1
|
Golombek DA, Eyre H, Spiousas I, Casiraghi LP, Hartikainen KM, Partonen T, Pyykkö M, Reynolds CF, Hynes WM, Bassetti CLA, Berk M, Hu K, Ibañez A. Sleep Capital: Linking Brain Health to Wellbeing and Economic Productivity Across the Lifespan. Am J Geriatr Psychiatry 2025; 33:92-106. [PMID: 39117505 DOI: 10.1016/j.jagp.2024.07.011] [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: 05/18/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 08/10/2024]
Abstract
INTRODUCTION AND FRAMEWORK Sleep capital contributes to individual and societal wellbeing, productivity, and economic outcomes and involves a novel aspect of brain capital. It encompasses the quality and quantity of sleep as integral components that influence cognitive abilities, mental and brain health, and physical health, affecting workplace productivity, learning, decision-making, and overall economic performance. Here, we bring a framework to understand the complex relationship between sleep quality, health, wellbeing, and economic productivity. Then we outline the multilevel impact of sleep on cognitive abilities, mental/brain health, and economic indicators, providing evidence for the substantial returns on investment in sleep health initiatives. Moreover, sleep capital is a key factor when considering brain health across the lifespan, especially for the aging population. DISCUSSION We propose specific elements and main variables to develop specific indexes of sleep capital to address its impacts on health, wellbeing and productivity. CONCLUSION Finally, we suggest policy recommendations, workplace interventions, and individual strategies to promote sleep health and brain capital. Investing in sleep capital is essential for fostering a healthier, happier, fairer and more productive society.
Collapse
Affiliation(s)
- Diego A Golombek
- Laboratorio Interdisciplinario del Tiempo (LITERA) (DAG, IS, LPC), Universidad de San Andrés/CONICET, Buenos Aires, Argentina.
| | - Harris Eyre
- Baker Institute for Public Policy (HE), Rice University, Houston, TX, USA; Global Brain Health Institute (HE), University of California San Francisco (UCSF), San Francisco, CA, USA; Department of Psychiatry and Behavioral Science (HE), (UCSF), San Francisco, CA, USA; Department of Psychiatry and Behavioral Science (HE), Baylor College of Medicine, Houston, TX, USA; Department of Psychiatry and Behavioral Science (HE), Houston Methodist, Houston, TX, USA; Department of Psychiatry and Behavioral Science (HE), The University of Texas Health Sciences Center at Houston, Houston, TX, USA; Institute for Mental and Physical Health and Clinical Translation (IMPACT) (HE), Deakin University, Geelong, Victoria, Australia; Euro-Mediterranean Economists Association (HE), Barcelona, Spain; Meadows Mental Health Policy Institute (HE), Dallas, TX, USA; Frontier Technology Lab, School of Engineering (HE), Stanford University, Palo Alto, CA, USA
| | - Ignacio Spiousas
- Laboratorio Interdisciplinario del Tiempo (LITERA) (DAG, IS, LPC), Universidad de San Andrés/CONICET, Buenos Aires, Argentina
| | - Leandro P Casiraghi
- Laboratorio Interdisciplinario del Tiempo (LITERA) (DAG, IS, LPC), Universidad de San Andrés/CONICET, Buenos Aires, Argentina
| | - Kaisa M Hartikainen
- Faculty of Medicine and Health Technology (KMH), Tampere University, Tampere, Finland; Behavioral Neurology Research Group (KMH), Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland; National Brain Health Programme (KMH), Helsinki, Finland
| | - Timo Partonen
- Finnish Institute for Health and Welfare (TP), Helsinki, Finland; Department of Psychiatry (TP), University of Helsinki, Helsinki, Finland
| | - Mika Pyykkö
- Finnish Brain Association and Finnish Centre for Health Promotion (MP), Helsinki, Finland
| | - Charles F Reynolds
- Graduate School of Public Health, University of Pittsburgh School of Medicine (CFR), Pittsburgh, PA, USA
| | - William M Hynes
- Institute for Global Prosperity (MH), University College London, London, UK; Santa Fe Institute (MH), Santa Fe, NM, USA; World Bank (MH), Washington, DC, USA
| | - Claudio L A Bassetti
- Neurology Department, Inselspital (CLAB), University of Bern, Bern, Switzerland; Swiss Brain Health Plan (CLAB), Bern, Switzerland
| | - Michael Berk
- School of Medicine (MB), Deakin University and Barwon Health. Institute for Mental and Physical Health and Clinical Translation (IMPACT), Victoria, Australia
| | - Kun Hu
- Division of Sleep Medicine (KH), Harvard Medical School, Boston, MA, USA; Medical Biodynamics Center (KH), Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Agustín Ibañez
- Latin American Brain Health institute (Brainlat) (CSCN) (AI), Universidad Adolfo Ibanez, Santiago, Chile; ChileGlobal Brain Health Institute, Trinity College Dublin, Ireland; Center for Social and Cognitive Neuroscience (CSCN) (AI), Universidad Adolfo Ibanez, Santiago, Chile; Universidad de San Andrés (AI), Buenos Aires, Argentina
| |
Collapse
|
2
|
Cacciatore S, Calvani R, Mancini J, Ciciarello F, Galluzzo V, Tosato M, Marzetti E, Landi F, Landi F, Bernabei R, Marzetti E, Calvani R, Mariotti L, Cacciatore S, Coelho-Junior HJ, Ciciarello F, Galluzzo V, Martone AM, Picca A, Russo A, Salini S, Tosato M, Abbatecola G, Agostino C, Ambrosio F, Banella F, Benvenuto C, Biscotti D, Brandi V, Bulla MM, Casciani C, Catalano L, Cocchi C, Colloca G, Cucinotta F, D'Angelo M, D'Elia M, D'Ignazio F, Elmi D, Finelli M, Fontanella FP, Fusco D, Gattari I, Gava G, Giani T, Giordano G, Giordano R, Giovanale F, Goracci S, Ialungo S, Labriola R, Levati E, Macaluso M, Marrella L, Massaro C, Montenero R, Notari MV, Paudice M, Persia M, Pirone F, Pompei S, Ragozzino R, Recupero C, Risoli A, Rizzo S, Romaniello D, Rubini G, Russo B, Satriano S, Savera G, Serafini E, Melechì AS, Simeoni F, Simoni S, Taccone C, Tagliacozzi E, Terranova R, Tupputi S, Vaccarella M, Venditti E, Zanchi C, Zuppardo M. Poor sleep quality is associated with probable sarcopenia in community-dwelling older adults: Results from the longevity check-up (lookup) 8. Exp Gerontol 2024; 200:112666. [PMID: 39709067 DOI: 10.1016/j.exger.2024.112666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/29/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Poor sleep quality may contribute to sarcopenia, but evidence remains sparse. This retrospective cross-sectional study investigated the association between subjective sleep quality and probable sarcopenia in a cohort of community-dwelling older adults enrolled in the Longevity Check-Up 8+ study. METHODS Participants were asked about their sleep quality over the past month, with four possible options ("very good", "quite good", "quite bad", very bad"). For the analysis, participants were grouped into good or bad sleep quality categories. Probable sarcopenia was operationalized according to handgrip strength values < 27 kg for men and < 16 kg for women. Logistic regression models were used to explore the relationship between sleep quality and probable sarcopenia. RESULTS 1971 participants were included in the analysis (mean age 73.4 ± 6.2 years, 50.0 % women). Bad sleep quality was reported by 28.3 % of participants and was more prevalent among women, physically inactive individuals, and those with dyslipidemia. Probable sarcopenia was more prevalent in participants with bad sleep quality (23.8 % vs. 18.7 %, p = 0.012). Logistic regression revealed that bad sleep quality was significantly associated with increased odds of probable sarcopenia in both unadjusted (odds ratio [OR] 1.36, 95 % confidence interval [CI] 1.07-1.72, p = 0.010) and fully adjusted models (OR 1.40, 95 % CI 1.08-1.81, p = 0.011). CONCLUSIONS Poor sleep quality is associated with increased likelihood of probable sarcopenia in older adults. This finding highlights the importance of addressing sleep quality in interventions aimed at preventing sarcopenia and promoting healthy aging.
Collapse
Affiliation(s)
- Stefano Cacciatore
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy.
| | - Riccardo Calvani
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Jasmine Mancini
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Francesca Ciciarello
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Vincenzo Galluzzo
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Matteo Tosato
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy.
| | - Francesco Landi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Francesco Landi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Roberto Bernabei
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Riccardo Calvani
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Luca Mariotti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Stefano Cacciatore
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Hélio José Coelho-Junior
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Francesca Ciciarello
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Vincenzo Galluzzo
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Anna Maria Martone
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Anna Picca
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Department of Medicine and Surgery, LUM University, Strada Statale 100 km 18, 70100 Casamassima, Italy
| | - Andrea Russo
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Sara Salini
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Matteo Tosato
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Gabriele Abbatecola
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Clara Agostino
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Fiorella Ambrosio
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Francesca Banella
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Carolina Benvenuto
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Damiano Biscotti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Vincenzo Brandi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Maria Modestina Bulla
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Caterina Casciani
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Lucio Catalano
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Camilla Cocchi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Giuseppe Colloca
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Federica Cucinotta
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Manuela D'Angelo
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Mariaelena D'Elia
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Federica D'Ignazio
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Daniele Elmi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Marta Finelli
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Francesco Pio Fontanella
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Domenico Fusco
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Ilaria Gattari
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Giordana Gava
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Tommaso Giani
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Giulia Giordano
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Rossella Giordano
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Francesca Giovanale
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Simone Goracci
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Silvia Ialungo
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Rosangela Labriola
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Elena Levati
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Myriam Macaluso
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Luca Marrella
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Claudia Massaro
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Rossella Montenero
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Maria Vittoria Notari
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Maria Paudice
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Martina Persia
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Flavia Pirone
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Simona Pompei
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Rosa Ragozzino
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Carla Recupero
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Antonella Risoli
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Stefano Rizzo
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Daria Romaniello
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Giulia Rubini
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Barbara Russo
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Stefania Satriano
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Giulia Savera
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Elisabetta Serafini
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Annalise Serra Melechì
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Francesca Simeoni
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Sofia Simoni
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Chiara Taccone
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Elena Tagliacozzi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Roberta Terranova
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Salvatore Tupputi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Matteo Vaccarella
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Emiliano Venditti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Chiara Zanchi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Maria Zuppardo
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
3
|
Xiong Y, Liang W, Wang X, Zhu H, Yi P, Wei G, Liu H, Lin Y, Zhang L, Ying J, Hua F. S100A8 knockdown activates the PI3K/AKT signaling pathway to inhibit microglial autophagy and improve cognitive impairment mediated by chronic sleep deprivation. Int Immunopharmacol 2024; 143:113375. [PMID: 39418730 DOI: 10.1016/j.intimp.2024.113375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE Cognitive dysfunction is one of the major symptoms of chronic sleep deprivation (CSD). Abnormal autophagy and apoptosis are thought to be important mechanisms. S100 Calcium Binding Protein A8 (S100A8) plays a key role in autophagy and apoptosis of microglia. This study investigated whether S100A8 knockdown can effectively inhibit aberrant autophagy in microglia and improve cognitive function by activating the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway under CSD conditions. METHODS CSD mouse models and BV2 cell autophagy models were established in vivo and in vitro. Transcriptome sequencing was used to determine the key regulator related to autophagy. The Morris water maze test was used to evaluate the cognitive behavior of the mice. RT-qPCR and western blot were conducted to examine S100A8 expression and autophagy signalling. HE, TUNEL, transmission electron microscopy, immunofluorescence, and histochemistry were performed to detect pathological changes, neuronal autophagy, apoptosis, or positive cells in hippocampal tissues, respectively. RESULTS Transcriptome sequencing showed that S100A8 was significantly elevated in CSD mice, and fluorescence colocalization results further suggested that S100A8 mainly colocalizes with microglia. In vivo studies revealed that knockdown of S100A8 alleviated CSD-induced cognitive impairment in mice. Through further mechanistic investigations employing both in vivo and in vitro models, we demonstrated that silencing S100A8 can activate the PI3K/AKT pathway, thereby reducing CSD-induced abnormal autophagy and apoptosis in microglia. Aberrant autophagy and apoptosis in microglia were reversed with the PI3K/AKT pathway inhibitor LY294002. CONCLUSION The S100A8/PI3K/AKT axis plays a crucial role in chronic sleep deprivation-mediated autophagy and apoptosis in microglia.
Collapse
Affiliation(s)
- Yanhong Xiong
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Weidong Liang
- Department of Anesthesiology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xifeng Wang
- Jiangxi Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hong Zhu
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Pengcheng Yi
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Gen Wei
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hailin Liu
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lieliang Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jun Ying
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
4
|
Lim CR, Ogawa S, Kumari Y. Exploring β-caryophyllene: a non-psychotropic cannabinoid's potential in mitigating cognitive impairment induced by sleep deprivation. Arch Pharm Res 2024. [DOI: 10.1007/s12272-024-01523-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
|
5
|
Wang Y, Niu W, Zhu S, Sun J, Lv J, Wang N, Zhang H, Zhang Z, Wang M, Cao L, Li S, Zhai Q, Ma L. STING Agonist cGAMP Attenuates Sleep Deprivation-Induced Neuroinflammation and Cognitive Deficits via TREM2 Up-Regulation. Inflammation 2024; 47:2129-2144. [PMID: 38668837 DOI: 10.1007/s10753-024-02029-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/03/2024] [Accepted: 04/15/2024] [Indexed: 11/30/2024]
Abstract
Sleep deprivation (SD) has been associated with several adverse effects, including cognitive deficit. Emerging evidence suggests microglia-associated neuroinflammation is a potential trigger of cognitive deficit after SD. Stimulator of interferon genes (STING) constitutes an important factor in host immune response to pathogenic organisms and is found in multiple cells, including microglia. STING is involved in neuroinflammation during neuronal degeneration, although how STING signaling affects SD-induced neuroinflammation remains unexplored. In the present study, the chronic sleep restriction (CSR) model was applied to examine the effects of STING signaling on cognition. The results revealed that cGAMP, a high-affinity and selective STING agonist, significantly improved cognitive deficit, alleviated neural injury, and relieved neuroinflammation in CSR mice by activating the STING-TBK1-IRF3 pathway. Moreover, triggering receptor expressed on myeloid cells 2 (TREM2) was upregulated in CSR mice treated with cGAMP, and this effect was abolished by STING knockout. TREM2 upregulation induced by cGAMP regulated the microglia from pro-inflammatory state to anti-inflammatory state, thereby relieving neuroinflammation in CSR mice. These findings indicate cGAMP-induced STING signaling activation alleviates SD-associated neuroinflammation and cognitive deficit by upregulating TREM2, providing a novel approach for the treatment of SD-related nerve injury.
Collapse
Affiliation(s)
- Yue Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Wen Niu
- Department of Physiology and Pathophysiology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Shan Zhu
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jianyu Sun
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jianrui Lv
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Ning Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Huijuan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Zhenni Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Meijuan Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Lingli Cao
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Shuwei Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Qian Zhai
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Lei Ma
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
6
|
Caldwell JA, Knapik JJ, Kusumpa S, Roy TC, Taylor KM, Lieberman HR. Insomnia and sleep apnea in the entire population of US Army soldiers: Associations with deployment and combat exposure 2010-2019, a retrospective cohort investigation. Sleep Health 2024:S2352-7218(24)00210-9. [PMID: 39438179 DOI: 10.1016/j.sleh.2024.09.004] [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: 06/20/2024] [Revised: 08/22/2024] [Accepted: 09/08/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVES This retrospective cohort study examined clinically diagnosed insomnia and sleep apnea and analyzed associations with deployment and combat exposure in active-duty soldiers (n=1,228,346) from 2010 to 2019. METHODS Retrospective data were obtained from the Soldier Performance, Health, and Readiness database. RESULTS Overseas soldier deployments peaked in 2010, decreasing thereafter as soldiers were withdrawn from Iraq and Afghanistan. From 2010 to 2012 insomnia incidence increased at a rate of 6.7 cases/1000 soldier-years, then decreased after 2012 at 5.3 cases/1000 soldier-years. Sleep apnea increased 2010-2016 at 1.9 cases/1000 soldier-years and generally declined thereafter. Risk of insomnia increased with deployment (hazard ratio=1.51; 95% confidence interval=1.49-1.52) and combat exposure (hazard ratio=1.15; 95% confidence interval=1.13-1.17). Risk of sleep apnea was increased by deployment (hazard ratio=1.89; 95% confidence interval, 1.86-1.92) and combat exposure (hazard ratio=1.09; 95% confidence interval, 1.07-1.11). Most relationships remained after accounting for other factors in multivariable analyses, except that the association between sleep apnea and combat exposure was reduced (hazard ratio=0.94; 95% confidence interval=0.92-0.97). CONCLUSIONS Insomnia risk decreased in the period nearly in parallel with a reduction in the number of deployments; nonetheless deployment and combat exposure increased insomnia risk in the period examined. Risk of sleep apnea increased in the period and was related to deployment but not combat exposure after accounting for demographics and comorbid conditions. Despite reductions in insomnia incidence and a slowing in sleep apnea incidence, sleep disorders remain highly prevalent, warranting continued emphasis on sleep-disorder screening and improving the soldier sleep habits.
Collapse
Affiliation(s)
- John A Caldwell
- US Army Research Institute of Environmental Medicine, Military Nutrition Division, Natick, Massachusetts, USA
| | - Joseph J Knapik
- US Army Research Institute of Environmental Medicine, Military Nutrition Division, Natick, Massachusetts, USA
| | - Soothesuk Kusumpa
- US Army Research Institute of Environmental Medicine, Military Nutrition Division, Natick, Massachusetts, USA; Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, USA
| | - Tanja C Roy
- Defense Centers for Public Health-Aberdeen, Clinical Public Health and Epidemiology, Aberdeen Proving Ground, Maryland, USA
| | - Kathryn M Taylor
- US Army Research Institute of Environmental Medicine, Military Nutrition Division, Natick, Massachusetts, USA
| | - Harris R Lieberman
- US Army Research Institute of Environmental Medicine, Military Nutrition Division, Natick, Massachusetts, USA.
| |
Collapse
|
7
|
Mace K, Zimmerman A, Chesi A, Doldur-Balli F, Kim H, Almeraya Del Valle E, Pack AI, Grant SFA, Kayser MS. Cross-species evidence for a developmental origin of adult hypersomnia with loss of synaptic adhesion molecules beat-Ia/CADM2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.615048. [PMID: 39386457 PMCID: PMC11463363 DOI: 10.1101/2024.09.25.615048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Idiopathic hypersomnia (IH) is a poorly-understood sleep disorder characterized by excessive daytime sleepiness despite normal nighttime sleep. Combining human genomics with behavioral and mechanistic studies in fish and flies, we uncover a role for beat-Ia/CADM2 , synaptic adhesion molecules of the immunoglobulin superfamily, in excessive sleepiness. Neuronal knockdown of Drosophila beat-Ia results in sleepy flies and loss of the vertebrate ortholog of beat-Ia , CADM2 , results in sleepy fish. We delineate a developmental function for beat-Ia in synaptic elaboration of neuropeptide F (NPF) neurites projecting to the suboesophageal zone (SEZ) of the fly brain. Brain connectome and experimental evidence demonstrate these NPF outputs synapse onto a subpopulation of SEZ GABAergic neurons to stabilize arousal. NPF is the Drosophila homolog of vertebrate neuropeptide Y (NPY), and an NPY receptor agonist restores sleep to normal levels in zebrafish lacking CADM2 . These findings point towards NPY modulation as a treatment target for human hypersomnia.
Collapse
|
8
|
Kohyama J. Re-Evaluating Recommended Optimal Sleep Duration: A Perspective on Sleep Literacy. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1098. [PMID: 39334630 PMCID: PMC11429570 DOI: 10.3390/children11091098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024]
Abstract
A significant number of adolescents experience sleepiness, primarily due to sleep deprivation. The detrimental effects of inadequate sleep on both physical and mental health are well documented, particularly during adolescence-a critical developmental stage that has far-reaching implications for later life outcomes. The International Classification of Diseases 11th Revision recently introduced the disorder termed 'insufficient sleep syndrome,' characterized by a persistent reduction in sleep quantity. However, diagnosing this condition based solely on sleep duration is challenging due to significant individual variation in what constitutes optimal sleep. Despite this, managing sleep debt remains difficult without a clear understanding of individual optimal sleep needs. This review aims to reassess recommended sleep durations, with a focus on enhancing sleep literacy. Beginning with an exploration of insufficient sleep syndrome, this review delves into research on optimal sleep duration and examines foundational studies on sleep debt's impact on the developing brain. Finally, it addresses the challenges inherent in sleep education programs from the perspective of sleep literacy. By doing so, this review seeks to contribute to a deeper understanding of the chronic sleep debt issues faced by adolescents, particularly those affected by insufficient sleep syndrome.
Collapse
Affiliation(s)
- Jun Kohyama
- Tokyo Bay Urayasu Ichikawa Medical Center, Urayasu 279-0001, Japan
| |
Collapse
|
9
|
Voigt RM, Ouyang B, Keshavarzian A. Outdoor nighttime light exposure (light pollution) is associated with Alzheimer's disease. Front Neurosci 2024; 18:1378498. [PMID: 39308948 PMCID: PMC11412842 DOI: 10.3389/fnins.2024.1378498] [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: 01/29/2024] [Accepted: 07/09/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction Alzheimer's disease (AD) prevalence has increased in the last century which can be attributed to increased lifespan, but environment is also important. Exposure to artificial light at night is one environmental factor that may influence AD. Methods This study evaluated the relationship between outdoor nighttime light exposure and AD prevalence in the United States using satellite acquired outdoor nighttime light intensity and Medicare data. Results Higher outdoor nighttime light was associated with higher prevalence of AD. While atrial fibrillation, diabetes, hyperlipidemia, hypertension, and stroke were associated more strongly with AD prevalence than nighttime light intensity, nighttime light was more strongly associated with AD prevalence than alcohol abuse, chronic kidney disease, depression, heart failure, and obesity. Startlingly, nighttime light exposure more strongly associated with AD prevalence in those under the age of 65 than any other disease factor examined. Discussion These data suggest light exposure at night may influence AD, but additional studies are needed.
Collapse
Affiliation(s)
- Robin M. Voigt
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
| | - Bichun Ouyang
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States
| | - Ali Keshavarzian
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
- Department of Physiology, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
10
|
Zhao X, Lu J, Zhang J, Liu C, Wang H, Wang Y, Du Q. Sleep restriction promotes brain oxidative stress and inflammation, and aggravates cognitive impairment in insulin-resistant mice. Psychoneuroendocrinology 2024; 166:107065. [PMID: 38718616 DOI: 10.1016/j.psyneuen.2024.107065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/17/2024] [Accepted: 04/28/2024] [Indexed: 06/16/2024]
Abstract
Sleep deprivation and insulin resistance (IR) are two risk factors for Alzheimer's disease. As the population of people with IR increases and sleep restriction (SR) due to staying up late becomes the "new normal", it is necessary to investigate the effects and molecular pathogenesis of chronic SR on cognitive function in insulin resistance. In this study, 4-week-old mice were fed a high-fat diet (HFD) for 8 weeks to establish IR model, and then the mice were subjected to SR for 21 days, and related indicators were assessed, including cognitive capacity, apoptosis, oxidative stress, glial cell activation, inflammation, blood-brain barrier (BBB) permeability and adiponectin levels, for exploring the potential regulatory mechanisms. Compared with control group, IR mice showed impaired cognitive capacity, meanwhile, SR not only promoted Bax/Bcl2-induced hippocampal neuronal cell apoptosis and Nrf2/HO1- induced oxidative stress, but also increased microglia activation and inflammatory factor levels and BBB permeability, thus aggravating the cognitive impairment in IR mice. Consequently, changing bad living habits and ensuring sufficient sleep are important intervention strategies to moderate the aggravation of IR-induced cognitive impairment.
Collapse
Affiliation(s)
- Xu Zhao
- Centre of General Practice, The Seventh Affiliated Hospital of Southern Medical University, Foshan 528200, China
| | - Jiancong Lu
- The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Jingyi Zhang
- Centre of General Practice, The Seventh Affiliated Hospital of Southern Medical University, Foshan 528200, China
| | - Ce Liu
- Department of Laboratory Medicine, The Seventh Affiliated Hospital of Southern Medical University, Foshan 528200, China
| | - Huijun Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Yan Wang
- Biomedical Research Center, Southern Medical University, Guangzhou 510515, China; Division of Gastroenterology and Hepatology, The Seventh Affiliated Hospital, Southern Medical University, Foshan 528200, China.
| | - Qingfeng Du
- Centre of General Practice, The Seventh Affiliated Hospital of Southern Medical University, Foshan 528200, China; School of Traditional Chinese medicine, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China.
| |
Collapse
|
11
|
Wang X, Li Y, Wang X, Wang R, Hao Y, Ren F, Wang P, Fang B. Faecalibacterium prausnitzii Supplementation Prevents Intestinal Barrier Injury and Gut Microflora Dysbiosis Induced by Sleep Deprivation. Nutrients 2024; 16:1100. [PMID: 38674791 PMCID: PMC11054126 DOI: 10.3390/nu16081100] [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: 02/24/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Sleep deprivation (SD) leads to impaired intestinal barrier function and intestinal flora disorder, especially a reduction in the abundance of the next generation of probiotic Faecalibacterium prausnitzii (F. prausnitzii). However, it remains largely unclear whether F. prausnitzii can ameliorate SD-induced intestinal barrier damage. A 72 h SD mouse model was used in this research, with or without the addition of F. prausnitzii. The findings indicated that pre-colonization with F. prausnitzii could protect against tissue damage from SD, enhance goblet cell count and MUC2 levels in the colon, boost tight-junction protein expression, decrease macrophage infiltration, suppress pro-inflammatory cytokine expression, and reduce apoptosis. We found that the presence of F. prausnitzii helped to balance the gut microbiota in SD mice by reducing harmful bacteria like Klebsiella and Staphylococcus, while increasing beneficial bacteria such as Akkermansia. Ion chromatography analysis revealed that F. prausnitzii pretreatment increased the fecal butyrate level in SD mice. Overall, these results suggested that incorporating F. prausnitzii could help reduce gut damage caused by SD, potentially by enhancing the intestinal barrier and balancing gut microflora. This provides a foundation for utilizing probiotics to protect against intestinal illnesses.
Collapse
Affiliation(s)
- Xintong Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (X.W.); (Y.L.); (R.W.); (Y.H.); (F.R.)
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (X.W.); (Y.L.); (R.W.); (Y.H.); (F.R.)
| | - Xifan Wang
- Department of Obstetrics and Gynecology, Columbia University, New York, NY 10032, USA;
| | - Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (X.W.); (Y.L.); (R.W.); (Y.H.); (F.R.)
| | - Yanling Hao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (X.W.); (Y.L.); (R.W.); (Y.H.); (F.R.)
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (X.W.); (Y.L.); (R.W.); (Y.H.); (F.R.)
- Food Laboratory of Zhongyuan, Luohe 462000, China
| | - Pengjie Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (X.W.); (Y.L.); (R.W.); (Y.H.); (F.R.)
- Food Laboratory of Zhongyuan, Luohe 462000, China
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (X.W.); (Y.L.); (R.W.); (Y.H.); (F.R.)
| |
Collapse
|
12
|
Andrillon T, Taillard J, Strauss M. Sleepiness and the transition from wakefulness to sleep. Neurophysiol Clin 2024; 54:102954. [PMID: 38460284 DOI: 10.1016/j.neucli.2024.102954] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 03/11/2024] Open
Abstract
The transition from wakefulness to sleep is a progressive process that is reflected in the gradual loss of responsiveness, an alteration of cognitive functions, and a drastic shift in brain dynamics. These changes do not occur all at once. The sleep onset period (SOP) refers here to this period of transition between wakefulness and sleep. For example, although transitions of brain activity at sleep onset can occur within seconds in a given brain region, these changes occur at different time points across the brain, resulting in a SOP that can last several minutes. Likewise, the transition to sleep impacts cognitive and behavioral levels in a graded and staged fashion. It is often accompanied and preceded by a sensation of drowsiness and the subjective feeling of a need for sleep, also associated with specific physiological and behavioral signatures. To better characterize fluctuations in vigilance and the SOP, a multidimensional approach is thus warranted. Such a multidimensional approach could mitigate important limitations in the current classification of sleep, leading ultimately to better diagnoses and treatments of individuals with sleep and/or vigilance disorders. These insights could also be translated in real-life settings to either facilitate sleep onset in individuals with sleep difficulties or, on the contrary, prevent or control inappropriate sleep onsets.
Collapse
Affiliation(s)
- Thomas Andrillon
- Paris Brain Institute, Sorbonne Université, Inserm-CNRS, Paris 75013, France; Monash Centre for Consciousness & Contemplative Studies, Monash University, Melbourne, VIC 3800, Australia
| | - Jacques Taillard
- Univ. Bordeaux, CNRS, SANPSY, UMR 6033, F-33000 Bordeaux, France
| | - Mélanie Strauss
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), CUB Hôpital Érasme, Services de Neurologie, Psychiatrie et Laboratoire du sommeil, Route de Lennik 808 1070 Bruxelles, Belgium; Neuropsychology and Functional Neuroimaging Research Group (UR2NF), Center for Research in Cognition and Neurosciences (CRCN), Université Libre de Bruxelles, B-1050 Brussels, Belgium.
| |
Collapse
|
13
|
Weiss JT, Blundell MZ, Singh P, Donlea JM. Sleep deprivation drives brain-wide changes in cholinergic presynapse abundance in Drosophila melanogaster. Proc Natl Acad Sci U S A 2024; 121:e2312664121. [PMID: 38498719 PMCID: PMC10990117 DOI: 10.1073/pnas.2312664121] [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: 07/24/2023] [Accepted: 02/19/2024] [Indexed: 03/20/2024] Open
Abstract
Sleep is an evolutionarily conserved state that supports brain functions, including synaptic plasticity, in species across the animal kingdom. Here, we examine the neuroanatomical and cell-type distribution of presynaptic scaling in the fly brain after sleep loss. We previously found that sleep loss drives accumulation of the active zone scaffolding protein Bruchpilot (BRP) within cholinergic Kenyon cells of the Drosophila melanogaster mushroom body (MB), but not in other classes of MB neurons. To test whether similar cell type-specific trends in plasticity occur broadly across the brain, we used a flp-based genetic reporter to label presynaptic BRP in cholinergic, dopaminergic, GABAergic, or glutamatergic neurons. We then collected whole-brain confocal image stacks of BRP intensity to systematically quantify BRP, a marker of presynapse abundance, across 37 neuropil regions of the central fly brain. Our results indicate that sleep loss, either by overnight (12-h) mechanical stimulation or chronic sleep disruption in insomniac mutants, broadly elevates cholinergic synapse abundance across the brain, while synapse abundance in neurons that produce other neurotransmitters undergoes weaker, if any, changes. Extending sleep deprivation to 24 h drives brain-wide upscaling in glutamatergic, but not other, synapses. Finally, overnight male-male social pairings induce increased BRP in excitatory synapses despite male-female pairings eliciting more waking activity, suggesting experience-specific plasticity. Within neurotransmitter class and waking context, BRP changes are similar across the 37 neuropil domains, indicating that similar synaptic scaling rules may apply across the brain during acute sleep loss and that sleep need may broadly alter excitatory-inhibitory balance in the central brain.
Collapse
Affiliation(s)
- Jacqueline T. Weiss
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, CA90095
- Neuroscience Interdepartmental Program, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Mei Z. Blundell
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, CA90095
| | - Prabhjit Singh
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, CA90095
| | - Jeffrey M. Donlea
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, CA90095
| |
Collapse
|
14
|
Fjell AM, Walhovd KB. Individual sleep need is flexible and dynamically related to cognitive function. Nat Hum Behav 2024; 8:422-430. [PMID: 38379065 DOI: 10.1038/s41562-024-01827-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 01/15/2024] [Indexed: 02/22/2024]
Abstract
Given that sleep deprivation studies consistently show that short sleep causes neurocognitive deficits, the effects of insufficient sleep on brain health and cognition are of great interest and concern. Here we argue that experimentally restricted sleep is not a good model for understanding the normal functions of sleep in naturalistic settings. Cross-disciplinary research suggests that human sleep is remarkably dependent on environmental conditions and social norms, thus escaping universally applicable rules. Sleep need varies over time and differs between individuals, showing a complex relationship with neurocognitive function. This aspect of sleep is rarely addressed in experimental work and is not reflected in expert recommendations about sleep duration. We recommend focusing on the role of individual and environmental factors to improve our understanding of the relationship between human sleep and cognition.
Collapse
Affiliation(s)
- Anders M Fjell
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway.
- Center for Computational Radiology and Artificial Intelligence, Oslo University Hospital, Oslo, Norway.
| | - Kristine B Walhovd
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway
- Center for Computational Radiology and Artificial Intelligence, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
15
|
Tang L, Zhang H, Liao Y, Zhou S, Yang Y, Zhang M, Guo Y, Xie T, Chen S, Ouyang W, Lin X, Wang S, Huang C, Zhang M, Zhuang J, Zhao J, Zhang R, Zhang C, Jin Z, Hu J, Liu Z. Chronic Sleep Deprivation Impairs Visual Functions via Oxidative Damage in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:307-320. [PMID: 38245252 DOI: 10.1016/j.ajpath.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/16/2023] [Accepted: 11/06/2023] [Indexed: 01/22/2024]
Abstract
Sleep deprivation (SD) is a global public health burden, and has a detrimental role in the nervous system. Retina is an important part of the central nervous system; however, whether SD affects retinal structures and functions remains largely unknown. Herein, chronic SD mouse model indicated that loss of sleep for 4 months could result in reductions in the visual functions, but without obvious morphologic changes of the retina. Ultrastructural analysis by transmission electron microscope revealed the deterioration of mitochondria, which was accompanied with the decrease of multiple mitochondrial proteins in the retina. Mechanistically, oxidative stress was provoked by chronic SD, which could be ameliorated after rest, and thus restore retinal homeostasis. Moreover, the supplementation of two antioxidants, α-lipoic acid and N-acetyl-l-cysteine, could reduce retinal reactive oxygen species, repair damaged mitochondria, and, as a result, improve the retinal functions. Overall, this work demonstrated the essential roles of sleep in maintaining the integrity and health of the retina. More importantly, it points towards supplementation of antioxidants as an effective intervention strategy for people experiencing sleep shortages.
Collapse
Affiliation(s)
- Liying Tang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Ophthalmology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Houjian Zhang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Yi Liao
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Shengmei Zhou
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yaqiong Yang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Mouxin Zhang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yuli Guo
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Tingyu Xie
- Department of Ophthalmology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Shikun Chen
- Department of Ophthalmology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Weijie Ouyang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiang Lin
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Shaopan Wang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Caihong Huang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Minjie Zhang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jingbin Zhuang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jiankai Zhao
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Rongrong Zhang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Changjun Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Zibing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Jiaoyue Hu
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang, China; Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.
| | - Zuguo Liu
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang, China; Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.
| |
Collapse
|
16
|
Curtis AF, Nair N, Hayse B, McGovney K, Mikula C, Halder P, Craggs JG, Kiselica A, McCrae CS. Preliminary investigation of the interactive role of physiological arousal and insomnia complaints in gray matter volume alterations in chronic widespread pain. J Clin Sleep Med 2024; 20:293-302. [PMID: 37823586 PMCID: PMC10835766 DOI: 10.5664/jcsm.10860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
STUDY OBJECTIVES Brain regions involved in insomnia and chronic pain are overlapping and diffuse. The interactive role of physiological arousal in associations between insomnia symptoms and neural regions is unknown. This preliminary study examined whether arousal interacted with sleep in associations with gray matter volume of frontal (dorsolateral prefrontal cortex, anterior cingulate cortex) and temporal (right/left hippocampus) regions in adults with chronic widespread pain and insomnia complaints. METHODS Forty-seven adults with chronic widespread pain and insomnia (mean age = 46.00, standard deviation = 13.88, 89% women) completed 14 daily diaries measuring sleep onset latency (SOL), wake time after sleep onset, and total sleep time (TST), as well as Holter monitor assessments of heart rate variability (measuring physiological arousal), and magnetic resonance imaging. Multiple regressions examined whether average SOL, wake time after sleep onset, or TST were independently or interactively (with arousal/heart rate variability) associated with dorsolateral prefrontal cortex, anterior cingulate cortex, and left/right hippocampus gray matter volumes. RESULTS Shorter TST was associated with lower right hippocampus volume. TST also interacted with arousal in its association with right hippocampal volume, Specifically, shorter TST was associated with lower volume at highest and average arousal levels. SOL interacted with arousal in its association with anterior cingulate cortex volume, such that, among individuals with lowest arousal, longer SOL was associated with lower volume. CONCLUSIONS Preliminary findings highlight the interactive roles of physiological arousal and insomnia symptoms in associations with neural structure in chronic widespread pain and insomnia. Individuals with the highest physiological arousal may be particularly vulnerable to the impact of shorter TST on hippocampal volume loss. Reducing SOL may only impact anterior cingulate cortex volume in those with lower physiological arousal. CITATION Curtis AF, Nair N, Hayse B, et al. Preliminary investigation of the interactive role of physiological arousal and insomnia complaints in gray matter volume alterations in chronic widespread pain. J Clin Sleep Med. 2024;20(2):293-302.
Collapse
Affiliation(s)
- Ashley F. Curtis
- College of Nursing, University of South Florida, Tampa, Florida
- Department of Psychiatry, University of Missouri-Columbia, Columbia, Missouri
- Department of Psychological Sciences, University of Missouri-Columbia, Columbia, Missouri
| | - Neetu Nair
- Department of Psychiatry, University of Missouri-Columbia, Columbia, Missouri
| | - Braden Hayse
- Department of Psychiatry, University of Missouri-Columbia, Columbia, Missouri
| | - Kevin McGovney
- Department of Psychological Sciences, University of Missouri-Columbia, Columbia, Missouri
| | - Cynthia Mikula
- Department of Health Psychology, University of Missouri-Columbia, Columbia, Missouri
| | - Puja Halder
- Department of Psychiatry, University of Missouri-Columbia, Columbia, Missouri
| | - Jason G. Craggs
- Department of Psychiatry, University of Missouri-Columbia, Columbia, Missouri
- Department of Physical Therapy, University of Missouri-Columbia, Columbia, Missouri
- Department of Psychiatry & Behavioral Neurosciences, University of South Florida, Tampa, FL
| | - Andrew Kiselica
- Department of Health Psychology, University of Missouri-Columbia, Columbia, Missouri
| | - Christina S. McCrae
- College of Nursing, University of South Florida, Tampa, Florida
- Department of Psychiatry, University of Missouri-Columbia, Columbia, Missouri
| |
Collapse
|
17
|
Jiang N, Yao C, Zhang Y, Sun X, Choudhary MI, Liu X. Ginsenoside Rg1 Attenuates Chronic Sleep Deprivation-Induced Hippocampal Mitochondrial Dysfunction and Improves Memory by the AMPK-SIRT3 Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:2362-2373. [PMID: 38236060 DOI: 10.1021/acs.jafc.3c04618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Ginsenoside Rg1 (Rg1) is the main bioactive ginseng component. This study investigates the effects of Rg1 on cognitive deficits triggered by chronic sleep deprivation stress (CSDS) and explores its underlying mechanisms. Rg1 effectively improved spatial working and recognition memory, as evidenced by various behavioral tests. RNA-sequence analysis revealed differential gene expression in the metabolic pathway. Treatment with Rg1 abrogated reductions in SOD and CAT activity, lowered MDA content, and increased Nrf2 and HO-1 protein levels. Rg1 administration alleviated hippocampal mitochondrial dysfunction by restoring normal ultrastructure and enhancing ATP activities and Mfn2 expression while regulating Drp-1 expression. Rg1 mitigated neuronal apoptosis by reducing the Bax/Bcl-2 ratio and the levels of cleaved caspase-3. Additionally, Rg1 upregulated AMPK and SIRT3 protein expressions. These findings suggest that Rg1 has potential as a robust intervention for cognitive dysfunction associated with sleep deprivation, acting through the modulation of mitochondrial function, oxidative stress, apoptosis, and the AMPK-SIRT3 axis.
Collapse
Affiliation(s)
- Ning Jiang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Caihong Yao
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Yiwen Zhang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Xinran Sun
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - M Iqbal Choudhary
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Xinmin Liu
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, Zhejiang, China
- Qian Xuesen Collaborative Research Center of Astrochemistry and Space Life Sciences, Ningbo University, Ningbo 315211, Zhejiang, China
| |
Collapse
|
18
|
Sang D, Lin K, Yang Y, Ran G, Li B, Chen C, Li Q, Ma Y, Lu L, Cui XY, Liu Z, Lv SQ, Luo M, Liu Q, Li Y, Zhang EE. Prolonged sleep deprivation induces a cytokine-storm-like syndrome in mammals. Cell 2023; 186:5500-5516.e21. [PMID: 38016470 DOI: 10.1016/j.cell.2023.10.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 08/17/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023]
Abstract
Most animals require sleep, and sleep loss induces serious pathophysiological consequences, including death. Previous experimental approaches for investigating sleep impacts in mice have been unable to persistently deprive animals of both rapid eye movement sleep (REMS) and non-rapid eye movement sleep (NREMS). Here, we report a "curling prevention by water" paradigm wherein mice remain awake 96% of the time. After 4 days of exposure, mice exhibit severe inflammation, and approximately 80% die. Sleep deprivation increases levels of prostaglandin D2 (PGD2) in the brain, and we found that elevated PGD2 efflux across the blood-brain-barrier-mediated by ATP-binding cassette subfamily C4 transporter-induces both accumulation of circulating neutrophils and a cytokine-storm-like syndrome. Experimental disruption of the PGD2/DP1 axis dramatically reduced sleep-deprivation-induced inflammation. Thus, our study reveals that sleep-related changes in PGD2 in the central nervous system drive profound pathological consequences in the peripheral immune system.
Collapse
Affiliation(s)
- Di Sang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China; National Institute of Biological Sciences, Beijing, China
| | - Keteng Lin
- National Institute of Biological Sciences, Beijing, China; College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yini Yang
- Peking University School of Life Sciences, Beijing, China
| | - Guangdi Ran
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Bohan Li
- Peking-Tsinghua Center for Life Sciences, Beijing, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Chen Chen
- National Institute of Biological Sciences, Beijing, China
| | - Qi Li
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Yan Ma
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Lihui Lu
- National Institute of Biological Sciences, Beijing, China
| | - Xi-Yang Cui
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Zhibo Liu
- Peking-Tsinghua Center for Life Sciences, Beijing, China; Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Sheng-Qing Lv
- Department of Neurosurgery, Xinqiao Hospital, Chongqing, China
| | - Minmin Luo
- National Institute of Biological Sciences, Beijing, China; School of Life Sciences, Tsinghua University, Beijing, China; Chinese Institute for Brain Research, Beijing, China
| | - Qinghua Liu
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Yulong Li
- Peking University School of Life Sciences, Beijing, China; Peking-Tsinghua Center for Life Sciences, Beijing, China; State Key Laboratory of Membrane Biology, Beijing, China; PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Eric Erquan Zhang
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| |
Collapse
|
19
|
Vaquer-Alicea A, Yu J, Liu H, Lucey BP. Plasma and cerebrospinal fluid proteomic signatures of acutely sleep-deprived humans: an exploratory study. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2023; 4:zpad047. [PMID: 38046221 PMCID: PMC10691441 DOI: 10.1093/sleepadvances/zpad047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/06/2023] [Indexed: 12/05/2023]
Abstract
Study Objectives Acute sleep deprivation affects both central and peripheral biological processes. Prior research has mainly focused on specific proteins or biological pathways that are dysregulated in the setting of sustained wakefulness. This exploratory study aimed to provide a comprehensive view of the biological processes and proteins impacted by acute sleep deprivation in both plasma and cerebrospinal fluid (CSF). Methods We collected plasma and CSF from human participants during one night of sleep deprivation and controlled normal sleep conditions. One thousand and three hundred proteins were measured at hour 0 and hour 24 using a high-scale aptamer-based proteomics platform (SOMAscan) and a systematic biological database tool (Metascape) was used to reveal altered biological pathways. Results Acute sleep deprivation decreased the number of upregulated and downregulated biological pathways and proteins in plasma but increased upregulated and downregulated biological pathways and proteins in CSF. Predominantly affected proteins and pathways were associated with immune response, inflammation, phosphorylation, membrane signaling, cell-cell adhesion, and extracellular matrix organization. Conclusions The identified modifications across biofluids add to evidence that acute sleep deprivation has important impacts on biological pathways and proteins that can negatively affect human health. As a hypothesis-driving study, these findings may help with the exploration of novel mechanisms that mediate sleep loss and associated conditions, drive the discovery of new sleep loss biomarkers, and ultimately aid in the identification of new targets for intervention to human diseases.
Collapse
Affiliation(s)
- Ana Vaquer-Alicea
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Jinsheng Yu
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Haiyan Liu
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Brendan P Lucey
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
20
|
Park J, Kim DY, Hwang GS, Han IO. Repeated sleep deprivation decreases the flux into hexosamine biosynthetic pathway/O-GlcNAc cycling and aggravates Alzheimer's disease neuropathology in adult zebrafish. J Neuroinflammation 2023; 20:257. [PMID: 37946213 PMCID: PMC10634120 DOI: 10.1186/s12974-023-02944-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
This study investigated chronic and repeated sleep deprivation (RSD)-induced neuronal changes in hexosamine biosynthetic pathway/O-linked N-acetylglucosamine (HBP/O-GlcNAc) cycling of glucose metabolism and further explored the role of altered O-GlcNAc cycling in promoting neurodegeneration using an adult zebrafish model. RSD-triggered degenerative changes in the brain led to impairment of memory, neuroinflammation and amyloid beta (Aβ) accumulation. Metabolite profiling of RSD zebrafish brain revealed a significant decrease in glucose, indicating a potential association between RSD-induced neurodegeneration and dysregulated glucose metabolism. While RSD had no impact on overall O-GlcNAcylation levels in the hippocampus region, changes were observed in two O-GlcNAcylation-regulating enzymes, specifically, a decrease in O-GlcNAc transferase (OGT) and an increase in O-GlcNAcase (OGA). Glucosamine (GlcN) treatment induced an increase in O-GlcNAcylation and recovery of the OGT level that was decreased in the RSD group. In addition, GlcN reversed cognitive impairment by RSD. GlcN reduced neuroinflammation and attenuated Aβ accumulation induced by RSD. Repeated treatment of zebrafish with diazo-5-oxo-l-norleucine (DON), an inhibitor of HBP metabolism, resulted in cognitive dysfunction, neuroinflammation and Aβ accumulation, similar to the effects of RSD. The pathological changes induced by DON were restored to normal upon treatment with GlcN. Both the SD and DON-treated groups exhibited a common decrease in glutamate and γ-aminobutyric acid compared to the control group. Overexpression of OGT in zebrafish brain rescued RSD-induced neuronal dysfunction and neurodegeneration. RSD induced a decrease in O-GlcNAcylation of amyloid precursor protein and increase in β-secretase activity, which were reversed by GlcN treatment. Based on the collective findings, we propose that dysregulation of HBP and O-GlcNAc cycling in brain plays a crucial role in RSD-mediated progression of neurodegeneration and Alzheimer's disease pathogenesis. Targeting of this pathway may, therefore, offer an effective regulatory approach for treatment of sleep-associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Jiwon Park
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, Korea
| | - Dong Yeol Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Korea
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Inn-Oc Han
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, Korea.
- Department of Physiology and Biophysics, College of Medicine, Inha University, 100 Inha Ro, Nam-Gu, Incheon, 22212, Korea.
| |
Collapse
|
21
|
Romigi A, Brown RE. Editorial: Consequences of sleep deprivation. Front Neurosci 2023; 17:1254248. [PMID: 37592947 PMCID: PMC10431962 DOI: 10.3389/fnins.2023.1254248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 08/19/2023] Open
Affiliation(s)
- Andrea Romigi
- IRCCS Neuromed Istituto Neurologico Mediterraneo Pozzilli (IS), Pozzilli, Italy
- Universita Telematica Internazionale UNINETTUNO, Rome, Italy
| | - Ritchie Edward Brown
- Laboratory of Neuroscience, VA Boston Healthcare System and Harvard Medical School, Department of Psychiatry, West Roxbury, MA, United States
- Boston VA Research Institute, Boston, MA, United States
| |
Collapse
|
22
|
Fjell AM, Sørensen Ø, Wang Y, Amlien IK, Baaré WFC, Bartrés-Faz D, Boraxbekk CJ, Brandmaier AM, Demuth I, Drevon CA, Ebmeier KP, Ghisletta P, Kievit R, Kühn S, Madsen KS, Nyberg L, Solé-Padullés C, Vidal-Piñeiro D, Wagner G, Watne LO, Walhovd KB. Is Short Sleep Bad for the Brain? Brain Structure and Cognitive Function in Short Sleepers. J Neurosci 2023; 43:5241-5250. [PMID: 37365003 PMCID: PMC10342221 DOI: 10.1523/jneurosci.2330-22.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 05/01/2023] [Accepted: 05/08/2023] [Indexed: 06/28/2023] Open
Abstract
Many sleep less than recommended without experiencing daytime sleepiness. According to prevailing views, short sleep increases risk of lower brain health and cognitive function. Chronic mild sleep deprivation could cause undetected sleep debt, negatively affecting cognitive function and brain health. However, it is possible that some have less sleep need and are more resistant to negative effects of sleep loss. We investigated this using a cross-sectional and longitudinal sample of 47,029 participants of both sexes (20-89 years) from the Lifebrain consortium, Human Connectome project (HCP) and UK Biobank (UKB), with measures of self-reported sleep, including 51,295 MRIs of the brain and cognitive tests. A total of 740 participants who reported to sleep <6 h did not experience daytime sleepiness or sleep problems/disturbances interfering with falling or staying asleep. These short sleepers showed significantly larger regional brain volumes than both short sleepers with daytime sleepiness and sleep problems (n = 1742) and participants sleeping the recommended 7-8 h (n = 3886). However, both groups of short sleepers showed slightly lower general cognitive function (GCA), 0.16 and 0.19 SDs, respectively. Analyses using accelerometer-estimated sleep duration confirmed the findings, and the associations remained after controlling for body mass index, depression symptoms, income, and education. The results suggest that some people can cope with less sleep without obvious negative associations with brain morphometry and that sleepiness and sleep problems may be more related to brain structural differences than duration. However, the slightly lower performance on tests of general cognitive abilities warrants closer examination in natural settings.SIGNIFICANCE STATEMENT Short habitual sleep is prevalent, with unknown consequences for brain health and cognitive performance. Here, we show that daytime sleepiness and sleep problems are more strongly related to regional brain volumes than sleep duration. However, participants sleeping ≤6 h had slightly lower scores on tests of general cognitive function (GCA). This indicates that sleep need is individual and that sleep duration per se is very weakly if at all related brain health, while daytime sleepiness and sleep problems may show somewhat stronger associations. The association between habitual short sleep and lower scores on tests of general cognitive abilities must be further scrutinized in natural settings.
Collapse
Affiliation(s)
- Anders M Fjell
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, 0373 Oslo, Norway
- Computational Radiology and Artificial Intelligence, Department of Radiology and Nuclear Medicine, Oslo University Hospital, 0424 Oslo, Norway
| | - Øystein Sørensen
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, 0373 Oslo, Norway
| | - Yunpeng Wang
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, 0373 Oslo, Norway
| | - Inge K Amlien
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, 0373 Oslo, Norway
| | - William F C Baaré
- Danish Research Centre for Magnetic Resonance (DRCMR), Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital-Amager and Hvidovre, 2650 Hvidovre, Copenhagen, Denmark
| | - David Bartrés-Faz
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and Institut de Neurociències, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Carl-Johan Boraxbekk
- Danish Research Centre for Magnetic Resonance (DRCMR), Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital-Amager and Hvidovre, 2650 Hvidovre, Copenhagen, Denmark
- Umeå Center for Functional Brain Imaging, Umeå University, 907 36 Umeå, Sweden
- Department of Radiation Sciences, Diagnostic Radiology, Umeå University, 907 36 Umeå, Sweden
- Institute of Sports Medicine Copenhagen (ISMC), Copenhagen University Hospital Bispebjerg, 2400 Copenhagen, Denmark
- Institute for Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, 2020 Copenhagen, Denmark
| | - Andreas M Brandmaier
- Center for Lifespan Psychology, Max Planck Institute for Human Development, 14195 Berlin, Germany
- Department of Psychology, MSB Medical School Berlin, Berlin, Germany
| | - Ilja Demuth
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolic Diseases (including Division of Lipid Metabolism), Biology of Aging working group, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10178 Berlin, Germany
- BCRT - Berlin Institute of Health Center for Regenerative Therapies, 13353 Berlin, Germany
| | - Christian A Drevon
- Vitas AS, The Science Park, 0349 Oslo, Norway
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of 0372 Oslo, Norway
| | - Klaus P Ebmeier
- Department of Psychiatry, University of Oxford, Oxford OX3 7JX, United Kingdom
| | - Paolo Ghisletta
- Faculty of Psychology and Educational Sciences, University of Geneva, 1205 Geneva, Switzerland
- UniDistance Suisse, 3900 Brig, Switzerland
- Swiss National Centre of Competence in Research LIVES, University of Geneva, 1205 Geneva, Switzerland
| | - Rogier Kievit
- Cognitive Neuroscience Department, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Simone Kühn
- Center for Lifespan Psychology, Max Planck Institute for Human Development, 14195 Berlin, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Kathrine Skak Madsen
- Danish Research Centre for Magnetic Resonance (DRCMR), Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital-Amager and Hvidovre, 2650 Hvidovre, Copenhagen, Denmark
- Radiography, Department of Technology, University College Copenhagen, 1799 Copenhagen, Denmark
| | - Lars Nyberg
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, 0373 Oslo, Norway
- Umeå Center for Functional Brain Imaging, Umeå University, 907 36 Umeå, Sweden
| | - Cristina Solé-Padullés
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and Institut de Neurociències, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Didac Vidal-Piñeiro
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, 0373 Oslo, Norway
| | - Gerd Wagner
- Department of Psychiatry and Psychotherapy, Jena University Hospital, 07743 Jena, Germany
| | - Leiv Otto Watne
- Oslo Delirium Research Group, Oslo University Hospital, 0424 Oslo, Norway
- Department of Geriatric Medicine, Akershus University Hospital, 1478 Lørenskog, Norway
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, 1478, Lørenskog, Norway
| | - Kristine B Walhovd
- Center for Lifespan Changes in Brain and Cognition, University of Oslo, 0373 Oslo, Norway
- Computational Radiology and Artificial Intelligence, Department of Radiology and Nuclear Medicine, Oslo University Hospital, 0424 Oslo, Norway
| |
Collapse
|
23
|
Que M, Li Y, Wang X, Zhan G, Luo X, Zhou Z. Role of astrocytes in sleep deprivation: accomplices, resisters, or bystanders? Front Cell Neurosci 2023; 17:1188306. [PMID: 37435045 PMCID: PMC10330732 DOI: 10.3389/fncel.2023.1188306] [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: 03/17/2023] [Accepted: 06/05/2023] [Indexed: 07/13/2023] Open
Abstract
Sleep plays an essential role in all studied animals with a nervous system. However, sleep deprivation leads to various pathological changes and neurobehavioral problems. Astrocytes are the most abundant cells in the brain and are involved in various important functions, including neurotransmitter and ion homeostasis, synaptic and neuronal modulation, and blood-brain barrier maintenance; furthermore, they are associated with numerous neurodegenerative diseases, pain, and mood disorders. Moreover, astrocytes are increasingly being recognized as vital contributors to the regulation of sleep-wake cycles, both locally and in specific neural circuits. In this review, we begin by describing the role of astrocytes in regulating sleep and circadian rhythms, focusing on: (i) neuronal activity; (ii) metabolism; (iii) the glymphatic system; (iv) neuroinflammation; and (v) astrocyte-microglia cross-talk. Moreover, we review the role of astrocytes in sleep deprivation comorbidities and sleep deprivation-related brain disorders. Finally, we discuss potential interventions targeting astrocytes to prevent or treat sleep deprivation-related brain disorders. Pursuing these questions would pave the way for a deeper understanding of the cellular and neural mechanisms underlying sleep deprivation-comorbid brain disorders.
Collapse
Affiliation(s)
- Mengxin Que
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yujuan Li
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Wang
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Gaofeng Zhan
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiao Luo
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Zhou
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
Steffey MA, Scharf VF, Risselada M, Buote NJ, Griffon D, Winter AL, Zamprogno H. A narrative review of the pathophysiology and impacts of insufficient and disrupted sleep. THE CANADIAN VETERINARY JOURNAL = LA REVUE VETERINAIRE CANADIENNE 2023; 64:579-587. [PMID: 37265804 PMCID: PMC10204879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Background Despite substantial ramifications of insufficient sleep on mental and physical health and general well-being, many individuals are unaware of what constitutes sufficient sleep, or of the short- and long-term extent of sleep deficiency effects, including those that may not be perceived as fatigue. Objectives and procedures This review describes the physiology of sleep, defines healthy standards, reviews the pathophysiology and health hazards of acute and chronic sleep insufficiency, and offers concepts for improving individual sleep hygiene. Online databases were searched to extract literature pertaining to sleep, sleep insufficiency, fatigue, and health, with emphasis on literature published in the preceding 5 years. Results The detrimental effects of acute and chronic sleep loss vary in their range and impact. Individuals often obtain a substandard quantity of sleep, a problem that is poorly recognized by individuals and society. This lack of recognition perpetuates a culture in which sleep insufficiency is accepted, resulting in serious and substantial negative impacts on mental and physical health. Conclusion and clinical relevance Sleep management is one of the most fundamental and changeable aspects of personal health. Improving awareness of the important physiological roles of sleep, healthy sleep habits, and the consequence of insufficient sleep is essential in promoting general well-being and mental and physical health.
Collapse
Affiliation(s)
- Michele A Steffey
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, 1 Shields Avenue, Davis, California 95616, USA (Steffey); Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, North Carolina 27607, USA (Scharf ); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, Indiana 47907, USA (Risselada); Department of Clinical Sciences, Cornell University College of Veterinary Medicine, 930 Campus Road, Ithaca, New York 14853, USA, (Buote); College of Veterinary Medicine, Western University of Health Sciences, 309 East Second Street, Pomona, California 91766, USA (Griffon); Merck Manuals Department, Merck Sharp & Dohme Corp., Rahway, New Jersey 07065, USA (Winter); Surgery Department, Evidensia Oslo Dyresykehus, Ensjøveien 14, 0655, Oslo, Norway (Zamprogno)
| | - Valery F Scharf
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, 1 Shields Avenue, Davis, California 95616, USA (Steffey); Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, North Carolina 27607, USA (Scharf ); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, Indiana 47907, USA (Risselada); Department of Clinical Sciences, Cornell University College of Veterinary Medicine, 930 Campus Road, Ithaca, New York 14853, USA, (Buote); College of Veterinary Medicine, Western University of Health Sciences, 309 East Second Street, Pomona, California 91766, USA (Griffon); Merck Manuals Department, Merck Sharp & Dohme Corp., Rahway, New Jersey 07065, USA (Winter); Surgery Department, Evidensia Oslo Dyresykehus, Ensjøveien 14, 0655, Oslo, Norway (Zamprogno)
| | - Marije Risselada
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, 1 Shields Avenue, Davis, California 95616, USA (Steffey); Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, North Carolina 27607, USA (Scharf ); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, Indiana 47907, USA (Risselada); Department of Clinical Sciences, Cornell University College of Veterinary Medicine, 930 Campus Road, Ithaca, New York 14853, USA, (Buote); College of Veterinary Medicine, Western University of Health Sciences, 309 East Second Street, Pomona, California 91766, USA (Griffon); Merck Manuals Department, Merck Sharp & Dohme Corp., Rahway, New Jersey 07065, USA (Winter); Surgery Department, Evidensia Oslo Dyresykehus, Ensjøveien 14, 0655, Oslo, Norway (Zamprogno)
| | - Nicole J Buote
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, 1 Shields Avenue, Davis, California 95616, USA (Steffey); Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, North Carolina 27607, USA (Scharf ); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, Indiana 47907, USA (Risselada); Department of Clinical Sciences, Cornell University College of Veterinary Medicine, 930 Campus Road, Ithaca, New York 14853, USA, (Buote); College of Veterinary Medicine, Western University of Health Sciences, 309 East Second Street, Pomona, California 91766, USA (Griffon); Merck Manuals Department, Merck Sharp & Dohme Corp., Rahway, New Jersey 07065, USA (Winter); Surgery Department, Evidensia Oslo Dyresykehus, Ensjøveien 14, 0655, Oslo, Norway (Zamprogno)
| | - Dominique Griffon
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, 1 Shields Avenue, Davis, California 95616, USA (Steffey); Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, North Carolina 27607, USA (Scharf ); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, Indiana 47907, USA (Risselada); Department of Clinical Sciences, Cornell University College of Veterinary Medicine, 930 Campus Road, Ithaca, New York 14853, USA, (Buote); College of Veterinary Medicine, Western University of Health Sciences, 309 East Second Street, Pomona, California 91766, USA (Griffon); Merck Manuals Department, Merck Sharp & Dohme Corp., Rahway, New Jersey 07065, USA (Winter); Surgery Department, Evidensia Oslo Dyresykehus, Ensjøveien 14, 0655, Oslo, Norway (Zamprogno)
| | - Alexandra L Winter
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, 1 Shields Avenue, Davis, California 95616, USA (Steffey); Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, North Carolina 27607, USA (Scharf ); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, Indiana 47907, USA (Risselada); Department of Clinical Sciences, Cornell University College of Veterinary Medicine, 930 Campus Road, Ithaca, New York 14853, USA, (Buote); College of Veterinary Medicine, Western University of Health Sciences, 309 East Second Street, Pomona, California 91766, USA (Griffon); Merck Manuals Department, Merck Sharp & Dohme Corp., Rahway, New Jersey 07065, USA (Winter); Surgery Department, Evidensia Oslo Dyresykehus, Ensjøveien 14, 0655, Oslo, Norway (Zamprogno)
| | - Helia Zamprogno
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, 1 Shields Avenue, Davis, California 95616, USA (Steffey); Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, 1060 William Moore Drive, Raleigh, North Carolina 27607, USA (Scharf ); Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, Indiana 47907, USA (Risselada); Department of Clinical Sciences, Cornell University College of Veterinary Medicine, 930 Campus Road, Ithaca, New York 14853, USA, (Buote); College of Veterinary Medicine, Western University of Health Sciences, 309 East Second Street, Pomona, California 91766, USA (Griffon); Merck Manuals Department, Merck Sharp & Dohme Corp., Rahway, New Jersey 07065, USA (Winter); Surgery Department, Evidensia Oslo Dyresykehus, Ensjøveien 14, 0655, Oslo, Norway (Zamprogno)
| |
Collapse
|
25
|
Gaudier-Diaz MM, Parekh SV, Penton RE, Robertson SD, Thomas A. Sleepy Mice Case Study: Implementation and Assessment. JOURNAL OF UNDERGRADUATE NEUROSCIENCE EDUCATION : JUNE : A PUBLICATION OF FUN, FACULTY FOR UNDERGRADUATE NEUROSCIENCE 2023; 21:A108-A116. [PMID: 37588653 PMCID: PMC10426825 DOI: 10.59390/rhsn3470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/10/2022] [Accepted: 10/14/2023] [Indexed: 08/18/2023]
Abstract
Case studies are a valuable teaching tool to engage students in course content using real-world scenarios. As part of the High-throughput Discovery Science & Inquiry-based Case Studies for Today's Students (HITS) Research Coordination Network (RCN), our team has created the Sleepy Mice Case Study for students to engage with RStudio and the Allen Institute for Brain Science's open access high-throughput sleep dataset on mice. Sleep is important for health, a familiar concern to college students, and was a basis for this case study. In this case, students completed an initial homework assignment, in-class work, and a final take-home application assignment. The case study was implemented in synchronous and asynchronous Introductory Neuroscience courses, a Biopsychology course, and a Human Anatomy and Physiology course, reflecting its versatility. The case can be used to teach course-specific learning objectives such as sleep-related content and/or science data processing skills. The case study was successful as shown by gains in student scores and confidence in achieving learning objectives. Most students reported enjoying learning about sleep deprivation course content using the case study. Best practices based on instructor experiences in implementation are also included to facilitate future use so that the Sleepy Mice Case Study can be used to teach content and/or research-related skills in various courses and modalities.
Collapse
Affiliation(s)
- Monica M. Gaudier-Diaz
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799
| | - Shveta V. Parekh
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799
| | - Rachel E. Penton
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799
| | - Sabrina D. Robertson
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799
| | - Aeisha Thomas
- Department of Biological and Health Sciences, Crown College, St. Bonifacius, MN 55375
| |
Collapse
|
26
|
Milman NE, Tinsley CE, Raju RM, Lim MM. Loss of sleep when it is needed most - Consequences of persistent developmental sleep disruption: A scoping review of rodent models. Neurobiol Sleep Circadian Rhythms 2023; 14:100085. [PMID: 36567958 PMCID: PMC9768382 DOI: 10.1016/j.nbscr.2022.100085] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Sleep is an essential component of development. Developmental sleep disruption (DSD) impacts brain maturation and has been associated with significant consequences on socio-emotional development. In humans, poor sleep during infancy and adolescence affects neurodevelopmental outcomes and may be a risk factor for the development of autism spectrum disorder (ASD) or other neuropsychiatric illness. Given the wide-reaching and enduring consequences of DSD, identifying underlying mechanisms is critical to best inform interventions with translational capacity. In rodents, studies have identified some mechanisms and neural circuits by which DSD causes later social, emotional, sensorimotor, and cognitive changes. However, these studies spanned methodological differences, including different developmental timepoints for both sleep disruption and testing, different DSD paradigms, and even different rodent species. In this scoping review on DSD in rodents, we synthesize these various studies into a cohesive framework to identify common neural mechanisms underlying DSD-induced dysfunction in brain and behavior. Ultimately, this review serves the goal to inform the generation of novel translational interventions for human developmental disorders featuring sleep disruption.
Collapse
Affiliation(s)
- Noah E.P. Milman
- Oregon Health and Science University, Dept. of Behavioral and Systems Neuroscience, Portland, OR, 97214, USA
- Veterans Affairs Portland Health Care System, Portland, OR, 97214, USA
| | - Carolyn E. Tinsley
- Oregon Health and Science University, Dept. of Behavioral and Systems Neuroscience, Portland, OR, 97214, USA
- Veterans Affairs Portland Health Care System, Portland, OR, 97214, USA
| | - Ravikiran M. Raju
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Miranda M. Lim
- Oregon Health and Science University, Dept. of Behavioral and Systems Neuroscience, Portland, OR, 97214, USA
- Veterans Affairs Portland Health Care System, Portland, OR, 97214, USA
| |
Collapse
|
27
|
Lodovichi C, Ratto GM. Control of circadian rhythm on cortical excitability and synaptic plasticity. Front Neural Circuits 2023; 17:1099598. [PMID: 37063387 PMCID: PMC10098176 DOI: 10.3389/fncir.2023.1099598] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/09/2023] [Indexed: 04/18/2023] Open
Abstract
Living organisms navigate through a cyclic world: activity, feeding, social interactions are all organized along the periodic succession of night and day. At the cellular level, periodic activity is controlled by the molecular machinery driving the circadian regulation of cellular homeostasis. This mechanism adapts cell function to the external environment and its crucial importance is underlined by its robustness and redundancy. The cell autonomous clock regulates cell function by the circadian modulation of mTOR, a master controller of protein synthesis. Importantly, mTOR integrates the circadian modulation with synaptic activity and extracellular signals through a complex signaling network that includes the RAS-ERK pathway. The relationship between mTOR and the circadian clock is bidirectional, since mTOR can feedback on the cellular clock to shift the cycle to maintain the alignment with the environmental conditions. The mTOR and ERK pathways are crucial determinants of synaptic plasticity and function and thus it is not surprising that alterations of the circadian clock cause defective responses to environmental challenges, as witnessed by the bi-directional relationship between brain disorders and impaired circadian regulation. In physiological conditions, the feedback between the intrinsic clock and the mTOR pathway suggests that also synaptic plasticity should undergo circadian regulation.
Collapse
Affiliation(s)
- Claudia Lodovichi
- Institute of Neuroscience, Consiglio Nazionale delle Ricerche (CNR), Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
- Padova Neuroscience Center, Universitá degli Studi di Padova, Padova, Italy
| | - Gian Michele Ratto
- Institute of Neuroscience, Consiglio Nazionale delle Ricerche (CNR), Padova, Italy
- Padova Neuroscience Center, Universitá degli Studi di Padova, Padova, Italy
- National Enterprise for NanoScience and NanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore, Pisa, Italy
| |
Collapse
|
28
|
Liu B, Li F, Xu Y, Wu Q, Shi J. Gastrodin Improves Cognitive Dysfunction in REM Sleep-Deprived Rats by Regulating TLR4/NF-κB and Wnt/β-Catenin Signaling Pathways. Brain Sci 2023; 13:brainsci13020179. [PMID: 36831722 PMCID: PMC9954436 DOI: 10.3390/brainsci13020179] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/15/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Gastrodin is the active ingredient in Gastrodia elata. Our previous studies demonstrated that gastrodin ameliorated cerebral ischemia-reperfusion and hypoperfusion injury and improved cognitive deficit in Alzheimer's disease. This study aims to examine the effects of gastrodin on REM sleep deprivation in rats. Gastrodin (100 and 150 mg/kg) was orally administered for 7 consecutive days before REM sleep deprivation. Seventy-two hours later, pentobarbital-induced sleep tests and a Morris water maze were performed to measure REM sleep quality and learning and memory ability. Histopathology was observed with hematoxylin-eosin staining, and the expression of the NF-κB and Wnt/β-catenin signaling pathways was examined using Western blot. After REM sleep deprivation, sleep latency increased and sleep duration decreased, and the ability of learning and memory was impaired. Neurons in the hippocampal CA1 region and the cortex were damaged. Gastrodin treatment significantly improved REM sleep-deprivation-induced sleep disturbance, cognitive deficits and neuron damage in the hippocampus CA1 region and cerebral cortex. A mechanism analysis revealed that the NF-κB pathway was activated and the Wnt/β-catenin pathway was inhibited after REM sleep deprivation, and gastrodin ameliorated these aberrant changes. Gastrodin improves REM sleep-deprivation-induced sleep disturbance and cognitive dysfunction by regulating the TLR4/NF-κB and Wnt/β-catenin signaling pathways and can be considered a potential candidate for the treatment of REM sleep deprivation.
Collapse
|
29
|
Beaugrand M, Muehlematter C, Markovic A, Camos V, Kurth S. Sleep as a protective factor of children's executive functions: A study during COVID-19 confinement. PLoS One 2023; 18:e0279034. [PMID: 36630329 PMCID: PMC9833525 DOI: 10.1371/journal.pone.0279034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/29/2022] [Indexed: 01/12/2023] Open
Abstract
Confinements due to the COVID-19 outbreak affected sleep and mental health of adults, adolescents and children. Already preschool children experienced acutely worsened sleep, yet the possible resulting effects on executive functions remain unexplored. Longitudinally, sleep quality predicts later behavioral-cognitive outcomes. Accordingly, we propose children's sleep behavior as essential for healthy cognitive development. By using the COVID-19 confinement as an observational-experimental intervention, we tested whether worsened children's sleep affects executive functions outcomes 6 months downstream. We hypothesized that acutely increased night awakenings and sleep latency relate to reduced later executive functions. With an online survey during the acute confinement phase we analyzed sleep behavior in 45 children (36-72 months). A first survey referred to the (retrospective) time before and (acute) situation during confinement, and a follow-up survey assessed executive functions 6 months later (6 months retrospectively). Indeed, acutely increased nighttime awakenings related to reduced inhibition at FOLLOW-UP. Associations were specific to the confinement-induced sleep-change and not the sleep behavior before confinement. These findings highlight that specifically acute changes of children's nighttime sleep during sensitive periods are associated with behavioral outcome consequences. This aligns with observations in animals that inducing poor sleep during developmental periods affects later brain function.
Collapse
Affiliation(s)
| | | | - Andjela Markovic
- University of Fribourg, Department of Psychology, Fribourg, Switzerland
- University Hospital Zurich, Department of Pulmonology, Zurich, Switzerland
| | - Valérie Camos
- University of Fribourg, Department of Psychology, Fribourg, Switzerland
| | - Salome Kurth
- University of Fribourg, Department of Psychology, Fribourg, Switzerland
- University Hospital Zurich, Department of Pulmonology, Zurich, Switzerland
| |
Collapse
|
30
|
Portillo E, Zi X, Kim Y, Tucker LB, Fu A, Miller LA, Valenzuela KS, Sullivan GM, Gauff AK, Yu F, Radomski KL, McCabe JT, Armstrong RC. Persistent hypersomnia following repetitive mild experimental traumatic brain injury: Roles of chronic stress and sex differences. J Neurosci Res 2023; 101:843-865. [PMID: 36624699 DOI: 10.1002/jnr.25165] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/15/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023]
Abstract
Traumatic brain injury (TBI) is often more complicated than a single head injury. An extreme example of this point may be military service members who experience a spectrum of exposures over a prolonged period under stressful conditions. Understanding the effects of complex exposures can inform evaluation and care to prevent persistent symptoms. We designed a longitudinal series of non-invasive procedures in adult mice to evaluate the effects of prolonged mild stress and head injury exposures. We assessed anxiety, depression, and sleep-wake dysfunction as symptoms that impact long-term outcomes after mild TBI. Unpredictable chronic mild stress (UCMS) was generated from a varied sequence of environmental stressors distributed within each of 21 days. Subsequently, mice received a mild blast combined with closed-head mild TBI on 5 days at 24-h intervals. In males and females, UCMS induced anxiety without depressive behavior. A major finding was reproducible sleep-wake dysfunction through 6- to 12-month time points in male mice that received UCMS with repetitive blast plus TBI events, or surprisingly after just UCMS alone. Specifically, male mice exhibited hypersomnia with increased sleep during the active/dark phase and fragmentation of longer wake bouts. Sleep-wake dysfunction was not found with TBI events alone, and hypersomnia was not found in females under any conditions. These results identify prolonged stress and sex differences as important considerations for sleep-wake dysfunction. Furthermore, this reproducible hypersomnia with impaired wakefulness is similar to the excessive daytime sleepiness reported in patients, including patients with TBI, which warrants further clinical screening, care, and treatment development.
Collapse
Affiliation(s)
- Edwin Portillo
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,The Center for Neuroscience and Regenerative Medicine, Bethesda, Maryland, USA
| | - Xiaomei Zi
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,The Center for Neuroscience and Regenerative Medicine, Bethesda, Maryland, USA
| | - Yeonho Kim
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Preclinical Behavior and Modeling Core, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Laura B Tucker
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Preclinical Behavior and Modeling Core, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Amanda Fu
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Preclinical Behavior and Modeling Core, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Lauren A Miller
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,The Center for Neuroscience and Regenerative Medicine, Bethesda, Maryland, USA
| | - Krystal S Valenzuela
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,The Center for Neuroscience and Regenerative Medicine, Bethesda, Maryland, USA
| | - Genevieve M Sullivan
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,The Center for Neuroscience and Regenerative Medicine, Bethesda, Maryland, USA
| | - Amina K Gauff
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,The Center for Neuroscience and Regenerative Medicine, Bethesda, Maryland, USA
| | - Fengshan Yu
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,The Center for Neuroscience and Regenerative Medicine, Bethesda, Maryland, USA
| | - Kryslaine L Radomski
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,The Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,The Center for Neuroscience and Regenerative Medicine, Bethesda, Maryland, USA
| | - Joseph T McCabe
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,The Center for Neuroscience and Regenerative Medicine, Bethesda, Maryland, USA.,Preclinical Behavior and Modeling Core, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Regina C Armstrong
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,The Center for Neuroscience and Regenerative Medicine, Bethesda, Maryland, USA
| |
Collapse
|
31
|
Calderón-Garcidueñas L, Torres-Jardón R, Greenough GP, Kulesza R, González-Maciel A, Reynoso-Robles R, García-Alonso G, Chávez-Franco DA, García-Rojas E, Brito-Aguilar R, Silva-Pereyra HG, Ayala A, Stommel EW, Mukherjee PS. Sleep matters: Neurodegeneration spectrum heterogeneity, combustion and friction ultrafine particles, industrial nanoparticle pollution, and sleep disorders-Denial is not an option. Front Neurol 2023; 14:1117695. [PMID: 36923490 PMCID: PMC10010440 DOI: 10.3389/fneur.2023.1117695] [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: 12/06/2022] [Accepted: 02/01/2023] [Indexed: 03/02/2023] Open
Abstract
Sustained exposures to ubiquitous outdoor/indoor fine particulate matter (PM2.5), including combustion and friction ultrafine PM (UFPM) and industrial nanoparticles (NPs) starting in utero, are linked to early pediatric and young adulthood aberrant neural protein accumulation, including hyperphosphorylated tau (p-tau), beta-amyloid (Aβ1 - 42), α-synuclein (α syn) and TAR DNA-binding protein 43 (TDP-43), hallmarks of Alzheimer's (AD), Parkinson's disease (PD), frontotemporal lobar degeneration (FTLD), and amyotrophic lateral sclerosis (ALS). UFPM from anthropogenic and natural sources and NPs enter the brain through the nasal/olfactory pathway, lung, gastrointestinal (GI) tract, skin, and placental barriers. On a global scale, the most important sources of outdoor UFPM are motor traffic emissions. This study focuses on the neuropathology heterogeneity and overlap of AD, PD, FTLD, and ALS in older adults, their similarities with the neuropathology of young, highly exposed urbanites, and their strong link with sleep disorders. Critical information includes how this UFPM and NPs cross all biological barriers, interact with brain soluble proteins and key organelles, and result in the oxidative, endoplasmic reticulum, and mitochondrial stress, neuroinflammation, DNA damage, protein aggregation and misfolding, and faulty complex protein quality control. The brain toxicity of UFPM and NPs makes them powerful candidates for early development and progression of fatal common neurodegenerative diseases, all having sleep disturbances. A detailed residential history, proximity to high-traffic roads, occupational histories, exposures to high-emission sources (i.e., factories, burning pits, forest fires, and airports), indoor PM sources (tobacco, wood burning in winter, cooking fumes, and microplastics in house dust), and consumption of industrial NPs, along with neurocognitive and neuropsychiatric histories, are critical. Environmental pollution is a ubiquitous, early, and cumulative risk factor for neurodegeneration and sleep disorders. Prevention of deadly neurological diseases associated with air pollution should be a public health priority.
Collapse
Affiliation(s)
- Lilian Calderón-Garcidueñas
- College of Health, The University of Montana, Missoula, MT, United States.,Universidad del Valle de México, Mexico City, Mexico
| | - Ricardo Torres-Jardón
- Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Glen P Greenough
- Department of Neurology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Randy Kulesza
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, Erie, PA, United States
| | | | | | | | | | | | | | - Héctor G Silva-Pereyra
- Instituto Potosino de Investigación Científica y Tecnológica A.C., San Luis Potosi, Mexico
| | - Alberto Ayala
- Sacramento Metropolitan Air Quality Management District, Sacramento, CA, United States.,Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, WV, United States
| | - Elijah W Stommel
- Department of Neurology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Partha S Mukherjee
- Interdisciplinary Statistical Research Unit, Indian Statistical Institute, Kolkata, India
| |
Collapse
|
32
|
Evans AK, Defensor E, Shamloo M. Selective Vulnerability of the Locus Coeruleus Noradrenergic System and its Role in Modulation of Neuroinflammation, Cognition, and Neurodegeneration. Front Pharmacol 2022; 13:1030609. [PMID: 36532725 PMCID: PMC9748190 DOI: 10.3389/fphar.2022.1030609] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/14/2022] [Indexed: 05/13/2024] Open
Abstract
Locus coeruleus (LC) noradrenergic (NE) neurons supply the main adrenergic input to the forebrain. NE is a dual modulator of cognition and neuroinflammation. NE neurons of the LC are particularly vulnerable to degeneration both with normal aging and in neurodegenerative disorders. Consequences of this vulnerability can be observed in both cognitive impairment and dysregulation of neuroinflammation. LC NE neurons are pacemaker neurons that are active during waking and arousal and are responsive to stressors in the environment. Chronic overactivation is thought to be a major contributor to the vulnerability of these neurons. Here we review what is known about the mechanisms underlying this neuronal vulnerability and combinations of environmental and genetic factors that contribute to confer risk to these important brainstem neuromodulatory and immunomodulatory neurons. Finally, we discuss proposed and potential interventions that may reduce the overall risk for LC NE neuronal degeneration.
Collapse
Affiliation(s)
- Andrew K. Evans
- School of Medicine, Stanford University, Stanford, CA, United States
| | | | - Mehrdad Shamloo
- School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
33
|
Simon KC, Nadel L, Payne JD. The functions of sleep: A cognitive neuroscience perspective. Proc Natl Acad Sci U S A 2022; 119:e2201795119. [PMID: 36279445 PMCID: PMC9636951 DOI: 10.1073/pnas.2201795119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023] Open
Abstract
This Special Feature explores the various purposes served by sleep, describing current attempts to understand how the many functions of sleep are instantiated in neural circuits and cognitive structures. Our feature reflects current experts' opinions about, and insights into, the dynamic processes of sleep. In the last few decades, technological advances have supported the updated view that sleep plays an active role in both cognition and health. However, these roles are far from understood. This collection of articles evaluates the dynamic nature of sleep, how it evolves across the lifespan, becomes a competitive arena for memory systems through the influence of the autonomic system, supports the consolidation and integration of new memories, and how lucid dreams might originate. This set of papers highlights new approaches and insights that will lay the groundwork to eventually understand the full range of functions supported by sleep.
Collapse
Affiliation(s)
| | - Lynn Nadel
- Psychology and Cognitive Science, University of Arizona, Tucson, AZ 85721
| | - Jessica D. Payne
- Department of Psychology, University of Notre Dame, Notre Dame, IN 46556
| |
Collapse
|