have implications for disease monitoring and therapeutic development. HIGHLIGHTS Brain functional connectivity (FC) in Alzheimer's disease is altered during sleep. Sleep FC measures correlate with cognitive decline in AD. Network hyperexcitability in AD has a distinct sleep connectivity signature.
Collapse
Affiliation(s)
- Sebastian G. Moguilner
- Department of NeurologyHarvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Courtney Berezuk
- Department of NeurologyHarvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Alex C. Bender
- Department of NeurologyHarvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Kyle R. Pellerin
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Stephen N. Gomperts
- Department of NeurologyHarvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Sydney S. Cash
- Department of NeurologyHarvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Rani A. Sarkis
- Department of NeurologyHarvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Alice D. Lam
- Department of NeurologyHarvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
14
|
Blanpain LT, Cole ER, Chen E, Park JK, Walelign MY, Gross RE, Cabaniss BT, Willie JT, Singer AC. Multisensory flicker modulates widespread brain networks and reduces interictal epileptiform discharges. Nat Commun 2024; 15:3156. [PMID: 38605017 PMCID: PMC11009358 DOI: 10.1038/s41467-024-47263-y] [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: 07/25/2023] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Modulating brain oscillations has strong therapeutic potential. Interventions that both non-invasively modulate deep brain structures and are practical for chronic daily home use are desirable for a variety of therapeutic applications. Repetitive audio-visual stimulation, or sensory flicker, is an accessible approach that modulates hippocampus in mice, but its effects in humans are poorly defined. We therefore quantified the neurophysiological effects of flicker with high spatiotemporal resolution in patients with focal epilepsy who underwent intracranial seizure monitoring. In this interventional trial (NCT04188834) with a cross-over design, subjects underwent different frequencies of flicker stimulation in the same recording session with the effect of sensory flicker exposure on local field potential (LFP) power and interictal epileptiform discharges (IEDs) as primary and secondary outcomes, respectively. Flicker focally modulated local field potentials in expected canonical sensory cortices but also in the medial temporal lobe and prefrontal cortex, likely via resonance of stimulated long-range circuits. Moreover, flicker decreased interictal epileptiform discharges, a pathological biomarker of epilepsy and degenerative diseases, most strongly in regions where potentials were flicker-modulated, especially the visual cortex and medial temporal lobe. This trial met the scientific goal and is now closed. Our findings reveal how multi-sensory stimulation may modulate cortical structures to mitigate pathological activity in humans.
Collapse
Affiliation(s)
- Lou T Blanpain
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
- Neuroscience Graduate Program, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, USA
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Eric R Cole
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Emily Chen
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - James K Park
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael Y Walelign
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Robert E Gross
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
- Departments of Neurosurgery and Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick and New Jersey Medical School, Newark, NJ, USA
| | - Brian T Cabaniss
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jon T Willie
- Departments of Neurological Surgery, Neurology, Psychiatry, and Biomedical Engineering, Washington University, St. Louis, MO, USA.
| | - Annabelle C Singer
- Neuroscience Graduate Program, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, USA.
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA.
| |
Collapse
|
15
|
Wang S, Wu M, Wu S, Lin F, Ji X, Yan J. A polysomnographic study of slow-wave sleep loss in elderly patients with epilepsy. Heliyon 2024; 10:e25904. [PMID: 38379992 PMCID: PMC10877289 DOI: 10.1016/j.heliyon.2024.e25904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 01/02/2024] [Accepted: 02/05/2024] [Indexed: 02/22/2024] Open
Abstract
Objective The primary objective is to explore what causes slow-wave sleep loss in elderly patients with epilepsy. The secondary objective is to identify the PSG characteristics in elderly patients with epilepsy. The clinical demographics, sleep architecture, sleep-related events, and interictal epileptiform discharges are to be evaluated in the objectives. Methods The video electroencephalography (VEEG) and polysomnogram (PSG) data from 44 elderly patients with epilepsy and 52 elderly patients with sleep disorders but without definite central nervous system diseases were analysed. This was a case-control study. The differences in the PSG sleep architecture parameters (total sleep time (TST), sleep efficiency, wake after sleep onset, etc.) and sleep-related events (apnea hypopnea index, oxygen desaturation index (ODI), periodic limb movement index, etc.) between the epilepsy and control groups. As Additionally, these parameters were assessed within the elderly patients with epilepsy, comparing the slow-wave sleep existence and slow-wave sleep loss groups, using VEEG and PSG. Results The epileptic group exhibited significantly lower TST (343.477 ± 96.3046min vs 389.115 ± 61.5727min, p < 0.05), rapid eye movement (%) (13.011 ± 7.5384 vs 16.992 ± 6.7025, p < 0.05), non-rapid eye movement stage 3 (%) (1.35[0,7.225] vs 3.65[0.425,13.75], p < 0.05), and sleep efficiency (%) (69.482 ± 14.1771% vs 77.242 ± 10.6171%, p < 0.05). Conversely, the ODI (25.6[9.825,51.775] events/hour vs 16.85[5.3,30.425] events/hour, p < 0.05) and spontaneous arousal index (4.0455[2.1805,6.9609] events/hour vs 2.9709[1.4747,5.0554] events/hour, p < 0.05) were significantly higher in elderly patients with epilepsy. The prevalence of obstructive sleep apnea-hypopnea syndrome (OSAHS) was significantly higher in the slow-wave sleep loss group than in the slow-wave sleep existence group (100% vs 77.8%, p < 0.05). The incidence of slow-wave sleep loss was lower in patients with epilepsy aged between 75 and 85 years compared to those aged between 65 and 75 years. Conclusion Elderly patients with epilepsy exhibit higher levels of ODI and spontaneous arousal index. Our findings indicate that OSAHS could be a contributing factor to slow-wave sleep loss in this population. The incidence of slow-wave sleep loss was lower in patients aged above 75 years among elderly patients with epilepsy.
Collapse
Affiliation(s)
| | | | - Sangru Wu
- Department of Neurology and Sleep Medical Center, Fujian Provincial Governmental Hospital, Fuzhou, China
| | - Fang Lin
- Department of Neurology and Sleep Medical Center, Fujian Provincial Governmental Hospital, Fuzhou, China
| | - Xiaolin Ji
- Department of Neurology and Sleep Medical Center, Fujian Provincial Governmental Hospital, Fuzhou, China
| | - Jinzhu Yan
- Department of Neurology and Sleep Medical Center, Fujian Provincial Governmental Hospital, Fuzhou, China
| |
Collapse
|
16
|
Sleem T, Decourt B, Sabbagh MN. Nonmedication Devices in Development for the Treatment of Alzheimer's Disease. J Alzheimers Dis Rep 2024; 8:241-255. [PMID: 38405349 PMCID: PMC10894612 DOI: 10.3233/adr-230115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 01/13/2024] [Indexed: 02/27/2024] Open
Abstract
Huge investments continue to be made in treatment for Alzheimer's disease (AD), with more than one hundred drugs currently in development. Pharmacological approaches and drug development, particularly those targeting amyloid-β, have dominated the therapeutic landscape. At the same time, there is also a growing interest in devices for treating AD. This review aimed to identify and describe devices under development for AD treatment. In this review, we queried the devices that are in development for the treatment of AD. PubMed was searched through the end of 2021 using the terms "device," "therapeutics," and "Alzheimer's" for articles that report on devices to treat AD. Ten devices with 31 references were identified as actively being developed for the treatment of AD. Many of these devices are far along in development. Device-based therapies are often overlooked when evaluating treatment approaches to AD. However, many devices for treating AD are in development and some show promising results.
Collapse
Affiliation(s)
- Tamara Sleem
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
| | - Boris Decourt
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX, USA
| | - Marwan N. Sabbagh
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
| |
Collapse
|
17
|
Hsieh JC, He W, Venkatraghavan D, Koptelova VB, Ahmad ZJ, Pyatnitskiy I, Wang W, Jeong J, Tang KKW, Harmeier C, Li C, Rana M, Iyer S, Nayak E, Ding H, Modur P, Mysliwiec V, Schnyer DM, Baird B, Wang H. Design of an injectable, self-adhesive, and highly stable hydrogel electrode for sleep recording. DEVICE 2024; 2:100182. [PMID: 39239460 PMCID: PMC11376683 DOI: 10.1016/j.device.2023.100182] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
High-quality and continuous electroencephalogram (EEG) monitoring is desirable for sleep research, sleep monitoring, and the evaluation and treatment of sleep disorders. Existing continuous EEG monitoring technologies suffer from fragile connections, long-term stability, and complex preparation for electrodes under real-life conditions. Here, we report an injectable and spontaneously cross-linked hydrogel electrode for long-term EEG applications. Specifically, our electrodes have a long-term low impedance on hairy scalp regions of 17.53 kΩ for more than 8 h of recording, high adhesiveness on the skin of 0.92 N cm-1 with repeated attachment capability, and long-term wearability during daily activities and overnight sleep. In addition, our electrodes demonstrate a superior signal-to-noise-ratio of 23.97 decibels (dB) in comparison with commercial wet electrodes of 17.98 dB and share a high agreement of sleep stage classification with commercial wet electrodes during multichannel recording. These results exhibit the potential of our on-site-formed electrodes for high-quality, prolonged EEG monitoring in various scenarios.
Collapse
Affiliation(s)
- Ju-Chun Hsieh
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Weilong He
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Dhivya Venkatraghavan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Victoria B Koptelova
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Zoya J Ahmad
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Ilya Pyatnitskiy
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Wenliang Wang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jinmo Jeong
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Kevin Kai Wing Tang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Cody Harmeier
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Conrad Li
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Manini Rana
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Sruti Iyer
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Eesha Nayak
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Hong Ding
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Pradeep Modur
- Department of Neurology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Vincent Mysliwiec
- Department of Psychiatry, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - David M Schnyer
- Department of Psychology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Benjamin Baird
- Department of Psychology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Huiliang Wang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Lead contact
| |
Collapse
|
18
|
Chen Y, Sun J, Tao J, Sun T. Treatments and regulatory mechanisms of acoustic stimuli on mood disorders and neurological diseases. Front Neurosci 2024; 17:1322486. [PMID: 38249579 PMCID: PMC10796816 DOI: 10.3389/fnins.2023.1322486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024] Open
Abstract
Acoustic stimuli such as music or ambient noise can significantly affect physiological and psychological health in humans. We here summarize positive effects of music therapy in premature infant distress regulation, performance enhancement, sleep quality control, and treatment of mental disorders. Specifically, music therapy exhibits promising effects on treatment of neurological disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD). We also highlight regulatory mechanisms by which auditory intervention affects an organism, encompassing modulation of immune responses, gene expression, neurotransmitter regulation and neural circuitry. As a safe, cost-effective and non-invasive intervention, music therapy offers substantial potential in treating a variety of neurological conditions.
Collapse
Affiliation(s)
- Yikai Chen
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China
| | - Julianne Sun
- Xiamen Institute of Technology Attached School, Xiamen, China
| | - Junxian Tao
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China
| | - Tao Sun
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China
| |
Collapse
|
19
|
Wilckens KA, Habte RF, Dong Y, Stepan ME, Dessa KM, Whitehead AB, Peng CW, Fletcher ME, Buysse DJ. A pilot time-in-bed restriction intervention behaviorally enhances slow-wave activity in older adults. FRONTIERS IN SLEEP 2024; 2:1265006. [PMID: 38938690 PMCID: PMC11210605 DOI: 10.3389/frsle.2023.1265006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Introduction Identifying intervention methods that target sleep characteristics involved in memory processing is a priority for the field of cognitive aging. Older adults with greater sleep efficiency and non-rapid eye movement slow-wave activity (SWA) (0.5-4 Hz electroencephalographic activity) tend to exhibit better memory and cognitive abilities. Paradoxically, long total sleep times are consistently associated with poorer cognition in older adults. Thus, maximizing sleep efficiency and SWA may be a priority relative to increasing mere total sleep time. As clinical behavioral sleep treatments do not consistently enhance SWA, and propensity for SWA increases with time spent awake, we examined with a proof-of concept pilot intervention whether a greater dose of time-in-bed (TiB) restriction (75% of habitual TiB) would increase both sleep efficiency and SWA in older adults with difficulties staying asleep without impairing memory performance. Methods Participants were adults ages 55-80 with diary-reported sleep efficiency <90% and wake after sleep onset (WASO) >20 min. Sleep diary, actigraphy, polysomnography (PSG), and paired associate memory acquisition and retention were assessed before and after a week-long TiB restriction intervention (n = 30). TiB was restricted to 75% of diary-reported habitual TiB. A comparison group of n = 5 participants repeated assessments while following their usual sleep schedule to obtain preliminary estimates of effect sizes associated with repeated testing. Results Subjective and objective sleep measures robustly improved in the TiB restriction group for sleep quality, sleep depth, sleep efficiency and WASO, at the expense of TiB and time spent in N1 and N2 sleep. As hypothesized, SWA increased robustly with TiB restriction across the 0.5-4 Hz range, as well as subjective sleep depth, subjective and objective WASO. Despite increases in sleepiness ratings, no impairments were found in memory acquisition or retention. Conclusion A TiB restriction dose equivalent to 75% of habitual TiB robustly increased sleep continuity and SWA in older adults with sleep maintenance difficulties, without impairing memory performance. These findings may inform long-term behavioral SWA enhancement interventions aimed at improving memory performance and risk for cognitive impairments.
Collapse
Affiliation(s)
| | - Rima F. Habte
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Yue Dong
- School of Nursing, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michelle E. Stepan
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kibra M. Dessa
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Alexis B. Whitehead
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Christine W. Peng
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Mary E. Fletcher
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Daniel J. Buysse
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
20
|
Wei J, Wang M, Guo Y, Liu Y, Dong X. Sleep structure assessed by objective measurement in patients with mild cognitive impairment: A meta-analysis. Sleep Med 2024; 113:397-405. [PMID: 38134714 DOI: 10.1016/j.sleep.2023.12.010] [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: 09/25/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
OBJECTIVES A meta-analysis was used to explore the characteristic changes in objective sleep structure of patients with mild cognitive impairment (MCI) compared with cognitively healthy older adults. MATERIALS AND METHODS PubMed, EMBAS, Cochrane Library, Scopus, and Web of Science were searched until November 2023. A literature quality evaluation was performed according to the Newcastle-Ottawa Scale, and a meta-analysis was performed by RevMan 5.3 software. RESULTS Fifteen studies with 771 participants were finally included. Compared with normal control groups, patients with MCI had a decreased total sleep time by 34.44 min, reduction in sleep efficiency by 7.96 %, increased waking after sleep onset by 19.61 min, and increased sleep latency by 6.97 min. Ten included studies showed that the patients with MCI had increased N1 sleep by 2.72 % and decreased N3 sleep by 0.78 %; however, there was no significant difference between the MCI and control groups in percentage of N2 sleep. Moreover, Twelve included studies reported the MCI groups had shorter REM sleep of 2.69 %. CONCLUSION Our results provide evidence of abnormal sleep architecture in patients with MCI. As a "plastic state," abnormal sleep architecture may be a promising therapeutic target for slowing cognitive decline and dementia prevention.
Collapse
Affiliation(s)
- Jianing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Min Wang
- Department of Nursing, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yuanli Guo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yanjin Liu
- Department of Nursing, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xiaofang Dong
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China.
| |
Collapse
|
21
|
Vickrey B, Lerner I. Overnight exposure to pink noise could jeopardize sleep-dependent insight and pattern detection. Front Hum Neurosci 2023; 17:1302836. [PMID: 38107593 PMCID: PMC10722168 DOI: 10.3389/fnhum.2023.1302836] [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: 09/27/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023] Open
Abstract
Accumulated evidence from the past decades suggests that sleep plays a crucial role in memory consolidation and the facilitation of higher-level cognitive processes such as abstraction and gist extraction. In addition, recent studies show that applying pink noise during sleep can further enhance sleep-dependent memory consolidation, potentially by modulating sleep physiology through stochastic resonance. However, whether this enhancement extends to higher cognitive processes remains untested. In this study, we investigated how the application of open-loop pink noise during sleep influences the gain of insight into hidden patterns. Seventy-two participants were assigned to three groups: daytime-wake, silent sleep, and sleep with pink noise. Each group completed the number reduction task, an established insight paradigm known to be influenced by sleep, over two sessions with a 12-h interval. Sleep groups were monitored by the DREEM 3 headband in home settings. Contrary to our prediction, pink noise did not induce an increase in insight compared to silent sleep and was statistically more similar to the wake condition despite evidence for its typical influence on sleep physiology. Particularly, we found that pink noise limited the time spent in the initial cycle of N1 just after sleep onset, while time spent in N1 positively predicted insight. These results echo recent suggestions that the time in the initial cycle of N1 plays a critical role in insight formation. Overall, our results suggest that open-loop pink noise during sleep may be detrimental to insight formation and creativity due to the alterations it causes to normal sleep architecture.
Collapse
Affiliation(s)
- Beverly Vickrey
- Department of Psychology, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Itamar Lerner
- Department of Psychology, The University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
22
|
Wunderlin M, Zeller CJ, Senti SR, Fehér KD, Suppiger D, Wyss P, Koenig T, Teunissen CE, Nissen C, Klöppel S, Züst MA. Acoustic stimulation during sleep predicts long-lasting increases in memory performance and beneficial amyloid response in older adults. Age Ageing 2023; 52:afad228. [PMID: 38163288 PMCID: PMC10758173 DOI: 10.1093/ageing/afad228] [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: 05/09/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Sleep and neurodegeneration are assumed to be locked in a bi-directional vicious cycle. Improving sleep could break this cycle and help to prevent neurodegeneration. We tested multi-night phase-locked acoustic stimulation (PLAS) during slow wave sleep (SWS) as a non-invasive method to improve SWS, memory performance and plasma amyloid levels. METHODS 32 healthy older adults (agemean: 68.9) completed a between-subject sham-controlled three-night intervention, preceded by a sham-PLAS baseline night. RESULTS PLAS induced increases in sleep-associated spectral-power bands as well as a 24% increase in slow wave-coupled spindles, known to support memory consolidation. There was no significant group-difference in memory performance or amyloid-beta between the intervention and control group. However, the magnitude of PLAS-induced physiological responses were associated with memory performance up to 3 months post intervention and beneficial changes in plasma amyloid. Results were exclusive to the intervention group. DISCUSSION Multi-night PLAS is associated with long-lasting benefits in memory and metabolite clearance in older adults, rendering PLAS a promising tool to build upon and develop long-term protocols for the prevention of cognitive decline.
Collapse
Affiliation(s)
- Marina Wunderlin
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000 Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, 3012 Bern, Switzerland
| | - Céline Jacqueline Zeller
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000 Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, 3012 Bern, Switzerland
| | - Samira Rafaela Senti
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000 Bern, Switzerland
| | - Kristoffer Daniel Fehér
- University Hospital of Psychiatry and Psychotherapy, University of Bern, 3000 Bern, Switzerland
| | - Debora Suppiger
- Department of Neonatology, University Hospital Zurich and University of Zurich, 8006 Zürich, Switzerland
| | - Patric Wyss
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000 Bern, Switzerland
| | - Thomas Koenig
- University Hospital of Psychiatry and Psychotherapy, University of Bern, 3000 Bern, Switzerland
| | - Charlotte Elisabeth Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Neurodegeneration, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, Netherlands
| | - Christoph Nissen
- University Hospital of Psychiatry and Psychotherapy, University of Bern, 3000 Bern, Switzerland
- Division of Psychiatric Specialties, Geneva University Hospitals (HUG), 1205 Geneva, Switzerland
| | - Stefan Klöppel
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000 Bern, Switzerland
| | - Marc Alain Züst
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000 Bern, Switzerland
| |
Collapse
|
23
|
Baxter BS, Mylonas D, Kwok KS, Talbot CE, Patel R, Zhu L, Vangel M, Stickgold R, Manoach DS. The effects of closed-loop auditory stimulation on sleep oscillatory dynamics in relation to motor procedural memory consolidation. Sleep 2023; 46:zsad206. [PMID: 37531587 PMCID: PMC11009689 DOI: 10.1093/sleep/zsad206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/13/2023] [Indexed: 08/04/2023] Open
Abstract
STUDY OBJECTIVES Healthy aging and many disorders show reduced sleep-dependent memory consolidation and corresponding alterations in non-rapid eye movement sleep oscillations. Yet sleep physiology remains a relatively neglected target for improving memory. We evaluated the effects of closed-loop auditory stimulation during sleep (CLASS) on slow oscillations (SOs), sleep spindles, and their coupling, all in relation to motor procedural memory consolidation. METHODS Twenty healthy young adults had two afternoon naps: one with auditory stimulation during SO upstates and another with no stimulation. Twelve returned for a third nap with stimulation at variable times in relation to SO upstates. In all sessions, participants trained on the motor sequence task prior to napping and were tested afterward. RESULTS Relative to epochs with no stimulation, upstate stimuli disrupted sleep and evoked SOs, spindles, and SO-coupled spindles. Stimuli that successfully evoked oscillations were delivered closer to the peak of the SO upstate and when spindle power was lower than stimuli that failed to evoke oscillations. Across conditions, participants showed similar significant post-nap performance improvement that correlated with the density of SO-coupled spindles. CONCLUSIONS Despite its strong effects on sleep physiology, CLASS failed to enhance motor procedural memory. Our findings suggest methods to overcome this failure, including better sound calibration to preserve sleep continuity and the use of real-time predictive algorithms to more precisely target SO upstates and to avoid disrupting endogenous SO-coupled spindles and their mnemonic function. They motivate continued development of CLASS as an intervention to manipulate sleep oscillatory dynamics and improve memory.
Collapse
Affiliation(s)
- Bryan S Baxter
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Dimitrios Mylonas
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Kristi S Kwok
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Christine E Talbot
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rudra Patel
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lin Zhu
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark Vangel
- Department of Biostatistics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert Stickgold
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Dara S Manoach
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| |
Collapse
|
24
|
Bressler S, Neely R, Yost RM, Wang D, Read HL. A wearable EEG system for closed-loop neuromodulation of sleep-related oscillations. J Neural Eng 2023; 20:056030. [PMID: 37726002 DOI: 10.1088/1741-2552/acfb3b] [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: 12/22/2022] [Accepted: 09/19/2023] [Indexed: 09/21/2023]
Abstract
Objective.Healthy sleep plays a critical role in general well-being. Enhancement of slow-wave sleep by targeting acoustic stimuli to particular phases of delta (0.5-2 Hz) waves has shown promise as a non-invasive approach to improve sleep quality. Closed-loop stimulation during other sleep phases targeting oscillations at higher frequencies such as theta (4-7 Hz) or alpha (8-12 Hz) could be another approach to realize additional health benefits. However, systems to track and deliver stimulation relative to the instantaneous phase of electroencephalogram (EEG) signals at these higher frequencies have yet to be demonstrated outside of controlled laboratory settings.Approach.Here we examine the feasibility of using an endpoint-corrected version of the Hilbert transform (ecHT) algorithm implemented on a headband wearable device to measure alpha phase and deliver phase-locked auditory stimulation during the transition from wakefulness to sleep, during which alpha power is greatest. First, the ecHT algorithm is implementedin silicoto evaluate the performance characteristics of this algorithm across a range of sleep-related oscillatory frequencies. Secondly, a pilot sleep study tests feasibility to use the wearable device by users in the home setting for measurement of EEG activity during sleep and delivery of real-time phase-locked stimulation.Main results.The ecHT is capable of computing the instantaneous phase of oscillating signals with high precision, allowing auditory stimulation to be delivered at the intended phases of neural oscillations with low phase error. The wearable system was capable of measuring sleep-related neural activity with sufficient fidelity for sleep stage scoring during the at-home study, and phase-tracking performance matched simulated results. Users were able to successfully operate the system independently using the companion smartphone app to collect data and administer stimulation, and presentation of auditory stimuli during sleep initiation did not negatively impact sleep onset.Significance.This study demonstrates the feasibility of closed-loop real-time tracking and neuromodulation of a range of sleep-related oscillations using a wearable EEG device. Preliminary results suggest that this approach could be used to deliver non-invasive neuromodulation across all phases of sleep.
Collapse
Affiliation(s)
- Scott Bressler
- Elemind Technologies, Inc., Cambridge, MA, United States of America
| | - Ryan Neely
- Elemind Technologies, Inc., Cambridge, MA, United States of America
| | - Ryan M Yost
- Elemind Technologies, Inc., Cambridge, MA, United States of America
| | - David Wang
- Elemind Technologies, Inc., Cambridge, MA, United States of America
| | - Heather L Read
- Department of Psychological Sciences-Behavioral Neuroscience Division, University of Connecticut, Storrs, CT, United States of America
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States of America
- Brain-Computer Interface Core, University of Connecticut, Storrs, CT, United States of America
| |
Collapse
|
25
|
Esfahani MJ, Farboud S, Ngo HVV, Schneider J, Weber FD, Talamini LM, Dresler M. Closed-loop auditory stimulation of sleep slow oscillations: Basic principles and best practices. Neurosci Biobehav Rev 2023; 153:105379. [PMID: 37660843 DOI: 10.1016/j.neubiorev.2023.105379] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023]
Abstract
Sleep is essential for our physical and mental well-being. During sleep, despite the paucity of overt behavior, our brain remains active and exhibits a wide range of coupled brain oscillations. In particular slow oscillations are characteristic for sleep, however whether they are directly involved in the functions of sleep, or are mere epiphenomena, is not yet fully understood. To disentangle the causality of these relationships, experiments utilizing techniques to detect and manipulate sleep oscillations in real-time are essential. In this review, we first overview the theoretical principles of closed-loop auditory stimulation (CLAS) as a method to study the role of slow oscillations in the functions of sleep. We then describe technical guidelines and best practices to perform CLAS and analyze results from such experiments. We further provide an overview of how CLAS has been used to investigate the causal role of slow oscillations in various sleep functions. We close by discussing important caveats, open questions, and potential topics for future research.
Collapse
Affiliation(s)
| | - Soha Farboud
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, the Netherlands
| | - Hong-Viet V Ngo
- Department of Psychology, University of Essex, United Kingdom; Department of Psychology, University of Lübeck, Germany; Center for Brain, Behaviour and Metabolism, University of Lübeck, Germany
| | - Jules Schneider
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Frederik D Weber
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, the Netherlands; Department of Sleep and Cognition, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands
| | - Lucia M Talamini
- Department of Psychology, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, the Netherlands
| | - Martin Dresler
- Donders Institute for Brain, Cognition and Behaviour, Radboudumc, the Netherlands.
| |
Collapse
|
26
|
Fougère M, Greco-Vuilloud J, Arnous C, Abel F, Lowe C, Elie V, Marchand S. Sensory stimulations potentializing digital therapeutics pain control. FRONTIERS IN PAIN RESEARCH 2023; 4:1168377. [PMID: 37745799 PMCID: PMC10511651 DOI: 10.3389/fpain.2023.1168377] [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: 02/17/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
For the past two decades, using Digital Therapeutics (DTx) to counter painful symptoms has emerged as a novel pain relief strategy. Several studies report that DTx significantly diminish pain while compensating for the limitations of pharmacological analgesics (e.g., addiction, side effects). Virtual reality (VR) is a major component of the most effective DTx for pain reduction. Notably, various stimuli (e.g., auditory, visual) appear to be frequently associated with VR in DTx. This review aims to compare the hypoalgesic power of specific stimuli with or without a VR environment. First, this review will briefly describe VR technology and known elements related to its hypoalgesic effect. Second, it will non-exhaustively list various stimuli known to have a hypoalgesic effect on pain independent of the immersive environment. Finally, this review will focus on studies that investigate a possible potentialized effect on pain reduction of these stimuli in a VR environment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Serge Marchand
- Lucine, Bordeaux, France
- Faculté de Médecine et des Sciences de la Santé, Centre de Recherche Clinique du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
27
|
LaGoy AD, Kubala AG, Deering S, Germain A, Markwald RR. Dawn of a New Dawn: Advances in Sleep Health to Optimize Performance. Sleep Med Clin 2023; 18:361-371. [PMID: 37532375 DOI: 10.1016/j.jsmc.2023.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Optimal sleep health is a critical component to high-level performance. In populations such as the military, public service (eg, firefighters), and health care, achieving optimal sleep health is difficult and subsequently deficiencies in sleep health may lead to performance decrements. However, advances in sleep monitoring technologies and mitigation strategies for poor sleep health show promise for further ecological scientific investigation within these populations. The current review briefly outlines the relationship between sleep health and performance as well as current advances in behavioral and technological approaches to improving sleep health for performance.
Collapse
Affiliation(s)
- Alice D LaGoy
- Sleep, Tactical Efficiency, and Endurance Laboratory, Warfighter Performance Department, Naval Health Research Center, 140 Sylvester Road, San Diego, CA 92106, USA; Leidos, Inc., San Diego, CA, USA
| | - Andrew G Kubala
- Sleep, Tactical Efficiency, and Endurance Laboratory, Warfighter Performance Department, Naval Health Research Center, 140 Sylvester Road, San Diego, CA 92106, USA; Leidos, Inc., San Diego, CA, USA
| | - Sean Deering
- Sleep, Tactical Efficiency, and Endurance Laboratory, Warfighter Performance Department, Naval Health Research Center, 140 Sylvester Road, San Diego, CA 92106, USA; Leidos, Inc., San Diego, CA, USA
| | | | - Rachel R Markwald
- Sleep, Tactical Efficiency, and Endurance Laboratory, Warfighter Performance Department, Naval Health Research Center, 140 Sylvester Road, San Diego, CA 92106, USA.
| |
Collapse
|
28
|
Tassone LM, Moyano MD, Laiño F, Brusco LI, Ramele RE, Forcato C. One-week sleep hygiene education improves episodic memory in young but not in older adults during social isolation. Front Psychol 2023; 14:1155776. [PMID: 37599745 PMCID: PMC10433204 DOI: 10.3389/fpsyg.2023.1155776] [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: 01/31/2023] [Accepted: 07/11/2023] [Indexed: 08/22/2023] Open
Abstract
Memory formation is a dynamic process that comprises different phases, such as encoding, consolidation and retrieval. It could be altered by several factors such as sleep quality, anxiety, and depression levels. In the last years, due to COVID-19 pandemic, there was a reduction in sleep quality, an increase in anxiety and depressive symptoms as well as an impairment in emotional episodic memory encoding, especially in young adults. Taking into account the profound impact of sleep quality in daily life a series of rules has been developed that are conducive to consistently achieving good sleep, known as sleep hygiene education. These interventions have been shown to be effective in improving sleep quality and duration and reducing depressive and anxiety symptoms. Here we propose the implementation of a brief sleep hygiene education to improve sleep quality and memory performance as well as to diminish anxiety and depressive scores. For that, participants were divided into two groups: Sleep hygiene education and control group. After that, they were evaluated for anxiety, depression, and sleep quality levels and trained on an episodic memory task. They were tested immediately after (short-term test) and also 1 week later (long-term test). This procedure was also performed before the sleep hygiene education and was taken as baseline level. We found that episodic memory performance for young adults improved for the SHE group after intervention but not for older adults, and no improvements in emotional variables were observed. Despite not observing a significant effect of the intervention for young and older adults regarding the sleep quality scores, we consider that there may be an improvement in sleep physiology that is not subjectively perceived, but would also have a positive impact on memory processes. These results show that even a sleep hygiene education of 1 week could improve cognition in young adults when acute memory and sleep impairment occurs, in this case, due to the isolation by COVID-19 pandemic. However, we suggest that longer interventions should be implemented for older adults who already experience a natural decline in cognitive processes such as episodic memory formation.
Collapse
Affiliation(s)
- Leonela Magali Tassone
- Laboratorio de Sueño y Memoria, Departamento de Ciencias de la Vida, Instituto Tecnológico de Buenos Aires (ITBA), Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| | - Malen Daiana Moyano
- Laboratorio de Sueño y Memoria, Departamento de Ciencias de la Vida, Instituto Tecnológico de Buenos Aires (ITBA), Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| | - Fernando Laiño
- Fundación Instituto Superior de Ciencias de la Salud, Buenos Aires, Argentina
| | - Luis Ignacio Brusco
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
- CENECON, Centro de Neuropsiquiatría y Neurología de la Conducta (CENECON), Buenos Aires, Argentina
| | - Rodrigo Ezequiel Ramele
- Centro de Inteligencia Computacional, Instituto Tecnológico de Buenos Aires (ITBA), Buenos Aires, Argentina
| | - Cecilia Forcato
- Laboratorio de Sueño y Memoria, Departamento de Ciencias de la Vida, Instituto Tecnológico de Buenos Aires (ITBA), Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
29
|
Wei Y, Luo M, Mai X, Feng L, Tang T, Yang D, Krishnan GP, Bazhenov M. The role of age-related sleep EEG changes in memory decline: experiments and computational modeling. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38083499 PMCID: PMC11214839 DOI: 10.1109/embc40787.2023.10340681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
The slow oscillation (SO) observed during deep sleep is known to facilitate memory consolidation. However, the impact of age-related changes in sleep electroencephalography (EEG) oscillations and memory remains unknown. In this study, we aimed to investigate the contribution of age-related changes in sleep SO and its role in memory decline by combining EEG recordings and computational modeling. Based on the detected SO events, we found that older adults exhibit lower SO density, lower SO frequency, and longer Up and Down state durations during N3 sleep compared to young and middle-aged groups. Using a biophysically detailed thalamocortical network model, we simulated the "aged" brain as a partial loss of synaptic connections between neurons in the cortex. Our simulations showed that the changes in sleep SO properties in the "aged" brain, similar to those observed in older adults, resulting in impaired memory consolidation. Overall, this study provides mechanistic insights into how age-related changes modulate sleep SOs and memory decline.Clinical Relevance- This study contributes towards finding feasible biomarkers and target mechanism for designing therapy in older adults with memory deficits, such as Alzheimer's disease patients.
Collapse
|
30
|
Zavecz Z, Shah VD, Murillo OG, Vallat R, Mander BA, Winer JR, Jagust WJ, Walker MP. NREM sleep as a novel protective cognitive reserve factor in the face of Alzheimer's disease pathology. BMC Med 2023; 21:156. [PMID: 37138290 PMCID: PMC10155344 DOI: 10.1186/s12916-023-02811-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/28/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) pathology impairs cognitive function. Yet some individuals with high amounts of AD pathology suffer marked memory impairment, while others with the same degree of pathology burden show little impairment. Why is this? One proposed explanation is cognitive reserve i.e., factors that confer resilience against, or compensation for the effects of AD pathology. Deep NREM slow wave sleep (SWS) is recognized to enhance functions of learning and memory in healthy older adults. However, that the quality of NREM SWS (NREM slow wave activity, SWA) represents a novel cognitive reserve factor in older adults with AD pathology, thereby providing compensation against memory dysfunction otherwise caused by high AD pathology burden, remains unknown. METHODS Here, we tested this hypothesis in cognitively normal older adults (N = 62) by combining 11C-PiB (Pittsburgh compound B) positron emission tomography (PET) scanning for the quantification of β-amyloid (Aβ) with sleep electroencephalography (EEG) recordings to quantify NREM SWA and a hippocampal-dependent face-name learning task. RESULTS We demonstrated that NREM SWA significantly moderates the effect of Aβ status on memory function. Specifically, NREM SWA selectively supported superior memory function in individuals suffering high Aβ burden, i.e., those most in need of cognitive reserve (B = 2.694, p = 0.019). In contrast, those without significant Aβ pathological burden, and thus without the same need for cognitive reserve, did not similarly benefit from the presence of NREM SWA (B = -0.115, p = 0.876). This interaction between NREM SWA and Aβ status predicting memory function was significant after correcting for age, sex, Body Mass Index, gray matter atrophy, and previously identified cognitive reserve factors, such as education and physical activity (p = 0.042). CONCLUSIONS These findings indicate that NREM SWA is a novel cognitive reserve factor providing resilience against the memory impairment otherwise caused by high AD pathology burden. Furthermore, this cognitive reserve function of NREM SWA remained significant when accounting both for covariates, and factors previously linked to resilience, suggesting that sleep might be an independent cognitive reserve resource. Beyond such mechanistic insights are potential therapeutic implications. Unlike many other cognitive reserve factors (e.g., years of education, prior job complexity), sleep is a modifiable factor. As such, it represents an intervention possibility that may aid the preservation of cognitive function in the face of AD pathology, both present moment and longitudinally.
Collapse
Affiliation(s)
- Zsófia Zavecz
- Department of Psychology, Center for Human Sleep Science, University of California Berkeley, Berkeley, CA, 94720, USA.
| | - Vyoma D Shah
- Department of Psychology, Center for Human Sleep Science, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Olivia G Murillo
- Department of Psychology, Center for Human Sleep Science, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Raphael Vallat
- Department of Psychology, Center for Human Sleep Science, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Bryce A Mander
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, 92617, USA
| | - Joseph R Winer
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94304, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Matthew P Walker
- Department of Psychology, Center for Human Sleep Science, University of California Berkeley, Berkeley, CA, 94720, USA.
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA.
| |
Collapse
|
31
|
Blanpain LT, Chen E, Park J, Walelign MY, Gross RE, Cabaniss BT, Willie JT, Singer AC. Multisensory Flicker Modulates Widespread Brain Networks and Reduces Interictal Epileptiform Discharges in Humans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.14.23286691. [PMID: 36993248 PMCID: PMC10055448 DOI: 10.1101/2023.03.14.23286691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Modulating brain oscillations has strong therapeutic potential. However, commonly used non-invasive interventions such as transcranial magnetic or direct current stimulation have limited effects on deeper cortical structures like the medial temporal lobe. Repetitive audio-visual stimulation, or sensory flicker, modulates such structures in mice but little is known about its effects in humans. Using high spatiotemporal resolution, we mapped and quantified the neurophysiological effects of sensory flicker in human subjects undergoing presurgical intracranial seizure monitoring. We found that flicker modulates both local field potential and single neurons in higher cognitive regions, including the medial temporal lobe and prefrontal cortex, and that local field potential modulation is likely mediated via resonance of involved circuits. We then assessed how flicker affects pathological neural activity, specifically interictal epileptiform discharges, a biomarker of epilepsy also implicated in Alzheimer's and other diseases. In our patient population with focal seizure onsets, sensory flicker decreased the rate interictal epileptiform discharges. Our findings support the use of sensory flicker to modulate deeper cortical structures and mitigate pathological activity in humans.
Collapse
Affiliation(s)
- Lou T. Blanpain
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
- Neuroscience Graduate Program, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, USA
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Emily. Chen
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - James Park
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael Y. Walelign
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Robert E. Gross
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Brian T. Cabaniss
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jon T. Willie
- Department of Neurosurgery, Washington University, St. Louis, MO, USA
| | - Annabelle C. Singer
- Neuroscience Graduate Program, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, USA
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| |
Collapse
|
32
|
Simor P, Bogdány T, Sifuentes-Ortega R, Rovai A, Peigneux P. Lateralized tactile stimulation during NREM sleep globally increases both slow and fast frequency activities. Psychophysiology 2023; 60:e14191. [PMID: 36153813 PMCID: PMC10078489 DOI: 10.1111/psyp.14191] [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: 11/22/2021] [Revised: 07/06/2022] [Accepted: 09/06/2022] [Indexed: 01/25/2023]
Abstract
Slow frequency activity during non-rapid eye movement (NREM) sleep emerges from synchronized activity of widely distributed thalamo-cortical and cortico-cortical networks, reflecting homeostatic and restorative properties of sleep. Slow frequency activity exhibits a reactive nature, and can be increased by acoustic stimulation. Although non-invasive brain stimulation is a promising technique in basic and clinical sleep research, sensory stimulation studies focusing on modalities other than the acoustic are scarce. We explored here the potential of lateralized vibro-tactile stimulation (VTS) of the finger to locally modify electroencephalographic activity during nocturnal NREM sleep. Eight seconds-long sequences of vibro-tactile pulses were delivered at a rate of 1 Hz either to the left or to the right index finger, in addition to a sham condition, in fourteen healthy participants. VTS markedly increased slow frequency activity that peaked between 1-4 Hz but extended to higher (~13 Hz) frequencies, with fronto-central dominance. Enhanced slow frequency activity was accompanied by increased (14-22 Hz) fast frequency power peaking over central and posterior locations. VTS increased the amplitude of slow waves, especially during the first 3-4 s of stimulation. Noticeably, we did not observe local-hemispheric effects, that is, VTS resulted in a global cortical response regardless of stimulation laterality. VTS moderately increased slow and fast frequency activities in resting wakefulness, to a much lower extent compared to NREM sleep. The concomitant increase in slow and fast frequency activities in response to VTS indicates an instant homeostatic response coupled with wake-like, high-frequency activity potentially reflecting transient periods of increased environmental processing.
Collapse
Affiliation(s)
- Péter Simor
- Institute of Psychology, ELTE Eötvös Loránd University, Budapest, Hungary.,UR2NF, Neuropsychology and Functional Neuroimaging Research Unit at CRCN-Center for Research in Cognition and Neurosciences, Brussels, Belgium.,UNI-ULB Neurosciences Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Tamás Bogdány
- Institute of Psychology, ELTE Eötvös Loránd University, Budapest, Hungary.,UR2NF, Neuropsychology and Functional Neuroimaging Research Unit at CRCN-Center for Research in Cognition and Neurosciences, Brussels, Belgium.,Doctoral School of Psychology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Rebeca Sifuentes-Ortega
- UR2NF, Neuropsychology and Functional Neuroimaging Research Unit at CRCN-Center for Research in Cognition and Neurosciences, Brussels, Belgium.,UNI-ULB Neurosciences Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Antonin Rovai
- UNI-ULB Neurosciences Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Laboratoire de Cartographie fonctionnelle du Cerveau (LCFC), ULB Neuroscience Institute (UNI), CUB-Hôpital Erasme, Université libre de Bruxelles (ULB), Brussels, Belgium.,Department of Functional Neuroimaging, Service of Nuclear Medicine, CUB-Hôpital Erasme, Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Philippe Peigneux
- UR2NF, Neuropsychology and Functional Neuroimaging Research Unit at CRCN-Center for Research in Cognition and Neurosciences, Brussels, Belgium.,UNI-ULB Neurosciences Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
33
|
Guttesen AÁV, Gaskell MG, Madden EV, Appleby G, Cross ZR, Cairney SA. Sleep loss disrupts the neural signature of successful learning. Cereb Cortex 2023; 33:1610-1625. [PMID: 35470400 PMCID: PMC9977378 DOI: 10.1093/cercor/bhac159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 11/13/2022] Open
Abstract
Sleep supports memory consolidation as well as next-day learning. The influential "Active Systems" account of offline consolidation suggests that sleep-associated memory processing paves the way for new learning, but empirical evidence in support of this idea is scarce. Using a within-subjects (n = 30), crossover design, we assessed behavioral and electrophysiological indices of episodic encoding after a night of sleep or total sleep deprivation in healthy adults (aged 18-25 years) and investigated whether behavioral performance was predicted by the overnight consolidation of episodic associations from the previous day. Sleep supported memory consolidation and next-day learning as compared to sleep deprivation. However, the magnitude of this sleep-associated consolidation benefit did not significantly predict the ability to form novel memories after sleep. Interestingly, sleep deprivation prompted a qualitative change in the neural signature of encoding: Whereas 12-20 Hz beta desynchronization-an established marker of successful encoding-was observed after sleep, sleep deprivation disrupted beta desynchrony during successful learning. Taken together, these findings suggest that effective learning depends on sleep but not necessarily on sleep-associated consolidation.
Collapse
Affiliation(s)
- Anna á V Guttesen
- Department of Psychology, University of York, Heslington, York, YO10 5DD, UK
| | - M Gareth Gaskell
- Department of Psychology, University of York, Heslington, York, YO10 5DD, UK
- York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, UK
| | - Emily V Madden
- Department of Psychology, University of York, Heslington, York, YO10 5DD, UK
| | - Gabrielle Appleby
- Department of Psychology, University of York, Heslington, York, YO10 5DD, UK
| | - Zachariah R Cross
- Cognitive Neuroscience Laboratory, Australian Research Centre for Interactive and Virtual Environments, Mawson Lakes Campus, Mawson Lakes, South Australia 5095, Australia
| | - Scott A Cairney
- Department of Psychology, University of York, Heslington, York, YO10 5DD, UK
- York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, UK
| |
Collapse
|
34
|
Zeller CJ, Züst MA, Wunderlin M, Nissen C, Klöppel S. The promise of portable remote auditory stimulation tools to enhance slow-wave sleep and prevent cognitive decline. J Sleep Res 2023:e13818. [PMID: 36631001 DOI: 10.1111/jsr.13818] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 01/13/2023]
Abstract
Dementia is the seventh leading cause of mortality, and a major source of disability and dependency in older individuals globally. Cognitive decline (and, to a lesser extent, normal ageing) are associated with sleep fragmentation and loss of slow-wave sleep. Evidence suggests a bidirectional causal link between these losses. Phase-locked auditory stimulation has emerged as a promising non-invasive tool to enhance slow-wave sleep, potentially ameliorating cognitive decline. In laboratory settings, auditory stimulation is usually supervised by trained experts. Different algorithms (simple amplitude thresholds, topographic correlation, sine-wave fitting, phase-locked loop, and phase vocoder) are used to precisely target auditory stimulation to a desired phase of the slow wave. While all algorithms work well in younger adults, the altered sleep physiology of older adults and particularly those with neurodegenerative disorders requires a tailored approach that can adapt to older adults' fragmented sleep and reduced amplitudes of slow waves. Moreover, older adults might require a continuous intervention that is not feasible in laboratory settings. Recently, several auditory stimulation-capable portable devices ('Dreem®', 'SmartSleep®' and 'SleepLoop®') have been developed. We discuss these three devices regarding their potential as tools for science, and as clinical remote-intervention tools to combat cognitive decline. Currently, SleepLoop® shows the most promise for scientific research in older adults due to high transparency and customizability but is not commercially available. Studies evaluating down-stream effects on cognitive abilities, especially in patient populations, are required before a portable auditory stimulation device can be recommended as a clinical preventative remote-intervention tool.
Collapse
Affiliation(s)
- Céline J Zeller
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Marc A Züst
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Marina Wunderlin
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Christoph Nissen
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland.,Division of Psychiatric Specialties, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Stefan Klöppel
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| |
Collapse
|
35
|
Jung H, Lee Y, Lee SH, Sohn JH. Auditory or Audiovisual Stimulation Ameliorates Cognitive Impairment and Neuropathology in ApoE4 Knock-In Mice. Int J Mol Sci 2023; 24:ijms24020938. [PMID: 36674449 PMCID: PMC9863367 DOI: 10.3390/ijms24020938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
We hypothesized that auditory stimulation could reduce the progression of Alzheimer’s disease (AD), and that audiovisual stimulation could have additional effects through multisensory integration. We exposed 12 month old Apoetm1.1(APOE*4)Adiuj mice (a mouse model of sporadic AD) to auditory (A) or audiovisual stimulation (AV) at 40 Hz for 14 days in a soundproof chamber system (no stimulation, N). Behavioral tests were performed before and after each session, and their brain tissues were assessed for amyloid-beta expression and apoptotic cell death, after 14 days. Furthermore, brain levels of acetylcholine and apoptosis-related proteins were analyzed. In the Y-maze test, the percentage relative alternation was significantly higher in group A than in group N mice. Amyloid-beta and TUNEL positivity in the hippocampal CA3 region was significantly lower in group A and group AV mice than in group N mice (p < 0.05). Acetylcholine levels were significantly higher in group A and group AV mice than in group N mice (p < 0.05). Compared to group N mice, expression of the proapoptotic proteins Bax and caspase-3 was lower in group A, and expression of the antiapoptotic protein Bcl-2 was higher in group AV. In a mouse model of early-stage sporadic AD, auditory or audiovisual stimulation improved cognitive performance and neuropathology.
Collapse
Affiliation(s)
- Harry Jung
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Yeonkyeong Lee
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Sang-Hwa Lee
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| | - Jong-Hee Sohn
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
- Correspondence:
| |
Collapse
|
36
|
Owen NE, Barker RA, Voysey ZJ. Sleep Dysfunction in Huntington's Disease: Impacts of Current Medications and Prospects for Treatment. J Huntingtons Dis 2023; 12:149-161. [PMID: 37248911 PMCID: PMC10473096 DOI: 10.3233/jhd-230567] [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] [Accepted: 05/06/2023] [Indexed: 05/31/2023]
Abstract
Sleep dysfunction is highly prevalent in Huntington's disease (HD). Increasing evidence suggests that such dysfunction not only impairs quality of life and exacerbates symptoms but may even accelerate the underlying disease process. Despite this, current HD treatment approaches neither consider the impact of commonly used medications on sleep, nor directly tackle sleep dysfunction. In this review, we discuss approaches to these two areas, evaluating not only literature from clinical studies in HD, but also that from parallel neurodegenerative conditions and preclinical models of HD. We conclude by summarizing a hierarchical framework of current medications with regard to their impact on sleep, and by outlining key emerging sleep therapies.
Collapse
Affiliation(s)
- Natalia E. Owen
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Roger A. Barker
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
- Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Zanna J. Voysey
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| |
Collapse
|
37
|
Fitzgerald ES, Stout JC, Glikmann-Johnston Y, Anderson C, Jackson ML. Sleep, Circadian Rhythms, and Cognitive Dysfunction in Huntington's Disease. J Huntingtons Dis 2023; 12:293-304. [PMID: 37599535 DOI: 10.3233/jhd-230578] [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: 08/22/2023]
Abstract
BACKGROUND In healthy people, sleep and circadian disruption are linked to cognitive deficits. People with Huntington's disease (HD), who have compromised brain function and sleep and circadian disturbances, may be even more susceptible to these cognitive effects. OBJECTIVE To conduct a comprehensive review and synthesis of the literature in HD on the associations of cognitive dysfunction with disturbed sleep and circadian rhythms. METHODS We searched MEDLINE via OVID, CINAHL Plus, EMBASE via OVID, and PubMed in May 2023. The first author then screened by title and abstract and conducted a full review of remaining articles. RESULTS Eight studies investigating the influence of sleep and/or circadian rhythms on cognitive function in HD were found. In manifest HD, poorer sleep was associated with worse cognitive function. For behavioral 24-hour (circadian) rhythms, two studies indicated that later wake times correlated with poorer cognitive function. No reported studies in HD examined altered physiological 24-hour (circadian) rhythms and cognitive impairment. CONCLUSION Some associations exist between poor sleep and cognitive dysfunction in manifest HD, yet whether these associations are present before clinical diagnosis is unknown. Whether circadian disturbances relate to cognitive impairment in HD also remains undetermined. To inform sleep and circadian interventions aimed at improving cognitive symptoms in HD, future research should include a range of disease stages, control for external factors, and utilize robust cognitive batteries targeted to the aspects of cognitive function known to be adversely affected in HD.
Collapse
Affiliation(s)
- Emily S Fitzgerald
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Julie C Stout
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Yifat Glikmann-Johnston
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Clare Anderson
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Melinda L Jackson
- School of Psychological Sciences, and Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| |
Collapse
|
38
|
Wilson DA, Fleming G, Williams CRO, Teixeira CM, Smiley JF, Saito M. Somatostatin neuron contributions to cortical slow wave dysfunction in adult mice exposed to developmental ethanol. Front Neurosci 2023; 17:1127711. [PMID: 37021136 PMCID: PMC10067632 DOI: 10.3389/fnins.2023.1127711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/06/2023] [Indexed: 04/07/2023] Open
Abstract
Introduction Transitions between sleep and waking and sleep-dependent cortical oscillations are heavily dependent on GABAergic neurons. Importantly, GABAergic neurons are especially sensitive to developmental ethanol exposure, suggesting a potential unique vulnerability of sleep circuits to early ethanol. In fact, developmental ethanol exposure can produce long-lasting impairments in sleep, including increased sleep fragmentation and decreased delta wave amplitude. Here, we assessed the efficacy of optogenetic manipulations of somatostatin (SST) GABAergic neurons in the neocortex of adult mice exposed to saline or ethanol on P7, to modulate cortical slow-wave physiology. Methods SST-cre × Ai32 mice, which selectively express channel rhodopsin in SST neurons, were exposed to ethanol or saline on P7. This line expressed similar developmental ethanol induced loss of SST cortical neurons and sleep impairments as C57BL/6By mice. As adults, optical fibers were implanted targeting the prefrontal cortex (PFC) and telemetry electrodes were implanted in the neocortex to monitor slow-wave activity and sleep-wake states. Results Optical stimulation of PFC SST neurons evoked slow-wave potentials and long-latency single-unit excitation in saline treated mice but not in ethanol mice. Closed-loop optogenetic stimulation of PFC SST neuron activation on spontaneous slow-waves enhanced cortical delta oscillations, and this manipulation was more effective in saline mice than P7 ethanol mice. Discussion Together, these results suggest that SST cortical neurons may contribute to slow-wave impairment after developmental ethanol.
Collapse
Affiliation(s)
- Donald A Wilson
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Child and Adolescent Psychiatry, New York University School of Medicine, New York, NY, United States
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, United States
| | - G Fleming
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - C R O Williams
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - C M Teixeira
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Child and Adolescent Psychiatry, New York University School of Medicine, New York, NY, United States
| | - J F Smiley
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States
| | - Mariko Saito
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
39
|
Ruch S, Schmidig FJ, Knüsel L, Henke K. Closed-loop modulation of local slow oscillations in human NREM sleep. Neuroimage 2022; 264:119682. [PMID: 36240988 DOI: 10.1016/j.neuroimage.2022.119682] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022] Open
Abstract
Slow-wave sleep is the deep non-rapid eye-movement (NREM) sleep stage that is most relevant for the recuperative function of sleep. Its defining property is the presence of slow oscillations (<2 Hz) in the scalp electroencephalogram (EEG). Slow oscillations are generated by a synchronous back and forth between highly active UP-states and silent DOWN-states in neocortical neurons. Growing evidence suggests that closed-loop sensory stimulation targeted at UP-states of EEG-defined slow oscillations can enhance the slow oscillatory activity, increase sleep depth, and boost sleep's recuperative functions. However, several studies failed to replicate such findings. Failed replications might be due to the use of conventional closed-loop stimulation algorithms that analyze the signal from one single electrode and thereby neglect the fact that slow oscillations vary with respect to their origins, distributions, and trajectories on the scalp. In particular, conventional algorithms nonspecifically target functionally heterogeneous UP-states of distinct origins. After all, slow oscillations at distinct sites of the scalp have been associated with distinct functions. Here we present a novel EEG-based closed-loop stimulation algorithm that allows targeting UP- and DOWN-states of distinct cerebral origins based on topographic analyses of the EEG: the topographic targeting of slow oscillations (TOPOSO) algorithm. We present evidence that the TOPOSO algorithm can detect and target local slow oscillations with specific, predefined voltage maps on the scalp in real-time. When compared to a more conventional, single-channel-based approach, TOPOSO leads to fewer but locally more specific stimulations in a simulation study. In a validation study with napping participants, TOPOSO targets auditory stimulation reliably at local UP-states over frontal, sensorimotor, and centro-parietal regions. Importantly, auditory stimulation temporarily enhanced the targeted local state. However, stimulation then elicited a standard frontal slow oscillation rather than local slow oscillations. The TOPOSO algorithm is suitable for the modulation and the study of the functions of local slow oscillations.
Collapse
Affiliation(s)
- Simon Ruch
- Institute for Neuromodulation and Neurotechnology, Department of Neurosurgery and Neurotechnology, University Hospital and University of Tuebingen, Otfried-Müller-Str. 45, Tübingen 72076, Germany; Cognitive Neuroscience of Memory and Consciousness, Institute of Psychology, University of Bern, Fabrikstrasse 8, 3012 Bern, Switzerland.
| | - Flavio Jean Schmidig
- Cognitive Neuroscience of Memory and Consciousness, Institute of Psychology, University of Bern, Fabrikstrasse 8, 3012 Bern, Switzerland
| | - Leona Knüsel
- Cognitive Neuroscience of Memory and Consciousness, Institute of Psychology, University of Bern, Fabrikstrasse 8, 3012 Bern, Switzerland
| | - Katharina Henke
- Cognitive Neuroscience of Memory and Consciousness, Institute of Psychology, University of Bern, Fabrikstrasse 8, 3012 Bern, Switzerland
| |
Collapse
|
40
|
Debellemanière E, Pinaud C, Schneider J, Arnal PJ, Casson AJ, Chennaoui M, Galtier M, Navarrete M, Lewis PA. Optimising sounds for the driving of sleep oscillations by closed-loop auditory stimulation. J Sleep Res 2022; 31:e13676. [PMID: 35762085 PMCID: PMC9788124 DOI: 10.1111/jsr.13676] [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: 10/02/2021] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 12/30/2022]
Abstract
Recent studies have shown that slow oscillations (SOs) can be driven by rhythmic auditory stimulation, which deepens slow-wave sleep (SWS) and improves memory and the immune-supportive hormonal milieu related to this sleep stage. While different attempts have been made to optimise the driving of the SOs by changing the number of click stimulations, no study has yet investigated the impact of applying more than five clicks in a row. Likewise, the importance of the type of sounds in eliciting brain responses is presently unclear. In a study of 12 healthy young participants (10 females; aged 18-26 years), we applied an established closed-loop stimulation method, which delivered sequences of 10 pink noises, 10 pure sounds (B note of 247 Hz), 10 pronounced "a" vowels, 10 sham, 10 variable sounds, and 10 "oddball" sounds on the up phase of the endogenous SOs. By analysing area under the curve, amplitude, and event related potentials, we explored whether the nature of the sound had a differential effect on driving SOs. We showed that every stimulus in a 10-click sequence, induces a SO response. Interestingly, all three types of sounds that we tested triggered SOs. However, pink noise elicited a more pronounced response compared to the other sounds, which was explained by a broader topographical recruitment of brain areas. Our data further suggest that varying the sounds may partially counteract habituation.
Collapse
Affiliation(s)
- Eden Debellemanière
- Unité Fatigue et VigilanceInstitut de recherche biomédicale des armées (IRBA)Brétigny sur OrgeFrance,EA7330 VIFASOM, Hôtel DieuUniversité Paris 5 DescartesParisFrance,Research Team, DreemParisFrance
| | | | - Jules Schneider
- School of Biological Sciencesthe University of ManchesterManchesterUK,CUBRIC, Psychology DepartmentCardiff UniversityCardiffUK
| | | | - Alexander J. Casson
- School of Electrical and Electronic Engineeringthe University of ManchesterManchesterUK
| | - Mounir Chennaoui
- Unité Fatigue et VigilanceInstitut de recherche biomédicale des armées (IRBA)Brétigny sur OrgeFrance,EA7330 VIFASOM, Hôtel DieuUniversité Paris 5 DescartesParisFrance
| | | | - Miguel Navarrete
- School of Biomedical EngineeringUniversity of Los AndesBogotáColombia
| | | |
Collapse
|
41
|
Moreira CG, Hofmann P, Müllner A, Baumann CR, Ginde VR, Kollarik S, Morawska MM, Noain D. Down-phase auditory stimulation is not able to counteract pharmacologically or physiologically increased sleep depth in traumatic brain injury rats. J Sleep Res 2022; 31:e13615. [PMID: 35474362 PMCID: PMC9786351 DOI: 10.1111/jsr.13615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/30/2022] [Accepted: 04/05/2022] [Indexed: 12/30/2022]
Abstract
Modulation of slow-wave activity, either via pharmacological sleep induction by administering sodium oxybate or sleep restriction followed by a strong dissipation of sleep pressure, has been associated with preserved posttraumatic cognition and reduced diffuse axonal injury in traumatic brain injury rats. Although these classical strategies provided promising preclinical results, they lacked the specificity and/or translatability needed to move forward into clinical applications. Therefore, we recently developed and implemented a rodent auditory stimulation method that is a scalable, less invasive and clinically meaningful approach to modulate slow-wave activity by targeting a particular phase of slow waves. Here, we assessed the feasibility of down-phase targeted auditory stimulation of slow waves and evaluated its comparative modulatory strength in relation to the previously employed slow-wave activity modulators in our rat model of traumatic brain injury. Our results indicate that, in spite of effectively reducing slow-wave activity in both healthy and traumatic brain injury rats via down-phase targeted stimulation, this method was not sufficiently strong to counteract the boost in slow-wave activity associated with classical modulators, nor to alter concomitant posttraumatic outcomes. Therefore, the usefulness and effectiveness of auditory stimulation as potential standalone therapeutic strategy in the context of traumatic brain injury warrants further exploration.
Collapse
Affiliation(s)
- Carlos G. Moreira
- Department of NeurologyUniversity Hospital Zurich, University of ZurichZurichSwitzerland
| | - Pascal Hofmann
- Department of NeurologyUniversity Hospital Zurich, University of ZurichZurichSwitzerland
| | - Adrian Müllner
- Department of NeurologyUniversity Hospital Zurich, University of ZurichZurichSwitzerland
| | - Christian R. Baumann
- Department of NeurologyUniversity Hospital Zurich, University of ZurichZurichSwitzerland,University Center of Competence Sleep & Health Zurich (CRPP)University of ZurichZurichSwitzerland,Neuroscience Center Zurich (ZNZ)ZurichSwitzerland
| | - Varun R. Ginde
- Department of NeurologyUniversity Hospital Zurich, University of ZurichZurichSwitzerland
| | - Sedef Kollarik
- Department of NeurologyUniversity Hospital Zurich, University of ZurichZurichSwitzerland
| | - Marta M. Morawska
- Department of NeurologyUniversity Hospital Zurich, University of ZurichZurichSwitzerland
| | - Daniela Noain
- Department of NeurologyUniversity Hospital Zurich, University of ZurichZurichSwitzerland,University Center of Competence Sleep & Health Zurich (CRPP)University of ZurichZurichSwitzerland,Neuroscience Center Zurich (ZNZ)ZurichSwitzerland
| |
Collapse
|
42
|
Wunderlin M, Koenig T, Zeller C, Nissen C, Züst MA. Automatized online prediction of slow-wave peaks during non-rapid eye movement sleep in young and old individuals: Why we should not always rely on amplitude thresholds. J Sleep Res 2022; 31:e13584. [PMID: 35274389 PMCID: PMC9787564 DOI: 10.1111/jsr.13584] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/02/2022] [Accepted: 02/25/2022] [Indexed: 12/30/2022]
Abstract
Brain-state-dependent stimulation during slow-wave sleep is a promising tool for the treatment of psychiatric and neurodegenerative diseases. A widely used slow-wave prediction algorithm required for brain-state-dependent stimulation is based on a specific amplitude threshold in the electroencephalogram. However, due to decreased slow-wave amplitudes in aging and psychiatric conditions, this approach might miss many slow-waves because they do not fulfill the amplitude criterion. Here, we compared slow-wave peaks predicted via an amplitude-based versus a multidimensional approach using a topographical template of slow-wave peaks in 21 young and 21 older healthy adults. We validate predictions against the gold-standard of offline detected peaks. Multidimensionally predicted peaks resemble the gold-standard regarding spatiotemporal dynamics but exhibit lower peak amplitudes. Amplitude-based prediction, by contrast, is less sensitive, less precise and - especially in the older group - predicts peaks that differ from the gold-standard regarding spatiotemporal dynamics. Our results suggest that amplitude-based slow-wave peak prediction might not always be the ideal choice. This is particularly the case in populations with reduced slow-wave amplitudes, like older adults or psychiatric patients. We recommend the use of multidimensional prediction, especially in studies targeted at populations other than young and healthy individuals.
Collapse
Affiliation(s)
- Marina Wunderlin
- University Hospital of Old Age Psychiatry and PsychotherapyUniversity of BernBernSwitzerland
| | - Thomas Koenig
- University Hospital of Psychiatry and PsychotherapyUniversity of BernBernSwitzerland,Interfaculty Research Cooperation ‐ Decoding SleepUniversity of BernBernSwitzerland
| | - Céline Zeller
- University Hospital of Old Age Psychiatry and PsychotherapyUniversity of BernBernSwitzerland
| | - Christoph Nissen
- University Hospital of Psychiatry and PsychotherapyUniversity of BernBernSwitzerland,Interfaculty Research Cooperation ‐ Decoding SleepUniversity of BernBernSwitzerland
| | - Marc Alain Züst
- University Hospital of Old Age Psychiatry and PsychotherapyUniversity of BernBernSwitzerland
| |
Collapse
|
43
|
Liu S, Lei Q, Liu Y, Zhang X, Li Z. Acoustic Stimulation Improves Memory and Reverses the Contribution of Chronic Sleep Deprivation to Pathology in 3xTgAD Mice. Brain Sci 2022; 12:1509. [PMID: 36358435 PMCID: PMC9688064 DOI: 10.3390/brainsci12111509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/02/2022] [Accepted: 11/02/2022] [Indexed: 07/30/2023] Open
Abstract
Acoustic stimulation during sleep is believed to enhance slow waves, which are critical to memory consolidation. However, clinical trials of acoustic stimulation have yielded mixed results concerning its effectiveness in improving human memory. A few studies have implied that acoustic stimulation ameliorates the pathology of Alzheimer's disease (AD) in mice with normal sleep. Here, we explored the effect of acoustic stimulation on 3xTgAD mice suffering from chronic sleep deprivation, as these data may shed light on the potential use of acoustic stimulation in AD patients with insomnia. Methods: Twenty-four 8-month-old 3xTgAD mice were randomly and equally divided into three groups: the normal sleep group (S group), the sleep deprivation group (SD group), and the acoustic stimulation group (AS group). During a 14-day sleep intervention, the SD and AS groups received 6 h of sleep deprivation per day, and the AS group also received acoustic stimulation in the dark phase. Then, the mice underwent Morris water maze (MWM) tests and arterial spin labelling (ASL) magnetic resonance imaging (MRI) scans and were sacrificed for pathological evaluation. Results: The three groups showed similar stress levels. The S and AS groups exhibited better spatial memory, better brain perfusion, and milder amyloid β (Aβ) and tau pathology than the SD group, although no significant discrepancies were found between the S and AS groups. Conclusion: Acoustic stimulation may exert a protective effect in 3xTgAD mice by improving spatial memory, enhancing the blood supply of the brain, and reversing the contribution of chronic sleep deprivation to Aβ and tau pathology to mimic the effect of normal sleep patterns.
Collapse
Affiliation(s)
- Shunjie Liu
- Department of Neurology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Qingfeng Lei
- Department of Neurology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Yunyun Liu
- Department of Neurology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Xiaofeng Zhang
- Department of Neurology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Zhong Li
- Department of Neurology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Shenzhen Research Institute, Sun Yat-sen University, Shenzhen 518000, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou 510080, China
| |
Collapse
|
44
|
Ngo HVV, Staresina BP. Shaping overnight consolidation via slow-oscillation closed-loop targeted memory reactivation. Proc Natl Acad Sci U S A 2022; 119:e2123428119. [PMID: 36279449 PMCID: PMC9636934 DOI: 10.1073/pnas.2123428119] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 05/19/2022] [Indexed: 11/23/2022] Open
Abstract
Sleep constitutes a privileged state for new memories to reactivate and consolidate. Previous work has demonstrated that consolidation can be bolstered experimentally either via delivery of reminder cues (targeted memory reactivation [TMR]) or via noninvasive brain stimulation geared toward enhancing endogenous sleep rhythms. Here, we combined both approaches, controlling the timing of TMR cues with respect to ongoing slow-oscillation (SO) phases. Prior to sleep, participants learned associations between unique words and a set of repeating images (e.g., car) while hearing a prototypical image sound (e.g., engine starting). Memory performance on an immediate test vs. a test the next morning quantified overnight memory consolidation. Importantly, two image sounds were designated as TMR cues, with one cue delivered at SO UP states and the other delivered at SO DOWN states. A novel sound was used as a TMR control condition. Behavioral results revealed a significant reduction of overnight forgetting for words associated with UP-state TMR compared with words associated with DOWN-state TMR. Electrophysiological results showed that UP-state cueing led to enhancement of the ongoing UP state and was followed by greater spindle power than DOWN-state cueing. Moreover, UP-state (and not DOWN-state) cueing led to reinstatement of target image representations. Together, these results unveil the behavioral and mechanistic effects of delivering reminder cues at specific phases of endogenous sleep rhythms and mark an important step for the endeavor to experimentally modulate memories during sleep.
Collapse
Affiliation(s)
- Hong-Viet V. Ngo
- Department of Psychology, University of Lübeck, 23562 Lübeck, Germany
- Centre for Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Bernhard P. Staresina
- Department of Experimental Psychology, University of Oxford, Oxford OX2 6GG, United Kingdom
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, Department of Psychiatry, University of Oxford, Oxford OX3 9DU, United Kingdom
- School of Psychology, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
45
|
Albrecht JN, Jaramillo V, Huber R, Karlen W, Baumann CR, Brotschi B. Technical feasibility of using auditory phase-targeted stimulation after pediatric severe traumatic brain injury in an intensive care setting. BMC Pediatr 2022; 22:616. [PMID: 36289537 PMCID: PMC9597971 DOI: 10.1186/s12887-022-03667-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/26/2022] [Accepted: 10/10/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Supplementary treatment options after pediatric severe traumatic brain injury (TBI) are needed to improve neurodevelopmental outcome. Evidence suggests enhancement of brain delta waves via auditory phase-targeted stimulation might support neuronal reorganization, however, this method has never been applied in analgosedated patients on the pediatric intensive care unit (PICU). Therefore, we conducted a feasibility study to investigate this approach: In a first recording phase, we examined feasibility of recording over time and in a second stimulation phase, we applied stimulation to address tolerability and efficacy. METHODS Pediatric patients (> 12 months of age) with severe TBI were included between May 2019 and August 2021. An electroencephalography (EEG) device capable of automatic delta wave detection and sound delivery through headphones was used to record brain activity and for stimulation (MHSL-SleepBand version 2). Stimulation tolerability was evaluated based on report of nurses, visual inspection of EEG data and clinical signals (heart rate, intracranial pressure), and whether escalation of therapy to reduce intracranial pressure was needed. Stimulation efficacy was investigated by comparing EEG power spectra of active stimulation versus muted stimulation (unpaired t-tests). RESULTS In total, 4 out of 32 TBI patients admitted to the PICU (12.5%) between 4 and 15 years of age were enrolled in the study. All patients were enrolled in the recording phase and the last one also to the stimulation phase. Recordings started within 5 days after insult and lasted for 1-4 days. Overall, 23-88 h of EEG data per patient were collected. In patient 4, stimulation was enabled for 50 min: No signs of patient stress reactions were observed. Power spectrums between active and muted stimulation were not statistically different (all P > .05). CONCLUSION Results suggests good feasibility of continuously applying devices needed for auditory stimulation over multiple days in pediatric patients with TBI on PICU. Very preliminary evidence suggests good tolerability of auditory stimuli, but efficacy of auditory stimuli to enhance delta waves remains unclear and requires further investigation. However, only low numbers of severe TBI patients could be enrolled in the study and, thus, future studies should consider an international multicentre approach.
Collapse
Affiliation(s)
- Joëlle Ninon Albrecht
- Child Development Center, University Children's Hospital Zurich, University of Zurich (UZH), Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, University of Zurich (UZH), Zurich, Switzerland
| | - Valeria Jaramillo
- Child Development Center, University Children's Hospital Zurich, University of Zurich (UZH), Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, University of Zurich (UZH), Zurich, Switzerland
- Surrey Sleep Research Centre, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
- Neuromodulation Laboratory, School of Psychology, University of Surrey, Guildford, UK
- Care Research and Technology Centre, UK Dementia Research Institute, at Imperial College, University of Surrey, London, Guildford, UK
| | - Reto Huber
- Child Development Center, University Children's Hospital Zurich, University of Zurich (UZH), Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, University of Zurich (UZH), Zurich, Switzerland
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry, University of Zurich (UZH), Zurich, Switzerland
| | - Walter Karlen
- Mobile Health Systems Lab, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Institute of Biomedical Engineering, University of Ulm, Ulm, Germany
| | | | - Barbara Brotschi
- Children's Research Center, University Children's Hospital Zurich, University of Zurich (UZH), Zurich, Switzerland.
- Department of Neonatology and Paediatric Intensive Care, University Children's Hospital Zurich, University of Zurich (UZH), Zurich, Switzerland.
| |
Collapse
|
46
|
Cassim TZ, McGregor KM, Nocera JR, García VV, Sinon CG, Kreuzer M, García PS. Effects of exercise on the sleep microarchitecture in the aging brain: A study on a sedentary sample. Front Syst Neurosci 2022; 16:855107. [PMID: 36387307 PMCID: PMC9644157 DOI: 10.3389/fnsys.2022.855107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 08/26/2022] [Indexed: 03/23/2024] Open
Abstract
Having a healthy sleep pattern plays a vital role in one's overall health. Sleep in the elderly is characterized by decreased slow-wave sleep and an increase of REM sleep. Furthermore, quantitative electroencephalographic (qEEG) studies have shown an age-related attenuation of total EEG power in sleep. However, exercise has been shown to improve sleep across all age groups. In this study, we used the Sleep Profiler™ EEG Sleep Monitor to observe EEG changes occurring during sleep following an aerobic exercise intervention. This study was done on older adults (N = 18, with only five subjects containing both pre- and post-data of sufficient quality for analysis) with an age range 60-85 years. The aerobics regimen was performed three times weekly for 12-weeks commencing with 20-min sessions. The time of each session progressed by 1-2 min/session as needed to a maximum time of 45 min per session. The macro-architecture (sleep stages) and microarchitecture (EEG) results were analyzed using MATLAB. For the microarchitecture, our results showed more deep sleep following the aerobic exercise regimen. Furthermore, for the microarchitecture, out results shows an increase in total EEG power post-exercise in both light (N1 and L1) and deep sleep (N2 and N3). These preliminary changes in sleep the microarchitecture suggest that non-pharmacologic methods might mitigate age-related EEG changes with potential implications for neurocognitive health.
Collapse
Affiliation(s)
- Tuan Z. Cassim
- Department of Anesthesiology, Neuroanesthesia Division, Columbia University Medical Center, New York-Presbyterian Hospital, New York, NY, United States
| | - Keith M. McGregor
- Department of Clinical and Diagnostic Sciences, School of Health Profession, University of Alabama at Birmingham, Birmingham, AL, United States
- Birmingham Veterans Affairs Geriatric Research, Education, and Clinical Center (GRECC), Birmingham, AL, United States
| | - Joe R. Nocera
- VA Rehabilitation R&D Center for Visual and Neurocognitive Rehabilitation, Atlanta VAMC, Decatur, GA, United States
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Violet V. García
- Stamps President’s Scholars Program, Georgia Institute of Technology, Atlanta, GA, United States
| | - Christopher G. Sinon
- Yerkes National Primate Research Center, Neuropharmacology and Neurologic Diseases, Emory University, Atlanta, GA, United States
| | - Matthias Kreuzer
- Department of Anesthesiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Paul S. García
- Department of Anesthesiology, Neuroanesthesia Division, Columbia University Medical Center, New York-Presbyterian Hospital, New York, NY, United States
| |
Collapse
|
47
|
Seok SC, McDevitt E, Mednick SC, Malerba P. Global and non-Global slow oscillations differentiate in their depth profiles. FRONTIERS IN NETWORK PHYSIOLOGY 2022; 2:947618. [PMID: 36926094 PMCID: PMC10013040 DOI: 10.3389/fnetp.2022.947618] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/10/2022] [Indexed: 03/18/2023]
Abstract
Sleep slow oscillations (SOs, 0.5-1.5 Hz) are thought to organize activity across cortical and subcortical structures, leading to selective synaptic changes that mediate consolidation of recent memories. Currently, the specific mechanism that allows for this selectively coherent activation across brain regions is not understood. Our previous research has shown that SOs can be classified on the scalp as Global, Local or Frontal, where Global SOs are found in most electrodes within a short time delay and gate long-range information flow during NREM sleep. The functional significance of space-time profiles of SOs hinges on testing if these differential SOs scalp profiles are mirrored by differential depth structure of SOs in the brain. In this study, we built an analytical framework to allow for the characterization of SO depth profiles in space-time across cortical and sub-cortical regions. To test if the two SO types could be differentiated in their cortical-subcortical activity, we trained 30 machine learning classification algorithms to distinguish Global and non-Global SOs within each individual, and repeated this analysis for light (Stage 2, S2) and deep (slow wave sleep, SWS) NREM stages separately. Multiple algorithms reached high performance across all participants, in particular algorithms based on k-nearest neighbors classification principles. Univariate feature ranking and selection showed that the most differentiating features for Global vs. non-Global SOs appeared around the trough of the SO, and in regions including cortex, thalamus, caudate nucleus, and brainstem. Results also indicated that differentiation during S2 required an extended network of current from cortical-subcortical regions, including all regions found in SWS and other basal ganglia regions, and amygdala and hippocampus, suggesting a potential functional differentiation in the role of Global SOs in S2 vs. SWS. We interpret our results as supporting the potential functional difference of Global and non-Global SOs in sleep dynamics.
Collapse
Affiliation(s)
- Sang-Cheol Seok
- Battelle Center for Mathematical Medicine, Nationwide Children’s Hospital, Columbus, OH, United States
| | | | - Sara C. Mednick
- Department of Cognitive Sciences, University of California, Irvine, Irvine, CA, United States
| | - Paola Malerba
- Battelle Center for Mathematical Medicine, Nationwide Children’s Hospital, Columbus, OH, United States
- Center for Biobehavioral Health, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
- School of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
48
|
Merlot B, Dispersyn G, Husson Z, Chanavaz-Lacheray I, Dennis T, Greco-Vuilloud J, Fougère M, Potvin S, Cotty-Eslous M, Roman H, Marchand S. Pain Reduction With an Immersive Digital Therapeutic Tool in Women Living With Endometriosis-Related Pelvic Pain: Randomized Controlled Trial. J Med Internet Res 2022; 24:e39531. [PMID: 36129733 PMCID: PMC9536521 DOI: 10.2196/39531] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/26/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Chronic pelvic pain is a common and disabling condition in women living with endometriosis. Pharmacological and surgical treatments are not always effective at controlling pain and present important restrictions. Digital therapeutics (DTx) are emerging as major nonpharmacological alternatives that aim to extend the analgesic therapeutic arsenal of patients. OBJECTIVE In this randomized controlled trial (RCT), we aimed to measure the immediate and 4-hour persisting effects of a single use 20-minute DTx (Endocare) on pain in women experiencing pelvic pain due to endometriosis. METHODS A total of 45 women with endometriosis participated in a randomized controlled study comparing the analgesic effect of a single use of a virtual reality digital treatment named Endocare (n=23, 51%) to a 2D digital control (n=22, 49%). Perceived pain and pain relief were measured before the treatment and 15, 30, 45, 60, and 240 minutes after the end of the treatment. RESULTS The clustered posttreatment pain was significantly reduced compared to the pretreatment for both Endocare and the control group (all P<.01). Endocare was significantly more effective than the control group (all P<.01). Endocare decreased the mean pain intensity from 6.0 (SD 1.31) before the treatment to 4.5 (SD 1.71) posttreatment, while the control only decreased it from 5.7 (SD 1.36) to 5.0 (SD 1.43). When comparing each posttreatment measures to the pretest, Endocare significantly reduced pain perception for all points in time up to 4 hours posttreatment. The differences did not reached significance for the control group. Moreover, Endocare was significantly superior to the control group 15, 30, and 45 minutes after the treatment (all P<.001). The mean perceived pain relief was significantly higher for Endocare at 28% (SD 2%) compared to the control, which was 15% (SD 1%) for all the posttreatment measurements (all P>.05). CONCLUSIONS Our study aimed to test the effects of a single use of a DTx treatment on reported pain at different time points in women diagnosed with endometriosis experiencing moderate-to-severe pelvic pain. Importantly, our results support that Endocare, a virtual reality immersive treatment, significantly reduce pain perception compared to a digital control in women living with endometriosis. Interestingly, we are the first to notice that the effect persisted up to 4 hours posttreatment. TRIAL REGISTRATION ClinicalTrials.gov NCT04650516; https://tinyurl.com/2a2eu9wv.
Collapse
Affiliation(s)
- Benjamin Merlot
- Franco European Multidisciplinary Endometriosis Institute (IFEMEndo), Bordeaux, France
| | | | - Zoé Husson
- Franco European Multidisciplinary Endometriosis Institute (IFEMEndo), Bordeaux, France
| | | | - Thomas Dennis
- Franco European Multidisciplinary Endometriosis Institute (IFEMEndo), Bordeaux, France
| | | | | | - Stéphane Potvin
- Centre de Recherche de l'Institut Universitaire en Santé Mentale de Montréal, Montréal, QC, Canada
| | | | - Horace Roman
- Franco European Multidisciplinary Endometriosis Institute (IFEMEndo), Bordeaux, France
| | - Serge Marchand
- Lucine, Bordeaux, France
- Faculté de Médecine et des Sciences de la Santé, Centre de Recherche Clinique du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
49
|
Mander BA, Dave A, Lui KK, Sprecher KE, Berisha D, Chappel-Farley MG, Chen IY, Riedner BA, Heston M, Suridjan I, Kollmorgen G, Zetterberg H, Blennow K, Carlsson CM, Okonkwo OC, Asthana S, Johnson SC, Bendlin BB, Benca RM. Inflammation, tau pathology, and synaptic integrity associated with sleep spindles and memory prior to β-amyloid positivity. Sleep 2022; 45:zsac135. [PMID: 35670275 PMCID: PMC9758508 DOI: 10.1093/sleep/zsac135] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 05/17/2022] [Indexed: 01/25/2023] Open
Abstract
STUDY OBJECTIVES Fast frequency sleep spindles are reduced in aging and Alzheimer's disease (AD), but the mechanisms and functional relevance of these deficits remain unclear. The study objective was to identify AD biomarkers associated with fast sleep spindle deficits in cognitively unimpaired older adults at risk for AD. METHODS Fifty-eight cognitively unimpaired, β-amyloid-negative, older adults (mean ± SD; 61.4 ± 6.3 years, 38 female) enriched with parental history of AD (77.6%) and apolipoprotein E (APOE) ε4 positivity (25.9%) completed the study. Cerebrospinal fluid (CSF) biomarkers of central nervous system inflammation, β-amyloid and tau proteins, and neurodegeneration were combined with polysomnography (PSG) using high-density electroencephalography and assessment of overnight memory retention. Parallelized serial mediation models were used to assess indirect effects of age on fast frequency (13 to <16Hz) sleep spindle measures through these AD biomarkers. RESULTS Glial activation was associated with prefrontal fast frequency sleep spindle expression deficits. While adjusting for sex, APOE ε4 genotype, apnea-hypopnea index, and time between CSF sampling and sleep study, serial mediation models detected indirect effects of age on fast sleep spindle expression through microglial activation markers and then tau phosphorylation and synaptic degeneration markers. Sleep spindle expression at these electrodes was also associated with overnight memory retention in multiple regression models adjusting for covariates. CONCLUSIONS These findings point toward microglia dysfunction as associated with tau phosphorylation, synaptic loss, sleep spindle deficits, and memory impairment even prior to β-amyloid positivity, thus offering a promising candidate therapeutic target to arrest cognitive decline associated with aging and AD.
Collapse
Affiliation(s)
- Bryce A Mander
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA
- Department of Cognitive Sciences, University of California, Irvine, CA, USA
| | - Abhishek Dave
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Department of Cognitive Sciences, University of California, Irvine, CA, USA
| | - Kitty K Lui
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- San Diego State University/University of California San Diego, Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Katherine E Sprecher
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Destiny Berisha
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Miranda G Chappel-Farley
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Ivy Y Chen
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Brady A Riedner
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Margo Heston
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital
, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital
, Mölndal, Sweden
| | - Cynthia M Carlsson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
- Geriatric Research Education and Clinical Center, Wm. S. Middleton Veterans Hospital, Madison, WI, USA
| | - Ozioma C Okonkwo
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
- Geriatric Research Education and Clinical Center, Wm. S. Middleton Veterans Hospital, Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
- Geriatric Research Education and Clinical Center, Wm. S. Middleton Veterans Hospital, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
- Geriatric Research Education and Clinical Center, Wm. S. Middleton Veterans Hospital, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
- Geriatric Research Education and Clinical Center, Wm. S. Middleton Veterans Hospital, Madison, WI, USA
| | - Ruth M Benca
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Psychiatry and Behavioral Medicine, Wake Forest University, Winston-Salem, NC, USA
| |
Collapse
|
50
|
Huwiler S, Carro Dominguez M, Huwyler S, Kiener L, Stich FM, Sala R, Aziri F, Trippel A, Schmied C, Huber R, Wenderoth N, Lustenberger C. Effects of auditory sleep modulation approaches on brain oscillatory and cardiovascular dynamics. Sleep 2022; 45:6632997. [PMID: 35793672 PMCID: PMC9453626 DOI: 10.1093/sleep/zsac155] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/01/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
Slow waves, the hallmark feature of deep nonrapid eye movement sleep, do potentially drive restorative effects of sleep on brain and body functions. Sleep modulation techniques to elucidate the functional role of slow waves thus have gained large interest. Auditory slow wave stimulation is a promising tool; however, directly comparing auditory stimulation approaches within a night and analyzing induced dynamic brain and cardiovascular effects are yet missing. Here, we tested various auditory stimulation approaches in a windowed, 10 s ON (stimulations) followed by 10 s OFF (no stimulations), within-night stimulation design and compared them to a SHAM control condition. We report the results of three studies and a total of 51 included nights and found a large and global increase in slow-wave activity (SWA) in the stimulation window compared to SHAM. Furthermore, slow-wave dynamics were most pronouncedly increased at the start of the stimulation and declined across the stimulation window. Beyond the changes in brain oscillations, we observed, for some conditions, a significant increase in the mean interval between two heartbeats within a stimulation window, indicating a slowing of the heart rate, and increased heart rate variability derived parasympathetic activity. Those cardiovascular changes were positively correlated with the change in SWA, and thus, our findings provide insight into the potential of auditory slow wave enhancement to modulate cardiovascular restorative conditions during sleep. However, future studies need to investigate whether the potentially increased restorative capacity through slow-wave enhancements translates into a more rested cardiovascular system on a subsequent day.
Collapse
Affiliation(s)
- Stephanie Huwiler
- Department of Health Sciences and Technology, Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Manuel Carro Dominguez
- Department of Health Sciences and Technology, Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Silja Huwyler
- Department of Health Sciences and Technology, Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Luca Kiener
- Department of Health Sciences and Technology, Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Fabia M Stich
- Department of Health Sciences and Technology, Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Rossella Sala
- Department of Health Sciences and Technology, Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Florent Aziri
- Department of Health Sciences and Technology, Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Anna Trippel
- Department of Health Sciences and Technology, Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Christian Schmied
- Department of Cardiology, University Heart Center Zurich, University of Zurich, Zurich, Switzerland
| | - Reto Huber
- Center of Competence Sleep and Health Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich, ETH Zurich, Zurich, Switzerland
- Child Development Centre, University Children’s Hospital, University of Zurich, Zurich, Switzerland
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Nicole Wenderoth
- Department of Health Sciences and Technology, Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich, ETH Zurich, Zurich, Switzerland
- Future Health Technologies, Singapore-ETH Center, Campus for Research Excellence and Technological Enterprise (CREATE), Singapore, Singapore
| | - Caroline Lustenberger
- Corresponding author. Caroline Lustenberger, Department of Health Sciences and Technology, Neural Control of Movement Lab, ETH Zurich, Zurich, 8092, Switzerland.
| |
Collapse
|