1
|
Chen X, Yan X, Jing C, Fu B, Jin W, Zhang S, Wang M, Liu F, Sun L. Ginsenoside Rc maintains sleep rhythm homeostasis by alleviating oxidative stress. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156634. [PMID: 40203472 DOI: 10.1016/j.phymed.2025.156634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/22/2025] [Accepted: 03/08/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Sleep disorders significantly impact physical health and quality of life. However, the current treatment strategies have several limitations. Panax ginseng has been traditionally employed to calm the mind, but its active components and their mechanisms remain elusive. PURPOSE This study aimed to elucidate the sleep-improving actions of Panax ginseng active component, ginseng Rc, and its potential mechanisms. METHODS Sleep duration and rhythm of Drosophila were assessed via a behavior analysis system. Furthermore, the potential addictive side effects of ginsenoside Rc were assessed through capillary ingestion. Changes in mRNA levels of core clock genes and stress response-related genes were determined via RT-qPCR. In addition, the potential mechanisms underlying the efficacy of ginsenoside Rc were evaluated by transcriptomic methodologies. A molecular operating environment (MOE)-Dock simulation was conducted to predict the binding affinity between Pink1 and ginsenoside Rc and verified by surface plasmon resonance. Lastly, Western blotting was carried out to assess Sir2 expression and acetylation of brain proteins. RESULTS It was observed that ginsenoside Rc improved sleep duration, latency, fragmentation, and amplitude. Furthermore, it upregulated the expression of the clock gene and was not addictive or dependency-inducing. Moreover, it increased antioxidant-related gene expression and reduced stress-related gene expression. In addition, transcriptomic analysis demonstrated that ginsenoside Rc also upregulated autophagy-related genes. Mechanistic studies showed that it improves sleep homeostasis by activating the Pink1/Sir2 signaling pathway, reducing oxidative stress, and modulating protein acetylation levels. CONCLUSION This study identified ginsenoside Rc, a novel compound from ginseng, and revealed that it can maintain sleep homeostasis. Mechanistically, ginsenoside Rc alleviated oxidative stress by targeting Pink1 and Sir2. These findings provide evidence for the potential clinical application of ginsenoside Rc for treating sleep disorders.
Collapse
Affiliation(s)
- Xuenan Chen
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Xiuci Yan
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Chenxu Jing
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China; Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Baoyu Fu
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China; Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Wenqi Jin
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Shuai Zhang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Manying Wang
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Fangbing Liu
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China.
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China; Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Changchun, China.
| |
Collapse
|
2
|
Tian Y, Kang L, Ha NT, Deng J, Liu D. Hydrogen peroxide in midbrain sleep neurons regulates sleep homeostasis. Cell Metab 2025:S1550-4131(25)00254-2. [PMID: 40378838 DOI: 10.1016/j.cmet.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 01/13/2025] [Accepted: 04/22/2025] [Indexed: 05/19/2025]
Abstract
Sleep could protect animals from oxidative damage, yet the dynamic interplay between the redox state and sleep homeostasis remains unclear. Here, we show that acute sleep deprivation (SD) in mice caused a general increase in brain oxidation, particularly in sleep-promoting regions. In vivo imaging of intracellular hydrogen peroxide (H2O2) real-time dynamics revealed that in nigra sleep neurons, the increase in cytosolic but not mitochondrial H2O2 reflects sleep debt and tracks spontaneous wakefulness by positively correlating with wake duration. By controllably manipulating intraneuronal H2O2, we discovered that H2O2 elevation is required for compensatory sleep and causally promotes sleep initiation, at least partly dependent on transient receptor potential melastatin 2 (TRPM2) channel. However, excessive H2O2 induced brain inflammation and sleep fragmentation. Together, our study demonstrates intraneuronal H2O2 as a crucial signaling molecule that translates brain redox imbalance into sleep drive and underscores the significance of oxidative eustress in sleep homeostasis.
Collapse
Affiliation(s)
- Yujing Tian
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Luwei Kang
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ngoc T Ha
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Juan Deng
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Danqian Liu
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
3
|
Ebrahim YM, Sadek MA, Sabry MO, Lotfy RM, El-Dessouki AM, Abou-Hussein D, El-Shiekh RA, ElBishbishy RM. Integrative sleep management: from molecular pathways to conventional and herbal treatments. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04183-y. [PMID: 40338321 DOI: 10.1007/s00210-025-04183-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/13/2025] [Indexed: 05/09/2025]
Abstract
Sleep is regarded as one of the most crucial factors in keeping a healthy lifestyle. To function normally, a person needs at least 6-8 h of sleep per day. Sleep influences not only our mood but also the efficiency with which we complete tasks. Sleep disorders exhibit diverse etiologies across different conditions and populations, with genetic and environmental factors playing a significant role in their development. Many issues emerge as a result of inadequate sleep. Unhealthy food and lifestyle choices have increased our susceptibility to sleep disorders. A well-balanced diet rich in essential vitamins and minerals can have a profound impact on sleep patterns, enhancing both the duration and quality of rest. The primary categories of sleep disorders include insomnia, sleep apnea (SA), narcolepsy, parasomnias, circadian rhythm disorders, and restless legs syndrome (RLS). The drugs used to treat sleep disorders are primarily habit-forming and have a history of withdrawal effects. This insufficiency in medication has prompted the hunt for newer, better options. Nutraceuticals are well-suited to the treatment of such illnesses. Its non-toxic, non-habit-forming properties, and practical efficiency have made it an outstanding choice. This review provides nutraceuticals used in sleep disorders. A comprehensive literature search was conducted utilizing several databases, including Google Scholar, Elsevier, Springer Nature, Wiley, PubMed, and EKB. Nutraceuticals are products that employ food or dietary components to treat or prevent disease. In the therapy of sleep disorders, nutraceuticals such as Artemisia annua, valerian, rosemary, jujube, Passionflower, lemon balm, ashwagandha, kava-kava, lavender, and chamomile have been shown to have remarkable benefits. These remedies exert their effects through multiple mechanisms, both directly by modulating neurotransmitter and hormonal pathways within sleep circuits, and indirectly by enhancing sleep quality through the alleviation of stress, inflammation, and oxidative stress. Clinical studies were piloted to validate the efficacy of natural sleep aids. Future research should focus on elucidating the precise mechanisms through which natural products influence sleep.
Collapse
Affiliation(s)
- Yasmina M Ebrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Mohamed A Sadek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Miral O Sabry
- Faculty of Science, National University of Singapore, Singapore Institute of Manufacturing Technology (SIMTech), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Rana M Lotfy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Ahmed M El-Dessouki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, 12566, Giza, Egypt
| | - Dina Abou-Hussein
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Rana M ElBishbishy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
4
|
Rorsman HO, Müller MA, Liu PZ, Sanchez LG, Kempf A, Gerbig S, Spengler B, Miesenböck G. Sleep pressure accumulates in a voltage-gated lipid peroxidation memory. Nature 2025; 641:232-239. [PMID: 40108451 PMCID: PMC12043502 DOI: 10.1038/s41586-025-08734-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 02/03/2025] [Indexed: 03/22/2025]
Abstract
Voltage-gated potassium (KV) channels contain cytoplasmically exposed β-subunits1-5 whose aldo-keto reductase activity6-8 is required for the homeostatic regulation of sleep9. Here we show that Hyperkinetic, the β-subunit of the KV1 channel Shaker in Drosophila7, forms a dynamic lipid peroxidation memory. Information is stored in the oxidation state of Hyperkinetic's nicotinamide adenine dinucleotide phosphate (NADPH) cofactor, which changes when lipid-derived carbonyls10-13, such as 4-oxo-2-nonenal or an endogenous analogue generated by illuminating a membrane-bound photosensitizer9,14, abstract an electron pair. NADP+ remains locked in the active site of KVβ until membrane depolarization permits its release and replacement with NADPH. Sleep-inducing neurons15-17 use this voltage-gated oxidoreductase cycle to encode their recent lipid peroxidation history in the collective binary states of their KVβ subunits; this biochemical memory influences-and is erased by-spike discharges driving sleep. The presence of a lipid peroxidation sensor at the core of homeostatic sleep control16,17 suggests that sleep protects neuronal membranes against oxidative damage. Indeed, brain phospholipids are depleted of vulnerable polyunsaturated fatty acyl chains after enforced waking, and slowing the removal of their carbonylic breakdown products increases the demand for sleep.
Collapse
Affiliation(s)
- H Olof Rorsman
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
| | - Max A Müller
- Institute of Inorganic and Analytical Chemistry, Justus-Liebig-Universität, Giessen, Germany
| | - Patrick Z Liu
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
| | | | - Anissa Kempf
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
- Biozentrum, Universität Basel, Basel, Switzerland
| | - Stefanie Gerbig
- Institute of Inorganic and Analytical Chemistry, Justus-Liebig-Universität, Giessen, Germany
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus-Liebig-Universität, Giessen, Germany
| | - Gero Miesenböck
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK.
| |
Collapse
|
5
|
Feeney SP, McCarthy JM, Petruconis CR, Tudor JC. Sleep loss is a metabolic disorder. Sci Signal 2025; 18:eadp9358. [PMID: 40198749 DOI: 10.1126/scisignal.adp9358] [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: 07/27/2024] [Accepted: 03/12/2025] [Indexed: 04/10/2025]
Abstract
Sleep loss dysregulates cellular metabolism and energy homeostasis. Highly metabolically active cells, such as neurons, enter a catabolic state during periods of sleep loss, which consequently disrupts physiological functioning. Specific to the central nervous system, sleep loss results in impaired synaptogenesis and long-term memory, effects that are also characteristic of neurodegenerative diseases. In this review, we describe how sleep deprivation increases resting energy expenditure, leading to the development of a negative energy balance-a state with insufficient metabolic resources to support energy expenditure-in highly active cells like neurons. This disruption of energetic homeostasis alters the balance of metabolites, including adenosine, lactate, and lipid peroxides, such that energetically costly processes, such as synapse formation, are attenuated. During sleep loss, metabolically active cells shunt energetic resources away from those processes that are not acutely essential, like memory formation, to support cell survival. Ultimately, these findings characterize sleep loss as a metabolic disorder.
Collapse
Affiliation(s)
- Sierra P Feeney
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Jordan M McCarthy
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Cecilia R Petruconis
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Jennifer C Tudor
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| |
Collapse
|
6
|
Horn CJ, Yuli S, Berry JA, Luong LT. A male-killing Spiroplasma endosymbiont has age-mediated impacts on Drosophila endurance and sleep. JOURNAL OF INSECT PHYSIOLOGY 2025; 161:104723. [PMID: 39551154 DOI: 10.1016/j.jinsphys.2024.104723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024]
Abstract
Endosymbiotic bacteria have a wide range of impacts on host physiology, behavior, metabolism, endurance, and mobility. Recent work found some endosymbionts also impact host sleep duration and quality. These effects may increase as flies age and endosymbiont titers increase. We tested the hypothesis that Spiroplasma poulsonni MSRO negatively impacts sleep in Drosophila melanogaster, and this in turn impairs fly endurance. In geotaxis climbing assays (a proxy for endurance), we found that MSRO impacted climbing endurance but in an age-dependent manner. Among younger flies, MSRO+ flies slept significantly less during dark periods (measured by a Drosophila Activity Monitoring System) compared to uninfected flies, but older MSRO+ flies did not show significant differences in amount of sleep compared to uninfected flies in the same cohort. While MSRO status impacted both sleep and endurance of hosts, endosymbiont-mediated sleep deprivation did not directly explain decreases in fly endurance. We discuss these results in the context of endosymbiont comparative biology.
Collapse
Affiliation(s)
- Collin J Horn
- Dalhousie University, Department of Psychology and Neuroscience, Canada; University of Alberta, Department of Biological Sciences, Canada.
| | - Sissi Yuli
- University of Alberta, Department of Biological Sciences, Canada
| | - Jacob A Berry
- University of Alberta, Department of Biological Sciences, Canada
| | - Lien T Luong
- University of Alberta, Department of Biological Sciences, Canada
| |
Collapse
|
7
|
Huang S, Piao C, Zhao Z, Beuschel CB, Turrel O, Toppe D, Sigrist SJ. Enhanced memory despite severe sleep loss in Drosophila insomniac mutants. PLoS Biol 2025; 23:e3003076. [PMID: 40111981 DOI: 10.1371/journal.pbio.3003076] [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: 02/03/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Sleep is crucial for cognitive functions and life span across species. While sleep homeostasis and cognitive processes are linked through cellular and synaptic plasticity, the signaling pathways connecting them remain unclear. Here, we show that Drosophila insomniac (inc) short sleep mutants, which lack an adaptor protein for the autism-associated Cullin-3 ubiquitin ligase, exhibited enhanced Pavlovian aversive olfactory learning and memory, unlike other sleep mutants with normal or reduced memory. Through a genetic modifier screen, we found that a mild reduction of Protein Kinase A (PKA) signaling specifically rescued the sleep and longevity phenotypes of inc mutants. However, this reduction further increased their excessive memory and mushroom body overgrowth. Since inc mutants displayed higher PKA signaling, we propose that inc loss-of-function suppresses sleep via increased PKA activity, which also constrains the excessive memory of inc mutants. Our data identify a signaling cascade for balancing sleep and memory functions, and provide a plausible explanation for the sleep phenotypes of inc mutants, suggesting that memory hyperfunction can provoke sleep deficits.
Collapse
Affiliation(s)
- Sheng Huang
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Chengji Piao
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Zhiying Zhao
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Christine B Beuschel
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Oriane Turrel
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - David Toppe
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
8
|
Nan M, Wang JB, Siokis M, St. Leger RJ. Latitudinal Clines in Climate and Sleep Patterns Shape Disease Outcomes in Drosophila melanogaster Infected by Metarhizium anisopliae. Ecol Evol 2025; 15:e71047. [PMID: 40027417 PMCID: PMC11868735 DOI: 10.1002/ece3.71047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/31/2025] [Accepted: 02/12/2025] [Indexed: 03/05/2025] Open
Abstract
Major latitudinal clines have been observed in Drosophila melanogaster, a human commensal that originated in tropical Africa and has subsequently dispersed globally to colonize temperate habitats. However, despite the crucial role pathogens play in species distribution, our understanding of how geographical factors influence disease susceptibility remains limited. This investigation explored the effects of latitudinal clines and biomes on disease resistance using the common fly pathogen Metarhizium anisopliae and 43 global Drosophila melanogaster populations. The findings revealed correlations between disease resistance and latitudinal gradients of sleep duration, temperature, and humidity. Although enhanced defenses may be driven by fungal diversity at tropical latitudes, the most disease-resistant tropical males also showed the highest susceptibility to desiccation. This suggests potential trade-offs between abiotic stress resistance, necessary for survival in temperate habitats, and disease resistance. Furthermore, the study uncovered interactions between sex, mating status, sleep, and abiotic stresses, affecting disease resistance. Notably, longer-sleeping males and virgin flies survived infections longer, with additional daytime sleep post-infection being protective, particularly in the most resistant fly lines. These observations support the hypothesis that sleep and disease defense are intertwined traits linked to organismal fitness and subject to joint clinal evolution.
Collapse
Affiliation(s)
- Mintong Nan
- Department of EntomologyUniversity of MarylandCollege ParkMarylandUSA
| | - Jonathan B. Wang
- Department of EntomologyUniversity of MarylandCollege ParkMarylandUSA
| | - Michail Siokis
- Department of EntomologyUniversity of MarylandCollege ParkMarylandUSA
| | | |
Collapse
|
9
|
Kim S, Kang SW, Kim SE, Kim HJ, Kim SA, Lee YW, Kim EY, Shin C, Lee HW. Genome-wide identification and functional validation of the WW domain containing oxidoreductase gene associated with sleep duration. Sci Rep 2025; 15:5552. [PMID: 39952983 PMCID: PMC11828923 DOI: 10.1038/s41598-024-81158-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 11/25/2024] [Indexed: 02/17/2025] Open
Abstract
Individual differences in sleep duration have been reported, and genetic components of sleep duration have been identified showing various heritability. To identify genetic variants that contribute to sleep duration, we conducted a human genome-wide identification on sleep duration and performed confirmatory experiments using a Drosophila model. Genome-wide association study in human was analyzed to determine the association of the genetic variants with self-aware sleep duration from two community-based cohort, Ansan (cohort 1, n = 4635) and Ansung (cohort 2, n = 4205), recruited from the Korean Genome and Epidemiology Study. Individual single nucleotide variants (rs16948804 and rs4887991) in the WW domain containing oxidoreductase (WWOX) gene were associated with self-aware sleep duration in human (p-values, 1.11 × 10- 7 and 2.05 × 10- 7, retrospectively). To examine the functional relevance of the WWOX gene identified in the genome-wide association study, we analyzed the sleep duration of Drosophila loss-of-function mutants. The deletion of Wwox in flies reduced sleep duration and quality with average bout length during daytime and increased night-time sleep duration (all of p-values < 0.01). Our findings suggested that WWOX expression is associated with sleep duration in both humans and Drosophila and genetic factors play a role in inter-individual variability in sleep characteristics.
Collapse
Affiliation(s)
- Soriul Kim
- Department of Paramedicine, Seowon University, Cheongju, South Korea
- Institute for Human Genomic Study, College of Medicine, Korea University, Seoul, South Korea
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - So Who Kang
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Song E Kim
- Departments of Neurology and Medical Science, Computational Medicine, System Health Science and Engineering and Artificial Intelligence Graduate Programs, Ewha Womans University School of Medicine and Ewha Medical Research Institute, 1071, Anyangcheon-ro, Yangcheon-gu, Seoul, 07985, South Korea
| | - Hyeon Jin Kim
- Institute for Human Genomic Study, College of Medicine, Korea University, Seoul, South Korea
- Department of Neurology, Asan Medical Center, Seoul, South Korea
| | - Sol Ah Kim
- Departments of Neurology and Medical Science, Computational Medicine, System Health Science and Engineering and Artificial Intelligence Graduate Programs, Ewha Womans University School of Medicine and Ewha Medical Research Institute, 1071, Anyangcheon-ro, Yangcheon-gu, Seoul, 07985, South Korea
| | - Yae Won Lee
- Departments of Neurology and Medical Science, Computational Medicine, System Health Science and Engineering and Artificial Intelligence Graduate Programs, Ewha Womans University School of Medicine and Ewha Medical Research Institute, 1071, Anyangcheon-ro, Yangcheon-gu, Seoul, 07985, South Korea
| | - Eun Young Kim
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Chol Shin
- Institute for Human Genomic Study, College of Medicine, Korea University, Seoul, South Korea.
- Institute of Human Genomic Study, College of Medicine, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| | - Hyang Woon Lee
- Departments of Neurology and Medical Science, Computational Medicine, System Health Science and Engineering and Artificial Intelligence Graduate Programs, Ewha Womans University School of Medicine and Ewha Medical Research Institute, 1071, Anyangcheon-ro, Yangcheon-gu, Seoul, 07985, South Korea.
- Computational Medicine, System Health Science and Engineering, Ewha Womans University, Seoul, South Korea.
- Artificial Intelligence Convergence Graduate Programs, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
10
|
Avila A, Lewandowski AS, Li Y, Gui J, Lee KA, Yang Z, Kim M, Lyles JT, Man K, Sehgal A, Chandler JD, Zhang SL. A carnitine transporter at the blood-brain barrier modulates sleep via glial lipid metabolism in Drosophila. Proc Natl Acad Sci U S A 2025; 122:e2421178122. [PMID: 39847335 PMCID: PMC11789159 DOI: 10.1073/pnas.2421178122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025] Open
Abstract
To regulate brain function, peripheral compounds must traverse the blood-brain barrier (BBB), an interface between the brain and the circulatory system. To determine whether specific transport mechanisms are relevant for sleep, we conducted a BBB-specific inducible RNAi knockdown (iKD) screen for genes affecting sleep in Drosophila. We observed reduced sleep with knockdown of solute carrier CG6126, a carnitine transporter, as determined by isotope flux. Our findings suggest that CG6126 regulation of sleep is through the role of the carnitine shuttle in regulating fatty acid metabolism as lipid droplets accumulate in the brains of CG6126 BBB iKD flies. Knocking down mitochondrial carnitine transferases in non-BBB glial cells mimicked the reduced sleep of the CG6126 BBB iKD flies, while bypassing the necessity of carnitine transport with dietary medium-chain fatty acids or palmitoylcarnitine rescued sleep. We propose that carnitine transport via CG6126 promotes brain fatty acid metabolism necessary for maintaining sleep.
Collapse
Affiliation(s)
- Ashley Avila
- Department of Cell Biology, Emory University, Atlanta, GA30322
| | | | - Yongjun Li
- HHMI, University of Pennsylvania, Philadelphia, PA19104
- Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA19104
| | - Jesse Gui
- Department of Cell Biology, Emory University, Atlanta, GA30322
| | - Kaeun A. Lee
- Department of Cell Biology, Emory University, Atlanta, GA30322
| | - Zhenglang Yang
- Department of Cell Biology, Emory University, Atlanta, GA30322
| | - Mari Kim
- Department of Cell Biology, Emory University, Atlanta, GA30322
| | - James T. Lyles
- Department of Pediatrics, Emory University, Atlanta, GA30322
| | - Kai Man
- Department of Cell Biology, Emory University, Atlanta, GA30322
| | - Amita Sehgal
- HHMI, University of Pennsylvania, Philadelphia, PA19104
- Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA19104
| | - Joshua D. Chandler
- Department of Pediatrics, Emory University, Atlanta, GA30322
- Children’s Healthcare of Atlanta, Emory University, Atlanta, GA30322
| | | |
Collapse
|
11
|
Ma H, Li J, Ma H. Associations of phenols, parabens, and phthalates with biological aging: stratified analyses by chronological age and lifestyle in NHANES 2005-2010. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2025:1-17. [PMID: 39801106 DOI: 10.1080/09603123.2025.2451626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/07/2025] [Indexed: 01/31/2025]
Abstract
Humans are widely exposed to phenols, parabens, and phthalates with health risks, while the effects of these chemicals on biological aging remain unclear. Among 3,441 adults in the National Health and Nutrition Examination Survey 2005-2010, phenol, paraben, and phthalate concentrations were measured and phenotypic age acceleration (PhenoAgeAccel) was calculated. Linear regression and weighted quantile sum (WQS) regression were used to evaluate the associations of single and mixed chemicals with PhenoAgeAccel. Stratified analyses by chronological age and lifestyle were also performed. Individual phthalates were positively associated with PhenoAgeAccel. The WQS model found the positive relationship between mixed chemicals with PhenoAgeAccel (β = 0.175, 95%CI: 0.001, 0.349). The adverse impacts of phenols and phthalates on biological aging were stronger in older participants with significant interactions. Adherence to healthier lifestyle might partly reduce the positive relationships of phenols and phthalates with biological aging, especially among older adults, which is expected to be a viable intervention in the future.
Collapse
Affiliation(s)
- Han Ma
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R.China
| | - Jinyue Li
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hanping Ma
- Office of Human Resources, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
12
|
Bongiovanni T, Santiago M, Zielinska K, Scheiman J, Barsa C, Jäger R, Pinto D, Rinaldi F, Giuliani G, Senatore T, Kostic AD. A Lactobacillus consortium provides insights into the sleep-exercise-microbiome nexus in proof of concept studies of elite athletes and in the general population. MICROBIOME 2025; 13:1. [PMID: 39748236 PMCID: PMC11697739 DOI: 10.1186/s40168-024-01936-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/18/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND The complex relationship among sleep, exercise, and the gut microbiome presents a unique opportunity to improve health and wellness. Here, we conducted the first large-scale investigation into the influence of a novel elite athlete-derived probiotic, consisting of a multi-strain Lactobacillus consortium, on sleep quality, exercise recovery, and gut microbiome composition in both elite athletes (n = 11) and the general population (n = 257). RESULTS Our two-phase study design, which included an open-label study followed by a controlled longitudinal study in a professional soccer team, allowed us to identify key interactions between probiotics, the gut microbiome, and the host. In the placebo-controlled study, we observed significant improvements in self-reported sleep quality by 69%, energy levels by 31%, and bowel movements by 37% after probiotic intervention relative to after placebo. These improvements were associated with a significant decrease in D-ROMS (a marker of oxidative stress) and a significantly higher free-testosterone/cortisol ratio. Multi-omics analyses revealed specific changes in microbiome composition and function, potentially providing mechanistic insights into these observed effects. CONCLUSION This study provides novel insights into how a multi-strain Lactobacillus probiotic modulates sleep quality, exercise recovery, and gut microbiome composition in both the general population and elite athletes, and introduces potential mechanisms through which this probiotic could be influencing overall health. Our results emphasize the untapped potential of tailored probiotic interventions derived from extremely fit and healthy individuals in improving several aspects of health and performance directly in humans. Video Abstract.
Collapse
Affiliation(s)
- Tindaro Bongiovanni
- Player Health and Performance, Palermo Football Club, Palermo, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | - Tullio Senatore
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | | |
Collapse
|
13
|
Giacon TA, Mrakic-Sposta S, Bosco G, Vezzoli A, Dellanoce C, Campisi M, Narici M, Paganini M, Foing B, Kołodziejczyk A, Martinelli M, Pavanello S. Environmental study and stress-related biomarkers modifications in a crew during analog astronaut mission EMMPOL 6. Eur J Appl Physiol 2025; 125:209-221. [PMID: 39320485 PMCID: PMC11753359 DOI: 10.1007/s00421-024-05575-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 08/12/2024] [Indexed: 09/26/2024]
Abstract
PURPOSE Human presence in space is increasingly frequent, but we must not forget that it is a hostile environment. We aimed to study the characteristics of experimental scenarios, to obtain data on human response to isolation, disruption of circadian rhythm and high levels of psychophysical stress. METHODS In these experiments, we evaluated stress response in five young healthy subjects inside an earth-based moon-settlement-like habitat during a 1-week long analog astronaut mission. Wearable devices were used to monitor daily step count of the subjects, physical activity, heart rate during physical exercise and at rest, and sleep parameters. From saliva and urine samples collected every day at awakening, we studied oxy-inflammation biomarkers and hormones (stress and appetite) were studied too. RESULTS At the end of the week, all subjects revealed an increase in oxidative stress and cortisol levels but no inflammation biomarkers variations, in conjunction with increasing time/daily exercise. Furthermore, a significant decrease in hours of sleep/day, sleep quality, and REM phase of sleep was recorded and correlated with the increase of reactive oxygen species. CONCLUSION Oxidative stress increased in a short period of time and may be attributed to the influence of psychological stress during confinement, as well as increased exercise and decreased amount of sleep. On a long-term basis, this could impact performance.
Collapse
Affiliation(s)
- T A Giacon
- Department of Biomedical Sciences, University of Padova, Via Marzolo 3, 35131, Padua, Italy
| | - Simona Mrakic-Sposta
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Piazza dell'Ospedale Maggiore, 3, 20162, Milan, Italy.
| | - G Bosco
- Department of Biomedical Sciences, University of Padova, Via Marzolo 3, 35131, Padua, Italy.
| | - A Vezzoli
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Piazza dell'Ospedale Maggiore, 3, 20162, Milan, Italy
| | - Cinzia Dellanoce
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Piazza dell'Ospedale Maggiore, 3, 20162, Milan, Italy
| | - M Campisi
- Occupational Medicine, Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
| | - M Narici
- Department of Biomedical Sciences, University of Padova, Via Marzolo 3, 35131, Padua, Italy
| | - M Paganini
- Department of Biomedical Sciences, University of Padova, Via Marzolo 3, 35131, Padua, Italy
| | - B Foing
- LUNEX EuroMoonMars, and EuroSpaceHub Academy, Leiden Observatory, Leiden, Netherlands
| | - A Kołodziejczyk
- Space Technology Centre, AGH University of Science and Technology, Kraków, Poland
- Analog Astronaut Training Centre, Kraków, Poland
| | - M Martinelli
- Institute of Science and Information Technologies "Alessandro Faedo", National Research Council (ISTI-CNR), Via G. Moruzzi 1, 56124, Pisa, Italy
| | - S Pavanello
- Occupational Medicine, Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
- University Center for Space Studies and Activities "Giuseppe Colombo"-CISAS, University of Padua, Padua, Italy
- University Hospital of Padova, Padua, Italy
| |
Collapse
|
14
|
Tener SJ, Kim CE, Lee J, Oraedu K, Gatto JA, Chang TY, Lam C, Schanta R, Jankowski MS, Park SJ, Hurley JM, Ulgherait M, Canman JC, Ja WW, Collins DB, Shirasu-Hiza M. Investigating the consequences of chronic short sleep for metabolism and survival of oxidative stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.01.626207. [PMID: 39677628 PMCID: PMC11642809 DOI: 10.1101/2024.12.01.626207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
In previous work, we found that short sleep caused sensitivity to oxidative stress; here we set out to characterize the physiological state of a diverse group of chronically short-sleeping mutants during hyperoxia as an acute oxidative stress. Using RNA-sequencing analysis, we found that short-sleeping mutants had a normal transcriptional oxidative stress response relative to controls. In both short-sleeping mutants and controls, hyperoxia led to downregulation of glycolytic genes and upregulation of genes involved in fatty acid metabolism, reminiscent of metabolic shifts during sleep. We hypothesized that short-sleeping mutants may be sensitive to hyperoxia because of defects in metabolism. Consistent with this, short-sleeping mutants were sensitive to starvation. Using metabolomics, we identified a pattern of low levels of long chain fatty acids and lysophospholipids in short-sleeping mutants relative to controls during hyperoxia, suggesting a defect in lipid metabolism. Though short-sleeping mutants did not have common defects in many aspects of lipid metabolism (basal fat stores, usage kinetics during hyperoxia, respiration rates, and cuticular hydrocarbon profiles), they were all sensitive to dehydration, suggesting a general defect in cuticular hydrocarbons, which protect against dehydration. To test the bi-directionality of sleep and lipid metabolism, we tested flies with both diet-induced obesity and genetic obesity. Flies with diet-induced obesity had no sleep or oxidative stress phenotype; in contrast, the lipid metabolic mutant, brummer , slept significantly more than controls but was sensitive to oxidative stress. Previously, all short sleepers tested were sensitive and all long sleepers resistant to oxidative stress. brummer mutants, the first exceptions to this rule, lack a key enzyme required to mobilize fat stores, suggesting that a defect in accessing lipid stores can cause sensitivity to oxidative stress. Taken together, we found that short-sleeping mutants have many phenotypes in common: sensitivity to oxidative stress, starvation, dehydration, and defects in lipid metabolites. These results argue against a specific role for sleep as an antioxidant and suggest the possibility that lipid metabolic defects underlie the sensitivity to oxidative stress of short-sleeping mutants.
Collapse
|
15
|
Yeewa R, Pohsa S, Yamsri T, Wongkummool W, Jantaree P, Potikanond S, Nimlamool W, Shotelersuk V, Lo Piccolo L, Jantrapirom S. The histone acylation reader ENL/AF9 regulates aging in Drosophila melanogaster. Neurobiol Aging 2024; 144:153-162. [PMID: 39405796 DOI: 10.1016/j.neurobiolaging.2024.10.002] [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: 03/04/2024] [Revised: 10/05/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024]
Abstract
Histone acylation plays a pivotal role in modulating gene expression, ensuring proper neurogenesis and responsiveness to various signals. Recently, the evolutionary conserved YAF9, ENL, AF9, TAF41, SAS5 (YEATS) domain found in four human paralogs, has emerged as a new class of histone acylation reader with a preference for the bulkier crotonyl group lysine over acetylation. Despite advancements, the role of either histone crotonylation or its readers in neurons remains unclear. In this study, we employed Drosophila melanogaster to investigate the role of ENL/AF9 (dENL/AF9) in the nervous system. Pan-neuronal dENL/AF9 knockdown not only extended the lifespan of flies but also enhanced their overall fitness during aging, including improved sleep quality and locomotion. Moreover, a decreased activity of dENL/AF9 in neurons led to an up-regulation of catalase gene expression which combined with reduced levels of malondialdehyde (MDA) and an enhanced tolerance to oxidative stress in aging flies. This study unveiled a novel function of histone crotonylation readers in aging with potential implications for understanding age-related conditions in humans.
Collapse
Affiliation(s)
- Ranchana Yeewa
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sureena Pohsa
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Titaree Yamsri
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Wasinee Wongkummool
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Phatcharida Jantaree
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Vorasuk Shotelersuk
- Centre of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Paediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Excellence Centre for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
| | - Luca Lo Piccolo
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Salinee Jantrapirom
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Drosophila Centre for Human Diseases and Drug Discovery (DHD), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
16
|
Otenaike TA, Farodoye OM, de Silva MM, Loreto JS, Adedara AO, Dos Santos MM, de Prestes AS, Barbosa NV, da Rocha JBT, Lobo LE, Wagner R, Abolaji AO, Loreto ELS. Nicotine and Vape: Drugs of the Same Profile Flock Together. J Biochem Mol Toxicol 2024; 38:e70075. [PMID: 39601203 DOI: 10.1002/jbt.70075] [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: 02/22/2024] [Revised: 09/10/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
Smoking, a major behavioral health burden, causes preventable and premature deaths globally. Nicotine, the addictive component present in tobacco products and Electronic cigarettes (E-cigarettes, vape), can bind to nicotinic acetylcholine receptors in the brain to trigger a dopamine release that reinforces smoking. Despite the widespread usage of nicotine, its mechanisms of toxicity, particularly in e-cigarettes, are poorly understood. Using Drosophila melanogaster as a model organism, this study aims to investigate the mechanism of the toxicity of nicotine and vape. Behavioral parameters, oxidative stress indicators, mRNA expression levels of Dopamine 1- receptor 1 (Dop1R1), Acetyl-coenzyme A synthetase (AcCoAs), and apoptotic proteins were assessed in the flies after a 5-day exposure to varying concentrations of nicotine (0.15, 0.25, and 0.35 mg/mL diet) and vape (0.06, 0.08, and 0.12 mg/mL diet). Furthermore, Gas Chromatography-Mass Spectrometry (GC/MS) and Gas Chromatography-Flame Ionization Detection (GC/FID) analyzes were conducted to gain more insight on the composition of the vape used in study. Findings indicate that both nicotine and vape exposure significantly reduced lifespan, impaired locomotor activity, and disrupted sleep patterns. Notably, nicotine exposure stimulated Dop1R1 transcription and altered Acetyl-CoA gene expression, impacting the viability and behavior of the flies. Elevated levels of reactive oxygen biomarkers were observed, contributing to cellular damage through oxidative stress and apoptotic mechanisms mediated by the Reaper and DIAP1 proteins. Additionally, the composition analysis of vape liquid revealed the presence of propylene glycol, nicotine, methyl esters, and an unidentified compound. This study highlights the complex interplay between nicotine, gene expression, and physiological responses in Drosophila.
Collapse
Affiliation(s)
- Titilayomi A Otenaike
- Doctoral Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), CEP, Porto Alegre, Brazil
- Drosophila Research and Training Centre, Ibadan, Nigeria
| | - Oluwabukola M Farodoye
- Doctoral Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), CEP, Porto Alegre, Brazil
- Drosophila Research and Training Centre, Ibadan, Nigeria
| | - Monica M de Silva
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Julia S Loreto
- Center for Natural and Exact Sciences, Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Adeola O Adedara
- Drosophila Research and Training Centre, Ibadan, Nigeria
- Center for Natural and Exact Sciences, Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Matheus M Dos Santos
- Center for Natural and Exact Sciences, Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Alessandro S de Prestes
- Center for Natural and Exact Sciences, Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Nilda V Barbosa
- Center for Natural and Exact Sciences, Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - João B T da Rocha
- Center for Natural and Exact Sciences, Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Luiz E Lobo
- Department of Technology and Food Science, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Roger Wagner
- Department of Technology and Food Science, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Amos O Abolaji
- Drosophila Research and Training Centre, Ibadan, Nigeria
- Drosophila Laboratory, Department of Biochemistry, College of Medicine, Molecular Drug Metabolism and Toxicology Unit, Ibadan, Nigeria
| | - Elgion L S Loreto
- Doctoral Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), CEP, Porto Alegre, Brazil
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| |
Collapse
|
17
|
Hao G, Zhao X, Fu W, Wu Y, Dai J, Qian Y, Xie T, Hou L, Shi W. The association between oxidative balance score and sleep duration: a mediation analysis of a cross-sectional study. Front Nutr 2024; 11:1423424. [PMID: 39588042 PMCID: PMC11586185 DOI: 10.3389/fnut.2024.1423424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/30/2024] [Indexed: 11/27/2024] Open
Abstract
Study objectives The Oxidative Balance Score (OBS), which reflects overall oxidation through diet and lifestyle, has been linked to sleep, but few studies have clarified this relationship. We investigated the association between OBS and sleep duration, and whether oxidative stress (OS) and inflammation mediate the underlying mechanisms. Methods Data were obtained from the National Health and Nutrition Examination Survey spanning the years 2007 to 2018. Multivariable logistic regression analyses were used to evaluate the association between OBS and the risk of sleep duration. Mediation analyses were conducted to investigate the role of OS and inflammatory markers. Results A significant negative association was found between OBS and sleep duration (p < 0.01). Meanwhile, compared to participants in OBS tertile 1, the ORs (95% CIs) of incident short sleep duration were 0.78 (0.72-0.86) and 0.72 (0.67-0.79) (both p < 0.01) for OBS tertile 2 and 3, respectively. And the ORs (95% CIs) of incident long sleep duration were 0.83 (0.73, 0.95) and 0.66 (0.57, 0.75) (both p < 0.01) for OBS tertiles 2 and 3 after adjustment for multivariate variables. A linear relationship between OBS and short/long sleep duration (p for non-linearity = 0.69/0.94, both p < 0.01) were revealed. Mediation analysis showed absolute neutrophil count, serum total bilirubin mediated the association between OBS and short/long sleep duration with 5.72, 13.41% proportion of mediation, respectively (both p < 0.001). Conclusion OBS is negatively associated with sleep duration. OS and inflammatory biomarkers mediate the relationship.
Collapse
Affiliation(s)
- Guihua Hao
- Department of Nursing, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- Shanghai Jiao Tong University School of Nursing, Shanghai, China
| | - Xiaomei Zhao
- Department of Nursing, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Weiwei Fu
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Yiwen Wu
- Department of Nursing, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jingjing Dai
- Department of Nursing, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yifeng Qian
- Department of Oral and Craniomaxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tian Xie
- Department of General Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lili Hou
- Department of Nursing, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wentao Shi
- Clinical Research Unit, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
18
|
Ortiz-Vega N, Lobato AG, Canic T, Zhu Y, Lazopulo S, Syed S, Zhai RG. Regulation of proteostasis by sleep through autophagy in Drosophila models of Alzheimer's disease. Life Sci Alliance 2024; 7:e202402681. [PMID: 39237365 PMCID: PMC11377308 DOI: 10.26508/lsa.202402681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024] Open
Abstract
Sleep and circadian rhythm dysfunctions are common clinical features of Alzheimer's disease (AD). Increasing evidence suggests that in addition to being a symptom, sleep disturbances can also drive the progression of neurodegeneration. Protein aggregation is a pathological hallmark of AD; however, the molecular pathways behind how sleep affects protein homeostasis remain elusive. Here we demonstrate that sleep modulation influences proteostasis and the progression of neurodegeneration in Drosophila models of tauopathy. We show that sleep deprivation enhanced Tau aggregational toxicity resulting in exacerbated synaptic degeneration. In contrast, sleep induction using gaboxadol led to reduced toxic Tau accumulation in neurons as a result of modulated autophagic flux and enhanced clearance of ubiquitinated Tau, suggesting altered protein processing and clearance that resulted in improved synaptic integrity and function. These findings highlight the complex relationship between sleep and regulation of protein homeostasis and the neuroprotective potential of sleep-enhancing therapeutics to slow the progression or delay the onset of neurodegeneration.
Collapse
Affiliation(s)
- Natalie Ortiz-Vega
- Department of Neurology, University of Chicago, Chicago, IL, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
- Graduate Program in Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Amanda G Lobato
- Department of Neurology, University of Chicago, Chicago, IL, USA
| | - Tijana Canic
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Physics, University of Miami, Coral Gables, FL, USA
| | - Yi Zhu
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Sheyum Syed
- Department of Physics, University of Miami, Coral Gables, FL, USA
| | - R Grace Zhai
- Department of Neurology, University of Chicago, Chicago, IL, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
19
|
Bond SM, Peralta AJ, Sirtalan D, Skeele DA, Huang H, Possidente DR, Vecsey CG. Differential regulation of sleep by blue, green, and red light in Drosophila melanogaster. Front Behav Neurosci 2024; 18:1476501. [PMID: 39539940 PMCID: PMC11557423 DOI: 10.3389/fnbeh.2024.1476501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Exposure to blue-enriched light from electronic devices is an emergent disruptor of human sleep, especially at particular times of day. Further dissection of this phenomenon necessitates modeling in a tractable model organism. Methods Thus, we investigated the effects of light color on sleep in Drosophila melanogaster. We measured sleep in red-eyed Canton-S (CS) and white-eyed w 1118 flies in baseline 12:12 light/dark conditions and experimental conditions with light-color (blue, red, or green) exposure for all 12 h of daylight or 3 h in the morning or evening. Results Blue light reduced daytime and nighttime sleep in CS but not in w 1118, potentially indicating a role for the compound eye in blue light's effects on fruit fly sleep. Red light, especially in the evening, reduced sleep during exposure in both strains. Green light had minimal effects on sleep in CS flies, but evening exposure reduced sleep in w 1118 flies, mimicking red light's effects. Discussion In conclusion, light's effects on sleep in D. melanogaster are dependent on wavelength and time-of-day. Future studies will aim to dissect these mechanisms genetically.
Collapse
Affiliation(s)
- Samuel M. Bond
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, United States
- Department of Neurobiology, UMass Chan Medical School, Worcester, MA, United States
| | - Aaliyah J. Peralta
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, United States
| | - Dilhan Sirtalan
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, United States
| | - Dominic A. Skeele
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, United States
| | - Haoyang Huang
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, United States
| | | | | |
Collapse
|
20
|
Zhang Y, Zhang C, Dai Q, Ma R. Continuous Theta Burst Stimulation Inhibits Oxidative Stress-Induced Inflammation and Autophagy in Hippocampal Neurons by Activating Glutathione Synthesis Pathway, Improving Cognitive Impairment in Sleep-Deprived Mice. Neuromolecular Med 2024; 26:40. [PMID: 39388015 DOI: 10.1007/s12017-024-08807-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/29/2024] [Indexed: 10/12/2024]
Abstract
Sleep deprivation (SD) has been reported to have a negative impact on cognitive function. Continuous theta burst stimulation (cTBS) shows certain effects in improving sleep and neurological diseases, and its molecular or cellular role in SD-induced cognition impairment still need further exploration. In this study, C57BL/6 mice were subjected to 48 h of SD and cTBS treatment, and cTBS treatment significantly improved SD-triggered impairment of spatial learning and memory abilities in mice. Additionally, cTBS reduced malondialdehyde levels, increased superoxide dismutase activities, and inhibited the production of inflammatory cytokines, alleviating oxidative stress and inflammation levels in hippocampal tissues of SD model mice. cTBS decreased LC3II/LC3I ratio, Beclin1 protein levels, and LC3B puncta intensity, and elevated p62 protein levels to suppress excessive autophagy in hippocampal tissues of SD-stimulated mice. Then, we proved that inhibiting oxidative stress alleviated inflammation, autophagy, and death of hippocampal neuron cells through an in vitro cellular model for oxidative stress, and cTBS treatment promoted the production of glutathione (GSH), the nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and the mRNA expression of GSH synthesis-related genes to enhance antioxidant capacity in hippocampal tissues of SD mice. An Nrf2 inhibitor ML385 or a GSH synthesis inhibitor BSO reversed the alleviating effects of cTBS treatment on oxidative stress-associated damage of hippocampal tissues and cognitive impairment in SD model mice. Altogether, our study demonstrated that cTBS mitigates oxidative stress-associated inflammation and autophagy through activating the Nrf2-mediated GSH synthesis pathway, improving cognitive impairment in SD mice.
Collapse
Affiliation(s)
- Yi Zhang
- Clinical Psychology Department, the People's Hospital of Xinjiang Uygur Autonomous Region, 91 Tianchi Road, Urumqi, 830001, China
| | - Cheng Zhang
- Clinical Psychology Department, the People's Hospital of Xinjiang Uygur Autonomous Region, 91 Tianchi Road, Urumqi, 830001, China
| | - Qing Dai
- Anesthesiology Department, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Rui Ma
- Clinical Psychology Department, the People's Hospital of Xinjiang Uygur Autonomous Region, 91 Tianchi Road, Urumqi, 830001, China.
| |
Collapse
|
21
|
Aghajani M, Aghajani M, Moghaddam EK, Faghihi M, Imani A. Acute sleep deprivation (ASD) and cardioprotection: Impact of ASD on oxytocin-mediated sympathetic nervous activation preceding myocardial infarction. Neuropeptides 2024; 107:102453. [PMID: 38959559 DOI: 10.1016/j.npep.2024.102453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 05/10/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION This study explored how acute sleep deprivation (ASD) before myocardial ischemia influences oxytocin release from paraventricular (PVN) neurons and its correlation with sympathetic nervous system (SNS) activity post-acute sleep loss, impacting subsequent left ventricular (LV) remodeling following myocardial infarction (MI). METHODS The study was conducted in two phases: induction of ASD, inducing MI, blood sampling, euthanizing animals and collecting their heart and brain for histological and gene expression evaluations. The animals in first and second phase were euthanized 24 h and 14 days after MI, respectively. RESULTS Pre-MI ASD, accompanied by increased serum epinephrine levels within 24 h of MI, upregulated oxytocin and cFos expression in the PVN. Also, pre-MI ASD resulted in decreased serum PAB levels 14 days post-MI (P < 0.001). While notable echocardiographic changes were seen in MI versus sham groups, ASD demonstrated protective effects. This was evidenced by reduced infarct size, elevated TIMP1, MMP2, and MMP9 in the LV of SD + MI animals versus MI alone (P < 0.05). Additionally, histological analysis showed reduced LV fibrosis in pre-MI ASD subjects (P < 0.05). CONCLUSION Our study supports the notion that activation of oxytocin neurons within the PVN subsequent to ASD interacts with autonomic centers in the central nervous system. This enhanced sympathetic outflow to the heart prior to MI triggers a preconditioning response, thereby mediating cardioprotection through decreased oxidative stress biomarkers and regulated extracellular matrix (ECM) turnover.
Collapse
Affiliation(s)
- Marjan Aghajani
- Physiology Department, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mozhgan Aghajani
- Rasoole-Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mahdieh Faghihi
- Physiology Department, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Imani
- Physiology Department, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Sleep Breathing Disorders Research Center (SBDRC), Tehran University of Medical Sciences, Tehran, Iran; Occupational Sleep Research Center, Baharloo Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Zhu Z, Yin Q, Duan X. Xiaoyin-anshen formula alleviates psoriasis complicated by sleep disturbances by regulating melatonin, antioxidant enzymes, and pro-inflammatory cytokines in mice. Front Pharmacol 2024; 15:1427985. [PMID: 39411067 PMCID: PMC11473356 DOI: 10.3389/fphar.2024.1427985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Background Psoriasis is a common autoimmune and chronic inflammatory dermatological disease that is mainly associated with aberrant immune response and oxidative stress (OS). OS, a crucial pathogenic factor in psoriasis, contributes to psoriasis-like inflammation mediated by the IL-23/IL-17 inflammatory axis. Sleep disturbances (SDs), highly prevalent in patients with psoriasis, exacerbate the condition by disrupting circadian rhythms and reducing melatonin levels, thus promoting OS and inflammation. Xiaoyin-Anshen formula (XYAS), a traditional Chinese medicine (TCM) formula, is composed of the Liangxue-Jiedu (LXJD) and Qingxin-Anshen (QXAS) TCM compounds and has been demonstrated to be effective in treating psoriasis complicated by SDs. However, its exact pharmacological mechanism remains uncertain. Thus, this study used animal experiments to verify whether XYAS can exert therapeutic effects on the disease by regulating melatonin (MLT) levels, protecting against OS, and inhibiting psoriasis-like skin inflammation. Methods A mouse model for psoriasis combined with SDs was established by smearing 62.5 mg of 5% imiquimod (IMQ) cream for seven consecutive days, along with a daily injection of p-chlorophenyl alanine (PCPA) solution at a dosage of 300 mg/kg at days 6-7. The IMQ cream was continued to be used for maintaining the model at days 8-14. Mice were randomly divided into groups: control, model, MLT, XYAS, LXJD, QXAS. Each group was treated according to its designation at days 8-14, receiving either an oral gavage of XYAS/LXJD/QXAS solution at a dosage of 2 mL/100 g per day, or a daily injection of MLT solution at a concentration of 0.25 mg/mL, with a dosage of 5 mg/kg. Immunohistological analysis, pentobarbital-induced sleep test, Western blotting, and enzyme-linked immunosorbent assay (ELISA) were performed to assess and compare pathological features, sleep conditions, localization and/or levels of manganese-dependent superoxide dismutase (mnSOD), mitochondrial cytochrome c (Cyt-C), MLT, retinoid-related orphan nuclear receptor-α (RORα), and pro-inflammatory cytokines interleukin (IL)-6, IL-17A, and tumor necrosis factor-alpha (TNF-α) among groups. Results MLT, XYAS, LXJD, and QXAS exhibited varying therapeutic effects on RORα regulation, OS inhibition, mitochondrial protection, and anti-inflammation. Compared to the model, the lesion severity/thickness and serum IL-6, IL-17A, and TNF-α levels were gradually reduced in the MLT, QXAS, LXJD, and XYAS. However, no statistical difference in TNF-α levels was identified between the MLT and the model groups. Additionally, skin MLT levels gradually increased in the MLT, QXAS, and XYAS groups, while RORα levels gradually increased in the MLT, QXAS, LXJD, and XYAS groups. All treatments increased mnSOD levels and reduced Cyt-C levels in skin lesions, with XYAS showing the most significant changes. Conclusion XYAS may treat psoriasis complicated by SDs through two main mechanisms: (1) Improving melatonin-RORα axis in the skin can lead to an increase in mnSOD and a reduction in Cyt-C levels, which provide protection against oxidative stress, mitochondrial damage, and psoriatic inflammation. (2) Reducing IL-6, IL-17A, and TNF-α production to suppress IL-23/Th17 pro-inflammatory signaling axis and epidermal hyperplasia in psoriasis.
Collapse
Affiliation(s)
- Zebing Zhu
- Department of Dermatology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Qiang Yin
- Department of Dermatology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xingwu Duan
- Department of Dermatology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
23
|
Goodman LD, Moulton MJ, Lin G, Bellen HJ. Does glial lipid dysregulation alter sleep in Alzheimer's and Parkinson's disease? Trends Mol Med 2024; 30:913-923. [PMID: 38755043 PMCID: PMC11466711 DOI: 10.1016/j.molmed.2024.04.010] [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: 02/17/2024] [Revised: 03/28/2024] [Accepted: 04/10/2024] [Indexed: 05/18/2024]
Abstract
In this opinion article, we discuss potential connections between sleep disturbances observed in Alzheimer's disease (AD) and Parkinson's disease (PD) and the dysregulation of lipids in the brain. Research using Drosophila has highlighted the role of glial-mediated lipid metabolism in sleep and diurnal rhythms. Relevant to AD, the formation of lipid droplets in glia, which occurs in response to elevated neuronal reactive oxygen species (ROS), is required for sleep. In disease models, this process is disrupted, arguing a connection to sleep dysregulation. Relevant to PD, the degradation of neuronally synthesized glucosylceramides by glia requires glucocerebrosidase (GBA, a PD-associated risk factor) and this regulates sleep. Loss of GBA in glia causes an accumulation of glucosylceramides and neurodegeneration. Overall, research primarily using Drosophila has highlighted how dysregulation of glial lipid metabolism may underlie sleep disturbances in neurodegenerative diseases.
Collapse
Affiliation(s)
- Lindsey D Goodman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Matthew J Moulton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Guang Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
24
|
Pan H, Lin S. Association between dietary total antioxidant capacity and sleep problems and depressive symptoms among U.S. adults. Front Nutr 2024; 11:1450815. [PMID: 39391679 PMCID: PMC11466051 DOI: 10.3389/fnut.2024.1450815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/03/2024] [Indexed: 10/12/2024] Open
Abstract
Background In this study, we aim to examine the impact of dietary total antioxidant capacity (TAC) on sleep problems and depressive symptoms (DS); besides, we seek to elucidate the potential mediating effect of dietary TAC on the relationships between sleep problems and DS. Methods Weighted Kruskal-Wallis tests for continuous variables and Chi-square tests for categorical variables were employed to discriminate between DS and non-DS participants. Multivariable logistic regression and restricted cubic spline analysis were applied to evaluate the associations of TAC with DS and sleep problems. Results Among the 21,805 participants, 1,947 participants suffered from DS. Weighted multivariable logistical regression indicated that shorter sleep hours were linked to an increased likelihood of risk of DS even after complete adjustments. Restricted cubic spline cure displayed that TAC was almost non-linearly correlated with DS and sleep problems. Mediation analysis indicated that sleep duration slightly mediated the association between TAC and DS (proportion of mediation: 3.12%, p < 0.001). Conclusion This study illustrated the inverse association between TAC value and sleep problems and DS. Furthermore, TAC slightly mediated the effect of sleep duration on the DS, and there was a nearly non-linear relationship between TAC and DS, and TAC and sleep problems.
Collapse
Affiliation(s)
- Hong Pan
- Department of Neurology, Deqing People’s Hospital, Huzhou, China
| | - Shasha Lin
- Department of Ultrasound, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
25
|
Ye M, Jeong W, Yu HJ, Kim KR, Rhie SJ, Kim Y, Kim J, Shim I. Effect of Earthing Mats on Sleep Quality in Rats. Int J Mol Sci 2024; 25:9791. [PMID: 39337279 PMCID: PMC11432166 DOI: 10.3390/ijms25189791] [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: 08/15/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Grounding, a therapeutic technique involving direct contact with the earth, has been proposed by various studies to potentially have beneficial effects on pressure, sleep quality, stress, inflammation, and mood. However, the scientific evidence supporting its sedative effects remains incomplete. This study examined the sedative effectiveness of an earthing mat on sleep quality and investigated the underlying neural mechanisms using electroencephalography (EEG) analysis in rodents, focusing on orexin and superoxide dismutase (SOD) levels in the brain. Rats were randomly assigned to four groups: the naïve normal group (Nor), the group exposed to an earthing mat for 7 days (A-7D), the group exposed to an earthing mat for 21 days (A-21D), and the group exposed to an electronic blanket for 21 days (EM). EEG results revealed that the A-21D group exhibited significantly reduced wake time and increased rapid eye movement (REM), non-rapid eye movement (NREM), and total sleep time compared to the Nor group (p < 0.05). Moreover, the A-21D group demonstrated a significant increase in NREM sleep (p < 0.001), REM sleep (p < 0.01), and total sleep time (p < 0.001), along with a decrease in wake time compared to the EM group (p < 0.001). The orexin level in the A-21D group was significantly lower compared to the Nor group (p < 0.01), while SOD1 expression was markedly elevated in the A-21D group compared to the Nor group (p < 0.001). These results suggest that the earthing mat may represent a promising new method for promoting sleep quality and could serve as an effective therapeutic technique.
Collapse
Affiliation(s)
- Minsook Ye
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02453, Republic of Korea; (M.Y.); (W.J.); (H.-j.Y.); (K.-r.K.)
| | - Woojin Jeong
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02453, Republic of Korea; (M.Y.); (W.J.); (H.-j.Y.); (K.-r.K.)
| | - Hyo-jeong Yu
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02453, Republic of Korea; (M.Y.); (W.J.); (H.-j.Y.); (K.-r.K.)
| | - Kyu-ri Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02453, Republic of Korea; (M.Y.); (W.J.); (H.-j.Y.); (K.-r.K.)
| | - Sung Ja Rhie
- Department of Beauty Design, Halla University, Wonju 26404, Republic of Korea;
| | - Yongsuk Kim
- DF World Corporation, Royal Building, 19 Saemunan-ro 5-gil, Jongno-gu, Seoul 03173, Republic of Korea;
| | - Jiyoun Kim
- World Home Doctor Corporation, 73 Anyangcheonseo-ro, Manan-gu, Anyang-si 14087, Republic of Korea;
| | - Insop Shim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02453, Republic of Korea; (M.Y.); (W.J.); (H.-j.Y.); (K.-r.K.)
| |
Collapse
|
26
|
Malik DM, Rhoades SD, Kain P, Sengupta A, Sehgal A, Weljie AM. Altered Metabolism during the Dark Period in Drosophila Short Sleep Mutants. J Proteome Res 2024; 23:3823-3836. [PMID: 38836855 DOI: 10.1021/acs.jproteome.4c00106] [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] [Indexed: 06/06/2024]
Abstract
Sleep is regulated via circadian mechanisms, but effects of sleep disruption on physiological rhythms, in particular metabolic cycling, remain unclear. To examine this question, we probed diurnal metabolic alterations of two Drosophila short sleep mutants, fumin and sleepless. Samples were collected with high temporal sampling (every 2 h) over 24 h under a 12:12 light:dark cycle, and profiling was done using an ion-switching LCMS/MS method. Fewer metabolites with 24 h oscillations were noted with short sleep (50 and 46 in fumin and sleepless, BH. Q < 0.2 by RAIN analysis) compared to a wild-type control (iso31, 63 with BH. Q < 0.2), and peak phases of the sleep mutants were consolidated into two major phase peaks at mid-day and middle of night. Overall, altered nicotinate/nicotinamide, alanine/aspartate/glutamate, acetylcholine, glyoxylate/dicarboxylate, and TCA cycle metabolism were observed in the short sleep mutants, indicative of increased energetic demand and oxidative stress compared to wild type. Both changes in cycling and discriminant models suggest unique alterations in the dark period indicative of constrained metabolic networks. Thus, we conclude that sleep loss alters metabolic function uniquely throughout the day, and further examination of specific mechanisms is warranted.
Collapse
Affiliation(s)
- Dania M Malik
- Pharmacology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Seth D Rhoades
- Pharmacology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Fulgens Consulting, LLC, Cambridge, Massachusetts 02142, United States
| | - Pinky Kain
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Amita Sehgal
- Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Aalim M Weljie
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
27
|
Liang C, Luong LT. Ghosts of parasites past influence current non-consumptive effects in Drosophila nigrospiracula. Int J Parasitol 2024; 54:551-558. [PMID: 38677400 DOI: 10.1016/j.ijpara.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/26/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
Parasites can indirectly impact hosts through non-consumptive effects (NCEs) via changes in behaviour, morphology, and/or physiology. These responses can be understood in terms of the ecology of fear (ectoparasites) or the ecology of disgust (endoparasites) framework. We tested the hypothesis that NCEs of parasite exposure (e.g., parasite avoidance and defense) trade off with other important behaviours such as feeding and resting. We predicted that when exposed to parasites (without infection), hosts will increase their defensive behaviors at the expense of feeding. We also posited that history of exposure (without infection), or previous infection would impact the expression of these NCEs. The study system involves a cactophilic fruit fly (Drosophila nigrospiracula) and a naturally occurring parasitic mite (Macrocheles subbadius). First, we assessed how prior mite exposure affected fly behaviour in response to current parasite exposure. Mite presence resulted in increased grooming and movement, but exposure history did not affect these behaviours. However, the interaction between previous and current exposure influenced host feeding and resting behaviours. We found that previously exposed flies increased feeding and decreased resting upon a secondary mite exposure. In a second experiment, we tested the role of infection history on current parasite exposure. Compared with naïve flies, previously infected flies were expected to increase defensive behaviours upon secondary exposure. Flies increased defensive and ambulatory behaviour in the presence of mites, and consequently less time was spent resting but feeding was unaffected. None of the behaviours measured were affected by previous infection status. In general, current parasite exposure resulted in NCEs. Moreover, our results showed that previous exposure (without infection) to parasites may have an even stronger effect upon secondary exposure than infection history. Our study highlights the importance of the ecology of fear and the role that exposure and infection history plays in generating NCEs of parasitism.
Collapse
Affiliation(s)
- C Liang
- University of Alberta, Department of Biological Sciences, 11455 Saskatchewan Drive, Edmonton, AB T6G 2E9, Canada.
| | - L T Luong
- University of Alberta, Department of Biological Sciences, 11455 Saskatchewan Drive, Edmonton, AB T6G 2E9, Canada
| |
Collapse
|
28
|
O'Hearn LA. Signals of energy availability in sleep: consequences of a fat-based metabolism. Front Nutr 2024; 11:1397185. [PMID: 39267859 PMCID: PMC11390529 DOI: 10.3389/fnut.2024.1397185] [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: 03/07/2024] [Accepted: 08/05/2024] [Indexed: 09/15/2024] Open
Abstract
Humans can flexibly switch between two primary metabolic modes, usually distinguished by whether substrate supply from glucose can meet energy demands or not. However, it is often overlooked that when glucose use is limited, the remainder of energy needs may still be met more or less effectively with fat and ketone bodies. Hence a fat-based metabolism marked by ketosis is often conflated with starvation and contexts of inadequate energy (including at the cellular level), even when energy itself is in ample supply. Sleep and satiation are regulated by common pathways reflecting energy metabolism. A conceptual analysis that distinguishes signals of inadequate energy in a glucose-dominant metabolism from signals of a fat-based metabolism that may well be energy sufficient allows a reexamination of experimental results in the study of sleep that may shed light on species differences and explain why ketogenic diets have beneficial effects simultaneously in the brain and the periphery. It may also help to distinguish clinically when a failure of a ketogenic diet to resolve symptoms is due to inadequate energy rather than the metabolic state itself.
Collapse
|
29
|
Turner BRH, Jenkinson PI, Huttman M, Mullish BH. Inflammation, oxidative stress and gut microbiome perturbation: A narrative review of mechanisms and treatment of the alcohol hangover. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1451-1465. [PMID: 38965644 DOI: 10.1111/acer.15396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/17/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024]
Abstract
Alcohol is the most widely abused substance in the world, the leading source of mortality in 15-49-year-olds, and a major risk factor for heart disease, liver disease, diabetes, and cancer. Despite this, alcohol is regularly misused in wider society. Consumers of excess alcohol often note a constellation of negative symptoms, known as the alcohol hangover. However, the alcohol hangover is not considered to have long-term clinical significance by clinicians or consumers. We undertook a critical review of the literature to demonstrate the pathophysiological mechanisms of the alcohol hangover. Hereafter, the alcohol hangover is re-defined as a manifestation of sickness behavior secondary to alcohol-induced inflammation, using the Bradford-Hill criteria to demonstrate causation above correlation. Alcohol causes inflammation through oxidative stress and endotoxemia. Alcohol metabolism is oxidative and increased intake causes relative tissue hypoxia and increased free radical generation. Tissue damage ensues through lipid peroxidation and the formation of DNA/protein adducts. Byproducts of alcohol metabolism such as acetaldehyde and congeners, sleep deprivation, and the activation of nonspecific inducible CYP2E1 in alcohol-exposed tissues exacerbate free radical generation. Tissue damage and cell death lead to inflammation, but in the intestine loss of epithelial cells leads to intestinal permeability, allowing the translocation of pathogenic bacteria to the systemic circulation (endotoxemia). This leads to a well-characterized cascade of systemic inflammation, additionally activating toll-like receptor 4 to induce sickness behavior. Considering the evidence, it is suggested that hangover frequency and severity may be predictors of the development of later alcohol-related diseases, meriting formal confirmation in prospective studies. In light of the mechanisms of alcohol-mediated inflammation, research into gut permeability and the gut microbiome may be an exciting future therapeutic avenue to prevent alcohol hangover and other alcohol-related diseases.
Collapse
Affiliation(s)
| | - Poppy I Jenkinson
- Department of Anaesthetics, Royal Surrey County Hospital, Surrey, UK
| | - Marc Huttman
- Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
| | - Benjamin H Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
30
|
Davinelli S, Medoro A, Savino R, Scapagnini G. Sleep and Oxidative Stress: Current Perspectives on the Role of NRF2. Cell Mol Neurobiol 2024; 44:52. [PMID: 38916679 PMCID: PMC11199221 DOI: 10.1007/s10571-024-01487-0] [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: 02/26/2024] [Accepted: 06/15/2024] [Indexed: 06/26/2024]
Abstract
Sleep is a fundamental conserved physiological state across evolution, suggesting vital biological functions that are yet to be fully clarified. However, our understanding of the neural and molecular basis of sleep regulation has increased rapidly in recent years. Among various processes implicated in controlling sleep homeostasis, a bidirectional relationship between sleep and oxidative stress has recently emerged. One proposed function of sleep may be the mitigation of oxidative stress in both brain and peripheral tissues, contributing to the clearance of reactive species that accumulate during wakefulness. Conversely, reactive species, such as reactive oxygen species (ROS) and reactive nitrogen species (RNS), at physiological levels, may act as signaling agents to regulate redox-sensitive transcriptional factors, enzymes, and other effectors involved in the regulation of sleep. As a primary sensor of intracellular oxidation, the transcription factor NRF2 is emerging as an indispensable component to maintain cellular redox homeostasis during sleep. Indeed, a number of studies have revealed an association between NRF2 dysfunction and the most common sleep conditions, including sleep loss, obstructive sleep apnea, and circadian sleep disturbances. This review examines the evidence of the intricate link between oxidative stress and NRF2 function in the context of sleep, and highlights the potential of NRF2 modulators to alleviate sleep disturbances.
Collapse
Affiliation(s)
- Sergio Davinelli
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Via F. De Sanctis, s.n.c., 86100, Campobasso, Italy.
| | - Alessandro Medoro
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Via F. De Sanctis, s.n.c., 86100, Campobasso, Italy
| | - Rosa Savino
- Department of Woman and Child, Neuropsychiatry for Child and Adolescent Unit, General Hospital "Riuniti" of Foggia, Viale Pinto Luigi, 1, 71122, Foggia, Italy
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Via F. De Sanctis, s.n.c., 86100, Campobasso, Italy
| |
Collapse
|
31
|
Tang L, Liu M, Mu J, Tian J. Association between circulating antioxidants and sleep disorders: comprehensive results from NHANES 2017-2018. Food Funct 2024; 15:6657-6672. [PMID: 38814101 DOI: 10.1039/d4fo00413b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Background: Oxidative stress plays an important role in the occurrence and pathological process of numerous human diseases. A bidirectional relationship was found between sleep disorders and oxidative stress. However, the association between circulating antioxidant levels and the risk of sleep disorders at the population-scale has yet to be determined. Methods: We used the dataset from the National Health and Nutrition Examination Survey (NHANES) 2017-2018 data release cycle and included 3062 adult participants aged 25-75 years. The circulating antioxidants levels in serum were measured, and the sleep status was assessed by self-reported sleep disorder questionnaire tests. We investigated the association and exposure-response relationship between the 12 main circulating antioxidants and sleep disorders using a generalized additive model (GAM), multiple linear, binary logistic, and restricted cubic spline (RCS) regression models. Multiple sensitivity analyses were conducted to validate the results of our study. Results: Significantly lower serum concentrations of ten antioxidants were observed in the group which had trouble sleeping symptoms compared to the control group. After adjusting for all the covariates, the binary logistic regression models indicated that six of the circulating antioxidants including alpha-carotene, alpha-cryptoxanthin, trans-beta-carotene, beta-cryptoxanthin, lutein and zeaxanthin, and vitamin C, showed a significant association with the risk of overall trouble sleeping symptoms, with odds ratios corresponding to 0.88 (95% CI: 0.80-0.96), 0.74 (95% CI: 0.62-0.87), 0.87 (95% CI: 0.79-0.97), 0.85 (95% CI: 0.75-0.95), 0.72 (95% CI: 0.61-0.84), and 0.83 (95% CI: 0.74-0.93), respectively. The GAM and multiple linear regression revealed similar associations whereas the RCS regression models further confirmed their significant negative exposure-response relationship. Conclusions: The circulating carotenoids and vitamin C levels were negatively correlated with the risk of sleep disorders. Higher circulating antioxidant levels were significantly associated with a lower risk of sleep disorders. The potential health risk of low circulating antioxidants levels was higher in the female population than in the male population.
Collapse
Affiliation(s)
- Liwei Tang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055 China.
| | - Min Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055 China.
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055 China
| | - Jingjing Mu
- Affiliated Psychological Hospital of Anhui Medical University, Anhui Mental Health Center, Hefei Fourth People's Hospital, Hefei 230022, China
| | - Jing Tian
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055 China.
| |
Collapse
|
32
|
Yano J, Nave C, Larratt K, Honey P, Roberts M, Jingco C, Fung ML, Trotter D, He X, Elezi G, Whitelegge JP, Wasserman S, Donlea JM. Elevated sleep quota in a stress-resilient Drosophila species. Curr Biol 2024; 34:2487-2501.e3. [PMID: 38772361 PMCID: PMC11163955 DOI: 10.1016/j.cub.2024.04.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/09/2024] [Accepted: 04/25/2024] [Indexed: 05/23/2024]
Abstract
Sleep is broadly conserved across the animal kingdom but can vary widely between species. It is currently unclear which selective pressures and regulatory mechanisms influence differences in sleep between species. The fruit fly Drosophila melanogaster has become a successful model system for examining sleep regulation and function, but little is known about the sleep patterns in many related fly species. Here, we find that fly species with adaptations to extreme desert environments, including D. mojavensis, exhibit strong increases in baseline sleep compared with D. melanogaster. Long-sleeping D. mojavensis show intact homeostasis, indicating that desert flies carry an elevated drive for sleep. In addition, D. mojavensis exhibit altered abundance or distribution of several sleep/wake-related neuromodulators and neuropeptides that are consistent with their reduced locomotor activity and increased sleep. Finally, we find that in a nutrient-deprived environment, the sleep patterns of individual D. mojavensis are strongly correlated with their survival time and that disrupting sleep via constant light stimulation renders D. mojavensis more sensitive to starvation. Our results demonstrate that D. mojavensis is a novel model for studying organisms with high sleep drive and for exploring sleep strategies that provide resilience in extreme environments.
Collapse
Affiliation(s)
- Jessica Yano
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Molecular, Cellular & Integrative Physiology Interdepartmental PhD Program, UCLA, Los Angeles, CA 90095, USA
| | - Ceazar Nave
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Katherine Larratt
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Phia Honey
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Department of Neuroscience, Wellesley College, Wellesley, MA 02481, USA
| | - Makayla Roberts
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Cassandra Jingco
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Melanie L Fung
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Damion Trotter
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Molecular, Cellular & Integrative Physiology Interdepartmental PhD Program, UCLA, Los Angeles, CA 90095, USA
| | - Xin He
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Gazmend Elezi
- Pasarow Mass Spectrometry Laboratory, Jane & Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Cotsen Institute of Archaeology, UCLA, Los Angeles, CA 90095, USA
| | - Julian P Whitelegge
- Pasarow Mass Spectrometry Laboratory, Jane & Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Sara Wasserman
- Department of Neuroscience, Wellesley College, Wellesley, MA 02481, USA
| | - Jeffrey M Donlea
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
33
|
Terzi A, Ngo KJ, Mourrain P. Phylogenetic conservation of the interdependent homeostatic relationship of sleep regulation and redox metabolism. J Comp Physiol B 2024; 194:241-252. [PMID: 38324048 PMCID: PMC11233307 DOI: 10.1007/s00360-023-01530-4] [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: 07/28/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 02/08/2024]
Abstract
Sleep is an essential and evolutionarily conserved process that affects many biological functions that are also strongly regulated by cellular metabolism. The interdependence between sleep homeostasis and redox metabolism, in particular, is such that sleep deprivation causes redox metabolic imbalances in the form of over-production of ROS. Likewise (and vice versa), accumulation of ROS leads to greater sleep pressure. Thus, it is theorized that one of the functions of sleep is to act as the brain's "antioxidant" at night by clearing oxidation built up from daily stress of the active day phase. In this review, we will highlight evidence linking sleep homeostasis and regulation to redox metabolism by discussing (1) the bipartite role that sleep-wake neuropeptides and hormones have in redox metabolism through comparing cross-species cellular and molecular mechanisms, (2) the evolutionarily metabolic changes that accompanied the development of sleep loss in cavefish, and finally, (3) some of the challenges of uncovering the cellular mechanism underpinning how ROS accumulation builds sleep pressure and cellularly, how this pressure is cleared.
Collapse
Affiliation(s)
- Aslihan Terzi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Keri J Ngo
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
| | - Philippe Mourrain
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- INSERM 1024, Ecole Normale Supérieure, Paris, France.
| |
Collapse
|
34
|
Zhang W, Liu D, Yuan M, Zhu LQ. The mechanisms of mitochondrial abnormalities that contribute to sleep disorders and related neurodegenerative diseases. Ageing Res Rev 2024; 97:102307. [PMID: 38614368 DOI: 10.1016/j.arr.2024.102307] [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: 03/09/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Sleep is a highly intricate biological phenomenon, and its disorders play a pivotal role in numerous diseases. However, the specific regulatory mechanisms remain elusive. In recent years, the role of mitochondria in sleep disorders has gained considerable attention. Sleep deprivation not only impairs mitochondrial morphology but also decreases the number of mitochondria and triggers mitochondrial dysfunction. Furthermore, mitochondrial dysfunction has been implicated in the onset and progression of various sleep disorder-related neurological diseases, especially neurodegenerative conditions. Therefore, a greater understanding of the impact of sleep disorders on mitochondrial dysfunction may reveal new therapeutic targets for neurodegenerative diseases. In this review, we comprehensively summarize the recent key findings on the mechanisms underlying mitochondrial dysfunction caused by sleep disorders and their role in initiating or exacerbating common neurodegenerative diseases. In addition, we provide fresh insights into the diagnosis and treatment of sleep disorder-related diseases.
Collapse
Affiliation(s)
- Wentao Zhang
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Dan Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Mei Yuan
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Affiliated Nanhua Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
35
|
Su AX, Ma ZJ, Li ZY, Li XY, Xia L, Ge YJ, Chen GH. Serum levels of neurotensin, pannexin-1, and sestrin-2 and the correlations with sleep quality or/and cognitive function in the patients with chronic insomnia disorder. Front Psychiatry 2024; 15:1360305. [PMID: 38803679 PMCID: PMC11128551 DOI: 10.3389/fpsyt.2024.1360305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Objectives To examine serum concentrations of neurotensin, pannexin-1 and sestrin-2, and their correlations with subjective and objective sleep quality and cognitive function in the patients with chronic insomnia disorder (CID). Methods Sixty-five CID patients were enrolled continuously and fifty-six good sleepers in the same period were served as healthy controls (HCs). Serum levels of neurotensin, pannexin-1 and sestrin-2 were measured by enzyme-linked immunosorbent assays. Sleep quality was assessed with the Pittsburgh Sleep Quality Index (PSQI) and polysomnography, and mood was evaluated by 17-item Hamilton Depression Rating Scale. General cognitive function was assessed with the Chinese-Beijing Version of Montreal Cognitive Assessment and spatial memory was evaluated by Blue Velvet Arena Test (BVAT). Results Relative to the HCs, the CID sufferers had higher levels of neurotensin (t=5.210, p<0.001) and pannexin-1 (Z=-4.169, p<0.001), and lower level of sestrin-2 (Z=-2.438, p=0.015). In terms of objective sleep measures, pannexin-1 was positively associated with total sleep time (r=0.562, p=0.002) and sleep efficiency (r=0.588, p=0.001), and negatively with wake time after sleep onset (r=-0.590, p=0.001) and wake time (r=-0.590, p=0.001); sestrin-2 was positively associated with percentage of rapid eye movement sleep (r=0.442, p=0.016) and negatively with non-rapid eye movement sleep stage 2 in the percentage (r=-0.394, p=0.034). Adjusted for sex, age and HAMD, pannexin-1 was still associated with the above objective sleep measures, but sestrin-2 was only negatively with wake time (r=-0.446, p=0.022). However, these biomarkers showed no significant correlations with subjective sleep quality (PSQI score). Serum concentrations of neurotensin and pannexin-1 were positively associated with the mean erroneous distance in the BVAT. Adjusted for sex, age and depression, neurotensin was negatively associated with MoCA score (r=-0.257, p=0.044), pannexin-1 was positively associated with the mean erroneous distance in the BVAT (r=0.270, p=0.033). Conclusions The CID patients had increased neurotensin and pannexin-1 and decreased sestrin-2 in the serum levels, indicating neuron dysfunction, which could be related to poor sleep quality and cognitive dysfunction measured objectively.
Collapse
Affiliation(s)
- Ai-Xi Su
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
- Department of General Medicine, the First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Zi-Jie Ma
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Zong-Yin Li
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Xue-Yan Li
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Lan Xia
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Yi-Jun Ge
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| |
Collapse
|
36
|
Yook S, Park HR, Joo EY, Kim H. Predicting the impact of CPAP on brain health: A study using the sleep EEG-derived brain age index. Ann Clin Transl Neurol 2024; 11:1172-1183. [PMID: 38396240 DOI: 10.1002/acn3.52032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/17/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
OBJECTIVE This longitudinal study investigated potential positive impact of CPAP treatment on brain health in individuals with obstructive sleep Apnea (OSA). To allow this, we aimed to employ sleep electroencephalogram (EEG)-derived brain age index (BAI) to quantify CPAP's impact on brain health and identify individually varying CPAP effects on brain aging using machine learning approaches. METHODS We retrospectively analyzed CPAP-treated (n = 98) and untreated OSA patients (n = 88) with a minimum 12-month follow-up of polysomnography. BAI was calculated by subtracting chronological age from the predicted brain age. To investigate BAI changes before and after CPAP treatment, we compared annual ΔBAI between CPAP-treated and untreated OSA patients. To identify individually varying CPAP effectiveness and factors influencing CPAP effectiveness, machine learning approaches were employed to predict which patient displayed positive outcomes (negative annual ΔBAI) based on their baseline clinical features. RESULTS CPAP-treated group showed lower annual ΔBAI than untreated (-0.6 ± 2.7 vs. 0.3 ± 2.6 years, p < 0.05). This BAI reduction with CPAP was reproduced independently in the Apnea, Bariatric surgery, and CPAP study cohort. Patients with more severe OSA at baseline displayed more positive annual ΔBAI (=accelerated brain aging) when untreated and displayed more negative annual ΔBAI (=decelerated brain aging) when CPAP-treated. Machine learning models achieved high accuracy (up to 86%) in predicting CPAP outcomes. INTERPRETATION CPAP treatment can alleviate brain aging in OSA, especially in severe cases. Sleep EEG-derived BAI has potential to assess CPAP's impact on brain health. The study provides insights into CPAP's effects and underscores BAI-based predictive modeling's utility in OSA management.
Collapse
Affiliation(s)
- Soonhyun Yook
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, 90033, USA
| | - Hea Ree Park
- Department of Neurology, Inje University College of Medicine, Ilsan Paik Hospital, Goyang, 10380, Korea
| | - Eun Yeon Joo
- Department of Neurology, Neuroscience Center, Samsung Medical Center, Samsung Biomedical Research Institute, School of Medicine, Sungkyunkwan University, Seoul, 06351, Korea
| | - Hosung Kim
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, 90033, USA
| |
Collapse
|
37
|
You Y, Li J, Zhang Y, Li X, Li X, Ma X. Exploring the potential relationship between short sleep risks and cognitive function from the perspective of inflammatory biomarkers and cellular pathways: Insights from population-based and mice studies. CNS Neurosci Ther 2024; 30:e14783. [PMID: 38797980 PMCID: PMC11128714 DOI: 10.1111/cns.14783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
AIMS The molecular mechanism of short-sleep conditions on cognition remains largely unknown. This research aimed to investigate associations between short sleep, inflammatory biomarkers and cognitive function in the US population (NHANES data 2011-2014) and explore cellular mechanisms in mice. METHODS Systemic immune-inflammation index (SII) was calculated using blood-cell based biomarkers. Further, we employed integrated bioinformatics and single-cell transcriptomics (GSE137665) to examine how short sleep exposure influenced the molecular pathways associated with inflammation in the brain. To explore the signaling pathways and biological processes of sleep deprivation, we carried out enrichment analyses utilizing the GO and KEGG databases. RESULTS Population results showed that, compared with normal sleep group, severe short sleep was associated with lower cognitive ability in all the four tests. Moreover, a higher SII level was correlated with lower scores of cognitive tests. In mice study, elevated activation of the inflammatory pathway was observed in cell subgroups of neurons within the sleep deprivation and recovery sleep cohorts. Additionally, heightened expression of oxidative stress and integrated stress response pathways was noted in GABAergic neurons during sleep deprivation. CONCLUSION This study contributed to the understanding of the influence of short sleep on cognitive function and its cellular mechanisms.
Collapse
Affiliation(s)
- Yanwei You
- Division of Sports Science & Physical EducationTsinghua UniversityBeijingChina
- IDG/McGovern Institute for Brain ResearchTsinghua UniversityBeijingChina
| | - Jinwei Li
- Department of Neurosurgery, West China HospitalSichuan UniversityChengduChina
| | - Yang Zhang
- Department of Vascular SurgeryFuwai Yunnan Cardiovascular Hospital, Affiliated Cardiovascular Hospital of Kunming Medical UniversityKunmingChina
| | - Xingtian Li
- Division of Sports Science & Physical EducationTsinghua UniversityBeijingChina
- IDG/McGovern Institute for Brain ResearchTsinghua UniversityBeijingChina
| | - Xinming Li
- Division of Sports Science & Physical EducationTsinghua UniversityBeijingChina
- IDG/McGovern Institute for Brain ResearchTsinghua UniversityBeijingChina
| | - Xindong Ma
- Division of Sports Science & Physical EducationTsinghua UniversityBeijingChina
- IDG/McGovern Institute for Brain ResearchTsinghua UniversityBeijingChina
| |
Collapse
|
38
|
Zeng Y, Guo Z, Wu M, Chen F, Chen L. Circadian rhythm regulates the function of immune cells and participates in the development of tumors. Cell Death Discov 2024; 10:199. [PMID: 38678017 PMCID: PMC11055927 DOI: 10.1038/s41420-024-01960-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024] Open
Abstract
Circadian rhythms are present in almost all cells and play a crucial role in regulating various biological processes. Maintaining a stable circadian rhythm is essential for overall health. Disruption of this rhythm can alter the expression of clock genes and cancer-related genes, and affect many metabolic pathways and factors, thereby affecting the function of the immune system and contributing to the occurrence and progression of tumors. This paper aims to elucidate the regulatory effects of BMAL1, clock and other clock genes on immune cells, and reveal the molecular mechanism of circadian rhythm's involvement in tumor and its microenvironment regulation. A deeper understanding of circadian rhythms has the potential to provide new strategies for the treatment of cancer and other immune-related diseases.
Collapse
Affiliation(s)
- Yuen Zeng
- Department of Immunology, School of Basic Medical Sciences, Air Force Medical University, Xi'an, China
| | - Zichan Guo
- Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Mengqi Wu
- Department of Immunology, School of Basic Medical Sciences, Air Force Medical University, Xi'an, China
| | - Fulin Chen
- Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Lihua Chen
- Department of Immunology, School of Basic Medical Sciences, Air Force Medical University, Xi'an, China.
| |
Collapse
|
39
|
Haynes PR, Pyfrom ES, Li Y, Stein C, Cuddapah VA, Jacobs JA, Yue Z, Sehgal A. A neuron-glia lipid metabolic cycle couples daily sleep to mitochondrial homeostasis. Nat Neurosci 2024; 27:666-678. [PMID: 38360946 PMCID: PMC11001586 DOI: 10.1038/s41593-023-01568-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 12/29/2023] [Indexed: 02/17/2024]
Abstract
Sleep is thought to be restorative to brain energy homeostasis, but it is not clear how this is achieved. We show here that Drosophila glia exhibit a daily cycle of glial mitochondrial oxidation and lipid accumulation that is dependent on prior wake and requires the Drosophila APOE orthologs NLaz and GLaz, which mediate neuron-glia lipid transfer. In turn, a full night of sleep is required for glial lipid clearance, mitochondrial oxidative recovery and maximal neuronal mitophagy. Knockdown of neuronal NLaz causes oxidative stress to accumulate in neurons, and the neuronal mitochondrial integrity protein, Drp1, is required for daily glial lipid accumulation. These data suggest that neurons avoid accumulation of oxidative mitochondrial damage during wake by using mitophagy and passing damage to glia in the form of lipids. We propose that a mitochondrial lipid metabolic cycle between neurons and glia reflects a fundamental function of sleep relevant for brain energy homeostasis.
Collapse
Affiliation(s)
- Paula R Haynes
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Elana S Pyfrom
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yongjun Li
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Carly Stein
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Vishnu Anand Cuddapah
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Jack A Jacobs
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Zhifeng Yue
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Amita Sehgal
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA.
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Wen D, Xie J, Yuan Y, Shen L, Yang Y, Chen W. The endogenous antioxidant ability of royal jelly in Drosophila is independent of Keap1/Nrf2 by activating oxidoreductase activity. INSECT SCIENCE 2024; 31:503-523. [PMID: 37632209 DOI: 10.1111/1744-7917.13252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/07/2023] [Accepted: 06/14/2023] [Indexed: 08/27/2023]
Abstract
Royal jelly (RJ) is a biologically active substance secreted by the hypopharyngeal and mandibular glands of worker honeybees. It is widely claimed that RJ reduces oxidative stress. However, the antioxidant activity of RJ has mostly been determined by in vitro chemical detection methods or by external administration drugs that cause oxidative stress. Whether RJ can clear the endogenous production of reactive oxygen species (ROS) in cells remains largely unknown. Here, we systematically investigated the antioxidant properties of RJ using several endogenous oxidative stress models of Drosophila. We found that RJ enhanced sleep quality of aging Drosophila, which is decreased due to an increase of oxidative damage with age. RJ supplementation improved survival and suppressed ROS levels in gut cells of flies upon exposure to hydrogen peroxide or to the neurotoxic agent paraquat. Moreover, RJ supplementation moderated levels of ROS in endogenous gut cells and extended lifespan after exposure of flies to heat stress. Sleep deprivation leads to accumulation of ROS in the gut cells, and RJ attenuated the consequences of oxidative stress caused by sleep loss and prolonged lifespan. Mechanistically, RJ prevented cell oxidative damage caused by heat stress or sleep deprivation, with the antioxidant activity in vivo independent of Keap1/Nrf2 signaling. RJ supplementation activated oxidoreductase activity in the guts of flies, suggesting its ability to inhibit endogenous oxidative stress and maintain health, possibly in humans.
Collapse
Affiliation(s)
- Dongjing Wen
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Jiayu Xie
- School of Medicine, Chongqing University, Chongqing, China
| | - Yao Yuan
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Lirong Shen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Yufeng Yang
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Wenfeng Chen
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| |
Collapse
|
41
|
Lutfy RH, Essawy AE, Mohammed HS, Shakweer MM, Salam SA. Transcranial Irradiation Mitigates Paradoxical Sleep Deprivation Effect in an Age-Dependent Manner: Role of BDNF and GLP-1. Neurochem Res 2024; 49:919-934. [PMID: 38114728 PMCID: PMC10902205 DOI: 10.1007/s11064-023-04071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/15/2023] [Accepted: 11/18/2023] [Indexed: 12/21/2023]
Abstract
The growing prevalence of aged sleep-deprived nations is turning into a pandemic state. Acute sleep deprivation (SD) accompanies aging, changing the hippocampal cellular pattern, neurogenesis pathway expression, and aggravating cognitive deterioration. The present study investigated the ability of Near Infra Red (NIR) light laser to ameliorate cognitive impairment induced by SD in young and senile rats. Wistar rats ≤ 2 months (young) and ≥ 14 months (senile) were sleep-deprived for 72 h with or without transcranial administration of NIR laser of 830 nm. Our results showed that NIR photobiomodulation (PBM) attenuated cognitive deterioration made by SD in young, but not senile rats, while both sleep-deprived young and senile rats exhibited decreased anxiety (mania)-like behavior in response to PBM. NIR PBM had an inhibitory effect on AChE, enhanced the production of ACh, attenuated ROS, and regulated cell apoptosis factors such as Bax and Bcl-2. NIR increased mRNA expression of BDNF and GLP-1 in senile rats, thus facilitating neuronal survival and differentiation. The present findings also revealed that age exerts an additive factor to the cellular assaults produced by SD where hippocampal damages made in 2-month rats were less severe than those of the aged one. In conclusion, NIR PBM seems to promote cellular longevity of senile hippocampal cells by combating ROS, elevating neurotrophic factors, thus improving cognitive performance. The present findings provide NIR as a possible candidate for hippocampal neuronal insults accompanying aging and SD.
Collapse
Affiliation(s)
- Radwa H Lutfy
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, 11829, Egypt
| | - Amina E Essawy
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
| | - Haitham S Mohammed
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt
| | - Marwa M Shakweer
- Department of Pathology, Faculty of Medicine, Badr University in Cairo (BUC), Cairo, Egypt
- Department of Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sherine Abdel Salam
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt.
| |
Collapse
|
42
|
Bose K, Agrawal R, Sairam T, Mil J, Butler MP, Dhandapany PS. Sleep fragmentation induces heart failure in a hypertrophic cardiomyopathy mouse model by altering redox metabolism. iScience 2024; 27:109075. [PMID: 38361607 PMCID: PMC10867644 DOI: 10.1016/j.isci.2024.109075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/11/2023] [Accepted: 01/26/2024] [Indexed: 02/17/2024] Open
Abstract
Sleep fragmentation (SF) disrupts normal biological rhythms and has major impacts on cardiovascular health; however, it has never been shown to be a risk factor involved in the transition from cardiac hypertrophy to heart failure (HF). We now demonstrate devastating effects of SF on hypertrophic cardiomyopathy (HCM). We generated a transgenic mouse model harboring a patient-specific myosin binding protein C3 (MYBPC3) variant displaying HCM, and measured the progression of pathophysiology in the presence and absence of SF. SF induces mitochondrial damage, sarcomere disarray, and apoptosis in HCM mice; these changes result in a transition of hypertrophy to an HF phenotype by chiefly targeting redox metabolic pathways. Our findings for the first time show that SF is a risk factor for HF transition and have important implications in clinical settings where HCM patients with sleep disorders have worse prognosis, and strategic intervention with regularized sleep patterns might help such patients.
Collapse
Affiliation(s)
- Karthikeyan Bose
- The Knight Cardiovascular Institute and Departments of Medicine, Molecular, and Medical Genetics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Radhika Agrawal
- Cardiovascular Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine, Bangalore (DBT-inStem), Bangalore, India
| | - Thiagarajan Sairam
- Cardiovascular Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine, Bangalore (DBT-inStem), Bangalore, India
| | - Jessenya Mil
- The Knight Cardiovascular Institute and Departments of Medicine, Molecular, and Medical Genetics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Matthew P. Butler
- Oregon Institute of Occupational Health Sciences, and Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239, USA
| | - Perundurai S. Dhandapany
- The Knight Cardiovascular Institute and Departments of Medicine, Molecular, and Medical Genetics, Oregon Health and Science University, Portland, OR 97239, USA
- Cardiovascular Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine, Bangalore (DBT-inStem), Bangalore, India
| |
Collapse
|
43
|
Guzman RM, Savolainen NG, Hayden OM, Lee M, Osbron CA, Liu Z, Yang H, Shaw DK, Omsland A, Goodman AG. Drosophila melanogaster Sting mediates Coxiella burnetii infection by reducing accumulation of reactive oxygen species. Infect Immun 2024; 92:e0056022. [PMID: 38363133 PMCID: PMC10929449 DOI: 10.1128/iai.00560-22] [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: 08/07/2023] [Accepted: 01/31/2024] [Indexed: 02/17/2024] Open
Abstract
The Gram-negative bacterium Coxiella burnetii is the causative agent of query fever in humans and coxiellosis in livestock. C. burnetii infects a variety of cell types, tissues, and animal species including mammals and arthropods, but there is much left to be understood about the molecular mechanisms at play during infection in distinct species. Human stimulator of interferon genes (STING) induces an innate immune response through the induction of type I interferons (IFNs), and IFN promotes or suppresses C. burnetii replication, depending on tissue type. Drosophila melanogaster contains a functional STING ortholog (Sting) which activates NF-κB signaling and autophagy. Here, we sought to address the role of D. melanogaster Sting during C. burnetii infection to uncover how Sting regulates C. burnetii infection in flies. We show that Sting-null flies exhibit higher mortality and reduced induction of antimicrobial peptides following C. burnetii infection compared to control flies. Additionally, Sting-null flies induce lower levels of oxidative stress genes during infection, but the provision of N-acetyl-cysteine (NAC) in food rescues Sting-null host survival. Lastly, we find that reactive oxygen species levels during C. burnetii infection are higher in Drosophila S2 cells knocked down for Sting compared to control cells. Our results show that at the host level, NAC provides protection against C. burnetii infection in the absence of Sting, thus establishing a role for Sting in protection against oxidative stress during C. burnetii infection.
Collapse
Affiliation(s)
- Rosa M. Guzman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Nathan G. Savolainen
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Olivia M. Hayden
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Miyoung Lee
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Chelsea A. Osbron
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Ziying Liu
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Hong Yang
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Dana K. Shaw
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Anders Omsland
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Alan G. Goodman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| |
Collapse
|
44
|
Zhao J, Ji Y, Zuo Y, Zhang L, Ku C, Wang W, Wang P, Yang Y, Kang Y, Wang F. Association of Oxidative Stress and Proinflammation with Insomnia in Perimenopause. J Womens Health (Larchmt) 2024; 33:379-387. [PMID: 38394165 DOI: 10.1089/jwh.2023.0316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024] Open
Abstract
Background: The levels of oxidative stress and proinflammatory factors in perimenopausal females increased, and they were also deeply troubled by insomnia. The occurrence of insomnia is related to the changes of oxidative stress and inflammation levels in the body. Perimenopausal insomnia may be related to mild systemic inflammation, and oxidative stress can promote chronic inflammation. However, the underlying mechanism behind the phenomenon is still unclear. Objective: The aim was to investigate whether the occurrence of perimenopausal insomnia disorder is related to higher levels of oxidative stress and inflammation in the body, and to explore the role of inducible nitric oxide synthase (iNOS) in perimenopausal insomnia. Methods: A total of 127 perimenopausal participants were recruited in this study. Participants with global scores of the Pittsburgh sleep quality index (PSQI) >7 were diagnosed with insomnia (n = 54). The patient health questionnaire-9 (PHQ-9) and generalized anxiety disorder-7 (GAD-7) were evaluated, and sociodemographic data were obtained. The serum concentrations of iNOS, interleukin 6 (IL6), and tumor necrosis factor α (TNFα) were measured using commercial assays. Results: In the insomnia group, IL6 levels were positively correlated with scores of component 5 and component 7 of PSQI, respectively. PHQ-9 and GAD-7 were positively correlated with the global score of PSQI component 7 and PSQI, respectively; PHQ-9 was positively correlated with the global score of PSQI component 1. Finally, PHQ-9, iNOS, and IL6 were found to be independent predictors of perimenopausal insomnia using logistic regression. Conclusions: Moderate oxidative stress caused by a certain concentration of iNOS plays a protective role in perimenopausal insomnia, while proinflammation and depression are potential risk factors.
Collapse
Affiliation(s)
- Jing Zhao
- Medical Neurobiology Lab, Inner Mongolia Medical University, Huhhot, China
- Department of Basic Medical Teaching and Research, Ordos Institute of Technology, Ordos, China
| | - Yubo Ji
- Department of Medical Psychology, Inner Mongolia Medical University, Huhhot, China
| | - Yanni Zuo
- Physical Examination Center, Beijing Changping Hospital of Chinese Medicine, Beijing, China
| | - Long Zhang
- Department of Gynaecology and Obstetrics, the First Affiliated Hospital of Inner Mongolia Medical University, Huhhot, China
| | - Congwen Ku
- Dongzhimen Hospital, the First Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Wenyan Wang
- School of Pharmacy, Yantai University, Yantai, China
| | - Pengxiang Wang
- Medical Neurobiology Lab, Inner Mongolia Medical University, Huhhot, China
| | - Yan Yang
- Urumqi Fourth People's Hospital, Urumqi, China
| | - Yimin Kang
- Medical Neurobiology Lab, Inner Mongolia Medical University, Huhhot, China
| | - Fan Wang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, China
| |
Collapse
|
45
|
Heiliczer S, Yanko R, Sharav Y, Aframian DJ, Klutstein M, Wilensky A, Haviv Y. Oxidative stress-mediated proapoptosis signaling: A novel theory on the mechanism underlying the pathogenesis of burning mouth syndrome. J Am Dent Assoc 2024; 155:258-267. [PMID: 37966403 DOI: 10.1016/j.adaj.2023.08.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 08/16/2023] [Accepted: 08/30/2023] [Indexed: 11/16/2023]
Abstract
BACKGROUND Burning mouth syndrome (BMS) is a chronic oral pain disorder characterized by a generalized burning sensation in the oral mucosa without apparent medical or dental causes. Despite various hypotheses proposed to explain BMS pathogenesis, a clear understanding of the cellular-level events and associated histologic and molecular findings is lacking. Advancing our understanding of BMS pathogenesis could facilitate the development of more targeted therapeutic interventions. TYPES OF STUDIES REVIEWED The authors conducted an extensive literature search and review of cellular mechanisms, focusing on evidence-based data that support a comprehensive hypothesis for BMS pathogenesis. The authors explored novel and detailed mechanisms that may account for the characteristic features of BMS. RESULTS The authors proposed that BMS symptoms arise from the uncontrolled activation of proapoptotic transmembrane calcium permeable channels expressed in intraoral mucosal nerve fibers. Elevated levels of reactive oxygen species or dysfunctional antiapoptosis pathways may lead to uncontrolled oxidative stress-mediated apoptosis signaling, resulting in upregulation of transmembrane transient receptor potential vanilloid type 1 and P2X 3 calcium channels in nociceptive fibers. Activation of these channels can cause nerve terminal depolarization, leading to generation of action potentials that are centrally interpreted as pain. CONCLUSIONS AND PRACTICAL IMPLICATIONS The authors present a novel hypothesis for BMS pathogenesis, highlighting the role of proapoptotic transmembrane calcium permeable channels and oxidative stress-mediated apoptosis signaling in the development of BMS symptoms. Understanding these underlying mechanisms could provide new insights into the development of targeted therapeutic interventions for BMS. Additional research is warranted to validate this hypothesis and explore potential avenues for effective management of BMS.
Collapse
|
46
|
Alaasam VJ, Hui C, Lomas J, Ferguson SM, Zhang Y, Yim WC, Ouyang JQ. What happens when the lights are left on? Transcriptomic and phenotypic habituation to light pollution. iScience 2024; 27:108864. [PMID: 38318353 PMCID: PMC10839644 DOI: 10.1016/j.isci.2024.108864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/30/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Artificial light at night (ALAN) is a ubiquitous pollutant worldwide. Exposure can induce immediate behavioral and physiological changes in animals, sometimes leading to severe health consequences. Nevertheless, many organisms persist in light-polluted environments and may have mechanisms of habituating, reducing responses to repeated exposure over time, but this has yet to be tested experimentally. Here, we tested whether zebra finches (Taeniopygia guttata) can habituate to dim (0.3 lux) ALAN, measuring behavior, physiology (oxidative stress and telomere attrition), and gene expression in a repeated measures design, over 6 months. We present evidence of tolerance to chronic exposure, persistent behavioral responses lasting 8 weeks post-exposure, and attenuation of responses to re-exposure. Oxidative stress decreased under chronic ALAN. Changes in the blood transcriptome revealed unique responses to past exposure and re-exposure. Results demonstrate organismal resilience to chronic stressors and shed light on the capacity of birds to persist in an increasingly light-polluted world.
Collapse
Affiliation(s)
| | - Cassandra Hui
- Department of Biology, University of Nevada-Reno, Reno 89503, NV, USA
| | - Johnathan Lomas
- Department of Biochemistry & Molecular Biology, University of Nevada-Reno, Reno 89503, NV, USA
| | | | - Yong Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda Genomic Resource Center, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Won Cheol Yim
- Department of Biochemistry & Molecular Biology, University of Nevada-Reno, Reno 89503, NV, USA
| | - Jenny Q. Ouyang
- Department of Biology, University of Nevada-Reno, Reno 89503, NV, USA
| |
Collapse
|
47
|
Tener SJ, Lin Z, Park SJ, Oraedu K, Ulgherait M, Van Beek E, Martínez-Muñiz A, Pantalia M, Gatto JA, Volpi J, Stavropoulos N, Ja WW, Canman JC, Shirasu-Hiza M. Neuronal knockdown of Cullin3 as a Drosophila model of autism spectrum disorder. Sci Rep 2024; 14:1541. [PMID: 38233464 PMCID: PMC10794434 DOI: 10.1038/s41598-024-51657-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 01/06/2024] [Indexed: 01/19/2024] Open
Abstract
Mutations in Cullin-3 (Cul3), a conserved gene encoding a ubiquitin ligase, are strongly associated with autism spectrum disorder (ASD). Here, we characterize ASD-related pathologies caused by neuron-specific Cul3 knockdown in Drosophila. We confirmed that neuronal Cul3 knockdown causes short sleep, paralleling sleep disturbances in ASD. Because sleep defects and ASD are linked to metabolic dysregulation, we tested the starvation response of neuronal Cul3 knockdown flies; they starved faster and had lower triacylglyceride levels than controls, suggesting defects in metabolic homeostasis. ASD is also characterized by increased biomarkers of oxidative stress; we found that neuronal Cul3 knockdown increased sensitivity to hyperoxia, an exogenous oxidative stress. Additional hallmarks of ASD are deficits in social interactions and learning. Using a courtship suppression assay that measures social interactions and memory of prior courtship, we found that neuronal Cul3 knockdown reduced courtship and learning compared to controls. Finally, we found that neuronal Cul3 depletion alters the anatomy of the mushroom body, a brain region required for memory and sleep. Taken together, the ASD-related phenotypes of neuronal Cul3 knockdown flies establish these flies as a genetic model to study molecular and cellular mechanisms underlying ASD pathology, including metabolic and oxidative stress dysregulation and neurodevelopment.
Collapse
Affiliation(s)
- Samantha J Tener
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Zhi Lin
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Scarlet J Park
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, 33458, USA
| | - Kairaluchi Oraedu
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Matthew Ulgherait
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Emily Van Beek
- Waksman Institute, Rutgers University, Piscataway, NJ, 08854, USA
| | - Andrés Martínez-Muñiz
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Meghan Pantalia
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jared A Gatto
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Julia Volpi
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | | | - William W Ja
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, 33458, USA
| | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
48
|
Han S, Kim DK, Jun SE, Kim N. Association of sleep quality and mitochondrial DNA copy number in healthy middle-aged adults. Sleep Med 2024; 113:19-24. [PMID: 37979503 DOI: 10.1016/j.sleep.2023.11.011] [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: 09/10/2023] [Revised: 11/03/2023] [Accepted: 11/04/2023] [Indexed: 11/20/2023]
Abstract
OBJECTIVES Mitochondria contribute to various compromised health, yet the association between sleep and mitochondria remains unclear. This study investigated the association between sleep quality and mitochondrial function in healthy middle-aged adults in the Republic of Korea. METHOD This cross-sectional study recruited 238 middle-aged adults using convenience sampling. Sleep quality was assessed using the Pittsburgh Sleep Quality Index (PSQI). Mitochondrial function, represented by mitochondrial DNA copy number (mtDNAcn), was measured using real-time quantitative polymerase chain reaction on peripheral blood leukocytes. Multivariate linear regression analyses were performed to determine the association between sleep quality and mtDNAcn. RESULTS Sleep quality was negatively associated with mtDNAcn (r = -.15, p = .025); the poor sleep quality group had a notably lower mtDNAcn compared to the good sleep quality group (t = 2.40, p = .017). Among the PSQI components, sleep latency was significantly associated with reduced mtDNAcn (r = -.18, p = .005). Univariate regression analysis revealed that mtDNAcn was significantly associated with education level (β = 0.15, p = .017), shift work (β = -0.17, p = .010), global PSQI score (β = -0.15, p = .025), and sleep latency (β = -0.18, p = .005). After adjusting for educational level and shift work in the final model, longer sleep latency was independently associated with reduced mtDNAcn (β = -.16, p = .011). CONCLUSIONS Poor sleep quality is associated with reduced mtDNAcn, suggesting a potential biological mechanism whereby poor sleep quality, specifically long sleep latency, accelerates cellular aging and impairs health through mitochondrial dysfunction. These findings enhance our understanding of the health effects of sleep quality and highlight the importance of screening and intervention strategies for mitochondrial dysfunction.
Collapse
Affiliation(s)
- Seolbin Han
- College of Nursing, Keimyung University, Daegu, 42601, Republic of Korea
| | - Dae-Kwang Kim
- School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea
| | - Sang-Eun Jun
- College of Nursing, Keimyung University, Daegu, 42601, Republic of Korea
| | - Nahyun Kim
- College of Nursing, Keimyung University, Daegu, 42601, Republic of Korea.
| |
Collapse
|
49
|
Bushana PN, Schmidt MA, Rempe MJ, Sorg BA, Wisor JP. Chronic dietary supplementation with nicotinamide riboside reduces sleep need in the laboratory mouse. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2023; 4:zpad044. [PMID: 38152423 PMCID: PMC10752388 DOI: 10.1093/sleepadvances/zpad044] [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: 06/28/2023] [Revised: 10/13/2023] [Indexed: 12/29/2023]
Abstract
Non-rapid eye movement sleep (NREMS) is accompanied by a reduction in cerebral glucose utilization. Enabling this metabolic change may be a central function of sleep. Since the reduction in glucose metabolism is inevitably accompanied by deceleration of downstream oxidation/reduction reactions involving nicotinamide adenine dinucleotide (NAD), we hypothesized a role for NAD in regulating the homeostatic dynamics of sleep at the biochemical level. We applied dietary nicotinamide riboside (NR), a NAD precursor, in a protocol known to improve neurological outcome measures in mice. Long-term (6-10 weeks) dietary supplementation with NR reduced the time that mice spent in NREMS by 17 percent and accelerated the rate of discharge of sleep need according to a mathematical model of sleep homeostasis (Process S). These findings suggest that increasing redox capacity by increasing nicotinamide availability reduces sleep need and increases the cortical capacity for energetically demanding high-frequency oscillations. In turn, this work demonstrates the impact of redox substrates on cortical circuit properties related to fatigue and sleep drive, implicating redox reactions in the homeostatic dynamics of cortical network events across sleep-wake cycles.
Collapse
Affiliation(s)
- Priyanka N Bushana
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Michelle A Schmidt
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Michael J Rempe
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Barbara A Sorg
- R.S. Dow Neuroscience Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| | - Jonathan P Wisor
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| |
Collapse
|
50
|
Brown EB, Zhang J, Lloyd E, Lanzon E, Botero V, Tomchik S, Keene AC. Neurofibromin 1 mediates sleep depth in Drosophila. PLoS Genet 2023; 19:e1011049. [PMID: 38091360 PMCID: PMC10763969 DOI: 10.1371/journal.pgen.1011049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/03/2024] [Accepted: 11/03/2023] [Indexed: 01/04/2024] Open
Abstract
Neural regulation of sleep and metabolic homeostasis are critical in many aspects of human health. Despite extensive epidemiological evidence linking sleep dysregulation with obesity, diabetes, and metabolic syndrome, little is known about the neural and molecular basis for the integration of sleep and metabolic function. The RAS GTPase-activating gene Neurofibromin (Nf1) has been implicated in the regulation of sleep and metabolic rate, raising the possibility that it serves to integrate these processes, but the effects on sleep consolidation and physiology remain poorly understood. A key hallmark of sleep depth in mammals and flies is a reduction in metabolic rate during sleep. Here, we examine multiple measures of sleep quality to determine the effects of Nf1 on sleep-dependent changes in arousal threshold and metabolic rate. Flies lacking Nf1 fail to suppress metabolic rate during sleep, raising the possibility that loss of Nf1 prevents flies from integrating sleep and metabolic state. Sleep of Nf1 mutant flies is fragmented with a reduced arousal threshold in Nf1 mutants, suggesting Nf1 flies fail to enter deep sleep. The effects of Nf1 on sleep can be localized to a subset of neurons expressing the GABAA receptor Rdl. Sleep loss has been associated with changes in gut homeostasis in flies and mammals. Selective knockdown of Nf1 in Rdl-expressing neurons within the nervous system increases gut permeability and reactive oxygen species (ROS) in the gut, raising the possibility that loss of sleep quality contributes to gut dysregulation. Together, these findings suggest Nf1 acts in GABA-sensitive neurons to modulate sleep depth in Drosophila.
Collapse
Affiliation(s)
- Elizabeth B. Brown
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
- Department of Biological Sciences, Florida State University, Tallahassee, Florida, United States of America
| | - Jiwei Zhang
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Evan Lloyd
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Elizabeth Lanzon
- Jupiter Life Science Initiative, Florida Atlantic University, Jupiter, Florida, United States of America
| | - Valentina Botero
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Seth Tomchik
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Alex C. Keene
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|