1
|
Killick R, Stranks L, Hoyos CM. Sleep Deficiency and Cardiometabolic Disease. Sleep Med Clin 2024; 19:653-670. [PMID: 39455184 DOI: 10.1016/j.jsmc.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
Epidemiologic studies have demonstrated that short sleep duration is associated with an increased risk of cardio-metabolic health outcomes including cardiovascular disease mortality, coronary heart disease, type 2 diabetes mellitus, hypertension, and metabolic syndrome. Experimental sleep restriction studies have sought to explain these findings. This review describes the main evidence of these associations and possible mechanisms explaining them. Whether sleep extension reverses these now widely acknowledged adverse health effects and the feasibility of implementing such strategies on a public health level is discussed.
Collapse
Affiliation(s)
- Roo Killick
- Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Lachlan Stranks
- Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, University of Sydney, Sydney, Australia; The University of Adelaide, Faculty of Health and Medical Sciences, Adelaide, Australia
| | - Camilla M Hoyos
- Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, University of Sydney, Sydney, Australia; The University of Sydney, Faculty of Science, School of Psychology and Brain and Mind Centre, Sydney, Australia.
| |
Collapse
|
2
|
Weng L, Hong H, Zhang Q, Xiao C, Zhang Q, Wang Q, Huang J, Lai D. Sleep Deprivation Triggers the Excessive Activation of Ovarian Primordial Follicles via β2 Adrenergic Receptor Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402393. [PMID: 39229959 PMCID: PMC11538700 DOI: 10.1002/advs.202402393] [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: 03/05/2024] [Revised: 07/23/2024] [Indexed: 09/05/2024]
Abstract
Sleep deprivation (SD) is observed to adversely affect the reproductive health of women. However, its precise physiological mechanisms remain largely elusive. In this study, using a mouse model of SD, it is demonstrated that SD induces the depletion of ovarian primordial follicles, a phenomenon not attributed to immune-mediated attacks or sympathetic nervous system activation. Rather, the excessive secretion of stress hormones, namely norepinephrine (NE) and epinephrine (E), by overactive adrenal glands, has emerged as a key mediator. The communication pathway mediated by the KIT ligand (KITL)-KIT between granulosa cells and oocytes plays a pivotal role in primordial follicle activation. SD heightened the levels of NE/E that stimulates the activation of the KITL-KIT/PI3K and mTOR signaling cascade in an β2 adrenergic receptor (ADRB2)-dependent manner, thereby promoting primordial follicle activation and consequent primordial follicle loss in vivo. In vitro experiments further corroborate these observations, revealing that ADRB2 upregulates KITL expression in granulosa cells via the activation of the downstream cAMP/PKA pathway. Together, these results reveal the significant involvement of ADRB2 signaling in the depletion of ovarian primordial follicles under sleep-deprived conditions. Additionally, ADRB2 antagonists are proposed for the treatment or prevention of excessive activation of primordial follicles induced by SD.
Collapse
Affiliation(s)
- Lichun Weng
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Hanqing Hong
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Qinyu Zhang
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Chengqi Xiao
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Qiuwan Zhang
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Qian Wang
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Ju Huang
- Songjiang Hospital and Songjiang Research InstituteShanghai Key Laboratory of Emotions and Affective DisordersShanghai Jiao Tong University School of MedicineShanghai201600China
| | - Dongmei Lai
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| |
Collapse
|
3
|
Wu P, Wang W, Huang C, Sun L, Wu X, Xu L, Xiao P. A rapid and reliable targeted LC-MS/MS method for quantitative analysis of the Tryptophan-NAD metabolic network disturbances in tissues and blood of sleep deprivation mice. Anal Chim Acta 2024; 1328:343125. [PMID: 39266191 DOI: 10.1016/j.aca.2024.343125] [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: 04/20/2024] [Revised: 08/12/2024] [Accepted: 08/18/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND TRY-NAD metabolic network includes TRY (tryptophan), 5-HT (5-hydroxytryptamine), KYN (kynurenine), and NAD (nicotinamide adenine dinucleotide) pathway, which plays a significant role in neurological diseases and ageing. It is important to monitor these metabolites for studying the pathological anatomy of disease and treatment of responses evaluation. Although previous studies have reported quantitative methods for several metabolites in the network, the bottlenecks of simultaneously quantifying the whole metabolic network are their similar structures, diverse physico-chemical properties, and instability. Standardized protocols for the whole metabolic network are still missing, which hinders the in-depth study of TRY-NAD metabolic network in laboratory research and clinical screening. RESULTS We developed a LC-MS/MS method for quantifying 28 metabolites in the TRY-NAD network simultaneously. Optimization was done for the mass spectral parameters, chromatographic conditions and sample pretreatment process. The developed method was fully validated in terms of standard curves, sensitivity, carryover, recovery, matrix effect, accuracy, precision, and stability. The pretreatment of 30 samples only takes 90 min, and the LC-MS/MS running time of one sample is only 13 min. With this method, we bring to light the chaos of global TRY-NAD metabolic network in sleep deprivation mice for the first time, including serum, clotted blood cells, hippocampus, cerebral cortex, and liver. NAD pathway levels in brain and blood decreased, whereas the opposite happened in the liver. The 5-HT pathway decreased and the concentration of KYN increased significantly in the brain. The concentration of many metabolites in KYN pathway (NAD+ de novo synthesis pathway) increased in the liver. SIGNIFICANCE This method is the first time to determine the metabolites of KYN, 5-HT and NAD pathway at the same time, and it is found that TRY-NAD metabolic network will be disordered after sleep deprivation. This work clarifies the importance of the pH of the extraction solution, the time and temperature control in pretreatment in standardized protocols building, and overcoming the problems of inconsistent sample pretreatment, separation, matrix effect interference and potential metabolite degradation. This method exhibits great prospects in providing more information on metabolic disturbances caused by sleep deprivation as well as neurological diseases and ageing.
Collapse
Affiliation(s)
- Peiling Wu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Wenjie Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Chuan Huang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Le Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Xiaoli Wu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Lijia Xu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China; Key Laboratory of Resources Conservation and Development of Southern Medicine of Hainan Province & Key Laboratory of State Administration of Traditional Chinese Medicine for Agarwood Sustainable Utilization, Hainan Branch of the Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Haikou, 570311, China.
| | - Peigen Xiao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China.
| |
Collapse
|
4
|
Malvandi H, Fallahi M, Saghi MH, Hassanzadeh N. Evaluation of electric field (E) exposure levels and its relationship with the sleep quality of residents around the BTS antennas in Sabzevar, Iran. RADIATION PROTECTION DOSIMETRY 2024; 200:1405-1415. [PMID: 39214550 DOI: 10.1093/rpd/ncae180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/24/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Mobile devices and base transceiver station (BTS) are the main sources of human exposure to radio frequency electromagnetic fields (RF-EMFs). Therefore, the aim of the present study was to evaluate the levels of exposure to RF-EMF in three different time intervals and three different distances from BTS antennas in Sabzevar. Additional goals were to investigate the electric field (E) difference between different microenvironments, between the suburbs and downtown, and evaluating the sleep quality of residents around BTS antennas at different distances. The results showed significant differences between the values of E Avg and E max Avg at different times (T1, T2, and T3), different distances (50, 100, and 300 m) from BTS antennas, and between BTS antennas located in the suburbs and downtown. No significant differences were observed between the values of E Avg and E max Avg in terms of microenvironments. Poor sleep quality (>5) was recorded in the residents around the BTS antennas at different distances, and a significant difference was observed between the sleep quality of the residents at a distance of ˂100 m compared to the residents at a distance of ˃300 m. The recorded levels of E in all places and times were below the human safety limits set by the Iranian National Standardization Organization, the Information and Communication Technologies Authority and the International Commission on Non-Ionizing Radiation Protection, indicating the absence of potential risk due to exposure to E in the study area.
Collapse
Affiliation(s)
- Hassan Malvandi
- Department of Environmental Sciences and Engineering, Faculty of Geography and Environmental Sciences, Hakim Sabzevari University, Tovhid Shahr, 9617976487, Sabzevar, Iran
- EthnoBiology Core, Hakim Sabzevari University, Tovhid Shahr, 9617976487, Sabzevar, Khorasan Razavi, Iran
| | - Majid Fallahi
- Non-Communicable Diseases Research Center, Department of Occupational Health Engineering, School of Public Health, Sabzevar University of Medical Sciences, Tovhid Shahr, 9617913112, Sabzevar, Iran
| | - Mohammad Hossien Saghi
- Non-Communicable Diseases Research Center, Department of Environmental Health Engineering School of Public Health, Sabzevar University of Medical Sciences, Tovhid Shahr, 9617913112, Sabzevar, Iran
| | - Nasrin Hassanzadeh
- Department of Environmental Sciences, Faculty of Environment and Natural Resource, Malayer University, Arak-Malayer Road, 65741-84621, Malayer, Iran
| |
Collapse
|
5
|
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] [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
|
6
|
Huang Z, Tian Z, Cui J, Wang G, Chen J. Prevalence of overweight/obesity, and associated factors among adolescents aged 12 ∼ 15 in Shandong Province, China: A cross-sectional study. Prev Med Rep 2024; 45:102831. [PMID: 39193377 PMCID: PMC11347837 DOI: 10.1016/j.pmedr.2024.102831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 08/29/2024] Open
Abstract
Overweight/obesity among adolescents in Shandong Province, China, has been rising, posing significant public health challenge. Comprehensive investigation is needed to develop effective interventions. Following the Strengthening the Reporting of Observational Studies in Epidemiology guidelines, a stratified random cluster sampling approach was used from September to October 2023 across 17 cities in Shandong Province. The study included 165 middle schools, surveying 99,638 students aged 12 ∼ 15. After applying exclusion criteria, 97,356 students (97.71% effective rate) completed anthropometric measurements and questionnaires. Overweight/obesity were assessed based on national and international standards. Univariable chi-square test and multivariable logistic regression were used to analyze factors influencing overweight/obesity. In 2023, the overweight/obesity rate among 12 ∼ 15-year-olds in Shandong was 19.75%. Significant factors included sex, age, residence, family income, parental weight status and activity, mother's gestational diabetes history, birth weight, physical activity, sleep, screen time, homework, and diet. Girls, older adolescents, and those with physically active parents or who themselves engaged in over 1.5 h of daily physical activity had lower odds of being overweight/obese. Adequate sleep and frequent consumption of vegetable and fruit were also protective. Higher odds were associated with urban residence, high family income, overweight/obese parents, maternal gestational diabetes, high birth weight, excessive screen time, extensive homework, and frequent fast food consumption. Overweight/obesity in Shandong adolescents is influenced by multiple determinants. Holistic interventions addressing genetic, behavioral, and environmental factors are essential for promoting healthier lifestyles and reducing the prevalence in this demographic.
Collapse
Affiliation(s)
- Zhihao Huang
- School of Big Data and Fundamental Sciences, Shandong Institute of Petroleum and Chemical Technology, Dongying, China
| | - Zhiqi Tian
- Department of Clinical Laboratory, Shengli Oilfield Central Hospital, China
| | - Jian Cui
- School of Big Data and Fundamental Sciences, Shandong Institute of Petroleum and Chemical Technology, Dongying, China
| | - Guan Wang
- School of Big Data and Fundamental Sciences, Shandong Institute of Petroleum and Chemical Technology, Dongying, China
| | - Jiyan Chen
- School of Big Data and Fundamental Sciences, Shandong Institute of Petroleum and Chemical Technology, Dongying, China
| |
Collapse
|
7
|
Witkowska A, Jaromirska J, Gabryelska A, Sochal M. Obstructive Sleep Apnea and Serotoninergic Signalling Pathway: Pathomechanism and Therapeutic Potential. Int J Mol Sci 2024; 25:9427. [PMID: 39273373 PMCID: PMC11395478 DOI: 10.3390/ijms25179427] [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: 07/18/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Obstructive Sleep Apnea (OSA) is a disorder characterized by repeated upper airway collapse during sleep, leading to apneas and/or hypopneas, with associated symptoms like intermittent hypoxia and sleep fragmentation. One of the agents contributing to OSA occurrence and development seems to be serotonin (5-HT). Currently, the research focuses on establishing and interlinking OSA pathogenesis and the severity of the disease on the molecular neurotransmitter omnipresent in the human body-serotonin, its pathway, products, receptors, drugs affecting the levels of serotonin, or genetic predisposition. The 5-HT system is associated with numerous physiological processes such as digestion, circulation, sleep, respiration, and muscle tone-all of which are considered factors promoting and influencing the course of OSA because of correlations with comorbid conditions. Comorbidities include obesity, physiological and behavioral disorders as well as cardiovascular diseases. Additionally, both serotonin imbalance and OSA are connected with psychiatric comorbidities, such as depression, anxiety, or cognitive dysfunction. Pharmacological agents that target 5-HT receptors have shown varying degrees of efficacy in reducing the Apnea-Hypopnea Index and improving OSA symptoms. The potential role of the 5-HT signaling pathway in modulating OSA provides a promising avenue for new therapeutic interventions that could accompany the primary treatment of OSA-continuous positive airway pressure. Thus, this review aims to elucidate the complex role of 5-HT and its regulatory mechanisms in OSA pathophysiology, evaluating its potential as a therapeutic target. We also summarize the relationship between 5-HT signaling and various physiological functions, as well as its correlations with comorbid conditions.
Collapse
Affiliation(s)
- Alicja Witkowska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
| | - Julia Jaromirska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
| | - Agata Gabryelska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
| | - Marcin Sochal
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
| |
Collapse
|
8
|
Scholz M, Steuer AE, Dobay A, Landolt HP, Kraemer T. Assessing the influence of sleep and sampling time on metabolites in oral fluid: implications for metabolomics studies. Metabolomics 2024; 20:97. [PMID: 39112673 PMCID: PMC11306311 DOI: 10.1007/s11306-024-02158-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 07/20/2024] [Indexed: 08/10/2024]
Abstract
INTRODUCTION The human salivary metabolome is a rich source of information for metabolomics studies. Among other influences, individual differences in sleep-wake history and time of day may affect the metabolome. OBJECTIVES We aimed to characterize the influence of a single night of sleep deprivation compared to sufficient sleep on the metabolites present in oral fluid and to assess the implications of sampling time points for the design of metabolomics studies. METHODS Oral fluid specimens of 13 healthy young males were obtained in Salivette® devices at regular intervals in both a control condition (repeated 8-hour sleep) and a sleep deprivation condition (total sleep deprivation of 8 h, recovery sleep of 8 h) and their metabolic contents compared in a semi-targeted metabolomics approach. RESULTS Analysis of variance results showed factor 'time' (i.e., sampling time point) representing the major influencer (median 9.24%, range 3.02-42.91%), surpassing the intervention of sleep deprivation (median 1.81%, range 0.19-12.46%). In addition, we found about 10% of all metabolic features to have significantly changed in at least one time point after a night of sleep deprivation when compared to 8 h of sleep. CONCLUSION The majority of significant alterations in metabolites' abundances were found when sampled in the morning hours, which can lead to subsequent misinterpretations of experimental effects in metabolomics studies. Beyond applying a within-subject design with identical sample collection times, we highly recommend monitoring participants' sleep-wake schedules prior to and during experiments, even if the study focus is not sleep-related (e.g., via actigraphy).
Collapse
Affiliation(s)
- Michael Scholz
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Andrea Eva Steuer
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Akos Dobay
- Forensic Machine Learning Technology Center, University of Zurich, Zurich, Switzerland
| | - Hans-Peter Landolt
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Sleep & Health Zurich, University of Zurich, Zurich, Switzerland
| | - Thomas Kraemer
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland.
- Sleep & Health Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
9
|
Castillo-Peinado LS, Calderón-Santiago M, Jurado-Gámez B, Priego-Capote F. Changes in human sweat metabolome conditioned by severity of obstructive sleep apnea and intermittent hypoxemia. J Sleep Res 2024; 33:e14075. [PMID: 37877569 DOI: 10.1111/jsr.14075] [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: 06/28/2023] [Revised: 09/13/2023] [Accepted: 10/04/2023] [Indexed: 10/26/2023]
Abstract
Obstructive sleep apnea (OSA) is a sleep disorder that has been associated with the incidence of other pathologies. Diagnosis is mainly based on the apnea-hypopnea index (AHI) obviating other repercussions such as intermittent hypoxemia, which has been found to be associated to cardiovascular complications. Blood-based samples and urine have been the most utilised biofluids in metabolomics studies related to OSA, while sweat could be an alternative due to its non-invasive and accessible sampling, its reduced complexity, and comparability with other biofluids. Therefore, this research aimed to evaluate metabolic overnight changes in sweat collected from patients with OSA classified according to the AHI and oxygen desaturation index (ODI), looking for potential cardiovascular repercussions. Pre- and post-sleeping sweat samples from all individuals (n = 61) were analysed by gas chromatography coupled to high-resolution mass spectrometry after appropriate sample preparation to detect as many metabolites as possible. Permanent significant alterations in the sweat were reported for pyruvate, serine, lactose, and hydroxybutyrate. The most relevant overnight metabolic alterations in sweat were reported for lactose, succinate, urea, and oxoproline, which presented significantly different effects on factors such as the AHI and ODI for OSA severity classification. Overall metabolic alterations mainly affected energy production-related processes, nitrogen metabolism, and oxidative stress. In conclusion, this research demonstrated the applicability of sweat for evaluation of OSA diagnosis and severity supported by the detected metabolic changes during sleep.
Collapse
Affiliation(s)
- Laura S Castillo-Peinado
- Department of Analytical Chemistry, Annex Marie Curie Building, Campus of Rabanales, University of Córdoba, Córdoba, Spain
- Maimónides Institute of Biomedical Research (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
- Energy and Environmental Chemistry University Institute (IQUEMA), Campus of Rabanales, University of Córdoba, Córdoba, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Mónica Calderón-Santiago
- Department of Analytical Chemistry, Annex Marie Curie Building, Campus of Rabanales, University of Córdoba, Córdoba, Spain
- Maimónides Institute of Biomedical Research (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
- Energy and Environmental Chemistry University Institute (IQUEMA), Campus of Rabanales, University of Córdoba, Córdoba, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Bernabé Jurado-Gámez
- Maimónides Institute of Biomedical Research (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
- Department of Respiratory Medicine, Reina Sofía University Hospital, Córdoba, Spain
| | - Feliciano Priego-Capote
- Department of Analytical Chemistry, Annex Marie Curie Building, Campus of Rabanales, University of Córdoba, Córdoba, Spain
- Maimónides Institute of Biomedical Research (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
- Energy and Environmental Chemistry University Institute (IQUEMA), Campus of Rabanales, University of Córdoba, Córdoba, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
10
|
Chen L, Wu K, He J, Hou J, Zhang Y, Liu L, Wang J, Xia Z. Circadian Regulation of the Lactate Metabolic Kinetics in Mice Using the [ 1H- 13C]-NMR Technique. Mol Neurobiol 2024; 61:5802-5813. [PMID: 38231323 DOI: 10.1007/s12035-024-03927-w] [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] [Accepted: 01/04/2024] [Indexed: 01/18/2024]
Abstract
Lactate is not only the energy substrate of neural cells, but also an important signal molecule in brain. In modern societies, disturbed circadian rhythms pose a global challenge. Therefore, exploring the influence of circadian period on lactate and its metabolic kinetics is essential for the advancement of neuroscientific research. In the present study, the different groups of mice (L: 8:00 a.m.; D: 20:00 p.m.; SD: 20:00 p.m. with 12 h acute sleep deprivation) were infused with [3-13C] lactate through the lateral tail vein for a duration of 2 min. After 30-min lactate metabolism, the animals were euthanized and the tissues of brain and liver were obtained and extracted, and then, the [1H-13C] NMR technology was employed to investigate the kinetic information of lactate metabolism in different brain regions and liver to detect the enrichment of various metabolic kinetic information. Results revealed the fluctuating lactate concentrations in the brain throughout the day, with lower levels during light periods and higher levels during dark periods. Most metabolites displayed strong sensitivity to circadian rhythm, exhibiting significant day-night variations. Conversely, only a few metabolites showed changes after acute sleep deprivation, primarily in the temporal brain region. Interestingly, in contrast to brain lactate metabolism, liver lactate metabolism exhibited a significant increase following acute sleep deprivation. This study explored the kinetics of lactate metabolism, hinted at potential clinical implications for disorders involving circadian rhythm disturbances, and providing a new research basis for clinical exploration of brain and liver lactate metabolism.
Collapse
Affiliation(s)
- Lili Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Kefan Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Jingang He
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, Hubei, 430071, People's Republic of China
| | - Jiabao Hou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Yuan Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Lian Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| | - Jie Wang
- Songjiang Hospital and Songjiang Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, People's Republic of China.
- Institute of Neuroscience and Brain Diseases; Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021, People's Republic of China.
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China.
| |
Collapse
|
11
|
Zheng ZJ, Zhang HY, Hu YL, Li Y, Wu ZH, Li ZP, Chen DR, Luo Y, Zhang XJ, Li C, Wang XY, Xu D, Qiu W, Li HP, Liao XP, Ren H, Sun J. Sleep Deprivation Induces Gut Damage via Ferroptosis. J Pineal Res 2024; 76:e12987. [PMID: 38975671 DOI: 10.1111/jpi.12987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024]
Abstract
Sleep deprivation (SD) has been associated with a plethora of severe pathophysiological syndromes, including gut damage, which recently has been elucidated as an outcome of the accumulation of reactive oxygen species (ROS). However, the spatiotemporal analysis conducted in this study has intriguingly shown that specific events cause harmful damage to the gut, particularly to goblet cells, before the accumulation of lethal ROS. Transcriptomic and metabolomic analyses have identified significant enrichment of metabolites related to ferroptosis in mice suffering from SD. Further analysis revealed that melatonin could rescue the ferroptotic damage in mice by suppressing lipid peroxidation associated with ALOX15 signaling. ALOX15 knockout protected the mice from the serious damage caused by SD-associated ferroptosis. These findings suggest that melatonin and ferroptosis could be targets to prevent devastating gut damage in animals exposed to SD. To sum up, this study is the first report that proposes a noncanonical modulation in SD-induced gut damage via ferroptosis with a clearly elucidated mechanism and highlights the active role of melatonin as a potential target to maximally sustain the state during SD.
Collapse
Affiliation(s)
- Zi-Jian Zheng
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Hai-Yi Zhang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Ya-Lin Hu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Yan Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Zhi-Hong Wu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Zhi-Peng Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Dong-Rui Chen
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Yang Luo
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Xiao-Jing Zhang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Cang Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Xiao-Yu Wang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Dan Xu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | - Xiao-Ping Liao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Hao Ren
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Jian Sun
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| |
Collapse
|
12
|
Windred DP, Anderson C, Jeppe KJ, Ftouni S, Grant LK, Nijagal B, Rajaratnam SMW, McConville M, Tull D, Lockley SW, Cain SW, Phillips AJK. Higher central circadian temperature amplitude is associated with greater metabolite rhythmicity in humans. Sci Rep 2024; 14:16796. [PMID: 39039133 PMCID: PMC11263371 DOI: 10.1038/s41598-024-67297-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: 02/26/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024] Open
Abstract
Robust circadian rhythms are essential for optimal health. The central circadian clock controls temperature rhythms, which are known to organize the timing of peripheral circadian rhythms in rodents. In humans, however, it is unknown whether temperature rhythms relate to the organization of circadian rhythms throughout the body. We assessed core body temperature amplitude and the rhythmicity of 929 blood plasma metabolites across a 40-h constant routine protocol, controlling for behavioral and environmental factors that mask endogenous temperature rhythms, in 23 healthy individuals (mean [± SD] age = 25.4 ± 5.7 years, 5 women). Valid core body temperature data were available in 17/23 (mean [± SD] age = 25.6 ± 6.3 years, 1 woman). Individuals with higher core body temperature amplitude had a greater number of metabolites exhibiting circadian rhythms (R2 = 0.37, p = .009). Higher core body temperature amplitude was also associated with less variability in the free-fitted periods of metabolite rhythms within an individual (R2 = 0.47, p = .002). These findings indicate that a more robust central circadian clock is associated with greater organization of circadian metabolite rhythms in humans. Metabolite rhythms may therefore provide a window into the strength of the central circadian clock.
Collapse
Affiliation(s)
- Daniel P Windred
- Flinders Health and Medical Research Institute (Sleep Health), Flinders University, Bedford Park, SA, Australia.
- School of Psychological Sciences, Monash University, Clayton, VIC, Australia.
| | - Clare Anderson
- School of Psychological Sciences, Monash University, Clayton, VIC, Australia
- School of Psychology, Centre for Human Brain Health, University of Birmingham, Edgbaston, UK
| | - Katherine J Jeppe
- School of Psychological Sciences, Monash University, Clayton, VIC, Australia
- Monash Proteomics and Metabolomics Platform, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Suzanne Ftouni
- School of Psychological Sciences, Monash University, Clayton, VIC, Australia
| | - Leilah K Grant
- School of Psychological Sciences, Monash University, Clayton, VIC, Australia
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Brunda Nijagal
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - Shantha M W Rajaratnam
- School of Psychological Sciences, Monash University, Clayton, VIC, Australia
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Malcolm McConville
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - Dedreia Tull
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - Steven W Lockley
- School of Psychological Sciences, Monash University, Clayton, VIC, Australia
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Sean W Cain
- Flinders Health and Medical Research Institute (Sleep Health), Flinders University, Bedford Park, SA, Australia.
- School of Psychological Sciences, Monash University, Clayton, VIC, Australia.
| | - Andrew J K Phillips
- Flinders Health and Medical Research Institute (Sleep Health), Flinders University, Bedford Park, SA, Australia.
- School of Psychological Sciences, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
13
|
Di T, Zhang L, Meng S, Liu W, Guo Y, Zheng E, Xie C, Xiang S, Jia T, Lu L, Sun Y, Shi J. The impact of REM sleep loss on human brain connectivity. Transl Psychiatry 2024; 14:270. [PMID: 38956035 PMCID: PMC11219886 DOI: 10.1038/s41398-024-02985-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024] Open
Abstract
Brain function is vulnerable to the consequences of inadequate sleep, an adverse trend that is increasingly prevalent. The REM sleep phase has been implicated in coordinating various brain structures and is hypothesized to have potential links to brain variability. However, traditional imaging research have encountered challenges in attributing specific brain region activity to REM sleep, remained understudied at the whole-brain connectivity level. Through the spilt-night paradigm, distinct patterns of REM sleep phases were observed among the full-night sleep group (n = 36), the early-night deprivation group (n = 41), and the late-night deprivation group (n = 36). We employed connectome-based predictive modeling (CPM) to delineate the effects of REM sleep deprivation on the functional connectivity of the brain (REM connectome) during its resting state. The REM sleep-brain connectome was characterized by stronger connectivity within the default mode network (DMN) and between the DMN and visual networks, while fewer predictive edges were observed. Notably, connections such as those between the cingulo-opercular network (CON) and the auditory network, as well as between the subcortex and visual networks, also made significant contributions. These findings elucidate the neural signatures of REM sleep loss and reveal common connectivity patterns across individuals, validated at the group level.
Collapse
Affiliation(s)
- Tianqi Di
- Department of Pharmacology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China
| | - Libo Zhang
- Department of Pharmacology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Shenzhen Public Service Platform for Clinical Application of Medical Imaging, Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen-PKU-HKUST Medical Center, Shenzhen, 518036, China
| | - Shiqiu Meng
- Department of Pharmacology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China
| | - Wangyue Liu
- Department of Pharmacology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
| | - Yang Guo
- Department of Pharmacology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
| | - Enyu Zheng
- Department of Pharmacology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
| | - Chao Xie
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, China
| | - Shitong Xiang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, China
| | - Tianye Jia
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, China
| | - Lin Lu
- Department of Pharmacology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
| | - Yan Sun
- Department of Pharmacology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China.
- Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China.
| | - Jie Shi
- Department of Pharmacology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China.
- Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China.
- The Key Laboratory for Neuroscience of the Ministry of Education and Health, Peking University, Beijing, 100191, China.
- The State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China.
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453000, China.
| |
Collapse
|
14
|
Daniels M, Margolis LM, Rood JC, Lieberman HR, Pasiakos SM, Karl JP. Comparative analysis of circulating metabolomic profiles identifies shared metabolic alterations across distinct multistressor military training exercises. Physiol Genomics 2024; 56:457-468. [PMID: 38738316 PMCID: PMC11368567 DOI: 10.1152/physiolgenomics.00008.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/26/2024] [Accepted: 05/03/2024] [Indexed: 05/14/2024] Open
Abstract
Military training provides insight into metabolic responses under unique physiological demands that can be comprehensively characterized by global metabolomic profiling to identify potential strategies for improving performance. This study identified shared changes in metabolomic profiles across three distinct military training exercises, varying in magnitude and type of stress. Blood samples collected before and after three real or simulated military training exercises were analyzed using the same untargeted metabolomic profiling platform. Exercises included a 2-wk survival training course (ST, n = 36), a 4-day cross-country ski march arctic training (AT, n = 24), and a 28-day controlled diet- and exercise-induced energy deficit (CED, n = 26). Log2-fold changes of greater than ±1 in 191, 121, and 64 metabolites were identified in the ST, AT, and CED datasets, respectively. Most metabolite changes were within the lipid (57-63%) and amino acid metabolism (18-19%) pathways and changes in 87 were shared across studies. The largest and most consistent increases in shared metabolites were found in the acylcarnitine, fatty acid, ketone, and glutathione metabolism pathways, whereas the largest decreases were in the diacylglycerol and urea cycle metabolism pathways. Multiple shared metabolites were consistently correlated with biomarkers of inflammation, tissue damage, and anabolic hormones across studies. These three studies of real and simulated military training revealed overlapping alterations in metabolomic profiles despite differences in environment and the stressors involved. Consistent changes in metabolites related to lipid metabolism, ketogenesis, and oxidative stress suggest a potential common metabolomic signature associated with inflammation, tissue damage, and suppression of anabolic signaling that may characterize the unique physiological demands of military training.NEW & NOTEWORTHY The extent to which metabolomic responses are shared across diverse military training environments is unknown. Global metabolomic profiling across three distinct military training exercises identified shared metabolic responses with the largest changes observed for metabolites related to fatty acids, acylcarnitines, ketone metabolism, and oxidative stress. These changes also correlated with alterations in markers of tissue damage, inflammation, and anabolic signaling and comprise a potential common metabolomic signature underlying the unique physiological demands of military training.
Collapse
Affiliation(s)
- Michael Daniels
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, United States
| | - Lee M Margolis
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Jennifer C Rood
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States
| | - Harris R Lieberman
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Stefan M Pasiakos
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
- Office of Dietary Supplements, National Institutes of Health, Bethesda, Maryland, United States
| | - J Philip Karl
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| |
Collapse
|
15
|
Liu S, Ma X, Chen Y, Zhao Y, Luo R, Wu Z, Li Y, Qian Y, Wang W, Dong S, Zhou Z, Li S, Xiao Y, Zhu X, Tian Y, Guo J. Multiplex influences on vigilance and biochemical variables induced by sleep deprivation. Front Sports Act Living 2024; 6:1412044. [PMID: 39005627 PMCID: PMC11239445 DOI: 10.3389/fspor.2024.1412044] [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: 04/04/2024] [Accepted: 06/11/2024] [Indexed: 07/16/2024] Open
Abstract
Introduction Sleep loss and sleep deprivation (SD) cause deleterious influences on health, cognition, mood and behaviour. Nevertheless, insufficient sleep and SD are prevalent across many industries and occur in various emergencies. The deleterious consequences of SD have yet to be fully elucidated. This study aimed to assess the extensive influences of SD on physiology, vigilance, and plasma biochemical variables. Methods Seventeen volunteers were recruited to participate in a 32.5-h SD experiment. Multiple physiological and cognitive variables, including tympanic temperature, blood oxygen saturation (SaO2), and vigilance were recorded. Urinal/salivary samples were collected and subjected to cortisol or cortisone analysis, and plasma samples were subjected to transcriptomic analysis of circular RNA (circRNA) expression using microarray. Plasma neurotransmitters were measured by targeted metabolic analysis, and the levels of inflammatory factors were assessed by antibody microarray. Results The volunteers showed significantly increased sleepiness and decreased vigilance during SD, and the changes in circadian rhythm and plasma biochemistry were observed. The plasma calcium (p = 0.0007) was induced by SD, while ischaemia-modified albumin (IMA, p = 0.0030) and total bile acid (TBA, p = 0.0157) decreased. Differentially expressed circRNAs in plasma were identified, which are involved in multiple signaling pathways including neuronal regulation and immunity. Accordingly, SD induced a decrease in 3-hydroxybutyric acid (3OBH, p = 0.0002) and an increase in thyroxine (T4, p < 0.0001) in plasma. The plasma anti-inflammatory cytokine IL-10 was downregulated while other ten inflammatory factors were upregulated. Conclusion This study demonstrates that SD influences biochemical, physiological, cognitive variables, and the significantly changed variables may serve as candidates of SD markers. These findings may further our understanding of the detrimental consequence of sleep disturbance at multiple levels.
Collapse
Affiliation(s)
- Shiqi Liu
- School of Life Sciences, Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Xiaohong Ma
- School of Life Sciences, Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Ying Chen
- Engineering Research Center of Human Circadian Rhythm and Sleep, Space Science and Technology Institute, Shenzhen, China
| | - Yuanyuan Zhao
- School of Life Sciences, Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Rujia Luo
- School of Life Sciences, Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Zhouying Wu
- School of Life Sciences, Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Yicheng Li
- School of Life Sciences, Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Yongyu Qian
- School of Life Sciences, Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Wenwen Wang
- School of Life Sciences, Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Shuohan Dong
- School of Life Sciences, Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Zengxuan Zhou
- School of Life Sciences, Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Silin Li
- School of Life Sciences, Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Yi Xiao
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Xinhai Zhu
- Sun Yat-sen University Instrumental Analysis & Research Center, Sun Yat-sen University, Guangzhou, China
| | - Yu Tian
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Jinhu Guo
- School of Life Sciences, Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
16
|
Bjørkum AA, Griebel L, Birkeland E. Human serum proteomics reveals a molecular signature after one night of sleep deprivation. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2024; 5:zpae042. [PMID: 39131770 PMCID: PMC11310596 DOI: 10.1093/sleepadvances/zpae042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 05/31/2024] [Indexed: 08/13/2024]
Abstract
Study Objectives Sleep deprivation is highly prevalent and caused by conditions such as night shift work or illnesses like obstructive sleep apnea. Compromised sleep affects cardiovascular-, immune-, and neuronal systems. Recently, we published human serum proteome changes after a simulated night shift. This pilot proteomic study aimed to further explore changes in human blood serum after 6 hours of sleep deprivation at night. Methods Human blood serum samples from eight self-declared healthy females were analyzed using Orbitrap Eclipse mass spectrometry (MS-MS) and high-pressure liquid chromatography. We used a within-participant design, in which the samples were taken after 6 hours of sleep at night and after 6 hours of sleep deprivation the following night. Systems biological databases and bioinformatic software were used to analyze the data and comparative analysis were done with other published sleep-related proteomic datasets. Results Out of 494 proteins, 66 were found to be differentially expressed proteins (DEPs) after 6 hours of sleep deprivation. Functional enrichment analysis revealed the associations of these DEPs with several biological functions related to the altered regulation of cellular processes such as platelet degranulation and blood coagulation, as well as associations with different curated gene sets. Conclusions This study presents serum proteomic changes after 6 hours of sleep deprivation, supports previous findings showing that short sleep deprivation affects several biological processes, and reveals a molecular signature of proteins related to pathological conditions such as altered coagulation and platelet function, impaired lipid and immune function, and cell proliferation. Data are available via ProteomeXchange with identifier PXD045729. This paper is part of the Genetic and other molecular underpinnings of sleep, sleep disorders, and circadian rhythms including translational approaches Collection.
Collapse
Affiliation(s)
- Alvhild Alette Bjørkum
- Department of Safety, Chemistry and Biomedical Laboratory Sciences, Western Norway University of Applied Sciences, Bergen, Norway
| | - Leandra Griebel
- Department of Safety, Chemistry and Biomedical Laboratory Sciences, Western Norway University of Applied Sciences, Bergen, Norway
- Department of Health Sciences and Technology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Even Birkeland
- The Proteomics Unit at The Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
17
|
Vakili O, Adibi Sedeh P, Pourfarzam M. Metabolic biomarkers in irritable bowel syndrome diagnosis. Clin Chim Acta 2024; 560:119753. [PMID: 38821336 DOI: 10.1016/j.cca.2024.119753] [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: 04/14/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Irritable bowel syndrome (IBS) is a chronic gastrointestinal (GI) disorder characterized by altered bowel habits and abdominal discomfort during defecation. It significantly impacts life quality and work productivity for those affected. Global data suggests a slightly higher prevalence in females than in males. Today, unambiguous diagnosis of IBS remains challenging due to the absence of a specific biochemical, histopathological, or radiological test. Current diagnosis relies heavily on thorough symptom evaluation. Efforts by the Rome committees have established standardized diagnostic criteria (Rome I-IV), improving consistency and clinical applicability. Recent studies in this framework, seem to have successfully employed metabolomics techniques to identify distinct metabolite profiles in breath and stool samples of IBS patients, differentiating them from healthy controls and those with other functional GI disorders, such as inflammatory bowel disease (IBD). Building on this success, researchers are investigating the presence of similar metabolites in easily accessible biofluids such as urine, potentially offering a less invasive diagnostic approach. Accordingly, this review focuses on key metabolites specifically detected in IBS patients' biological specimens, with a focus on urinary metabolites, using various methods, particularly mass spectrometry (MS)-based techniques, including gas chromatography-MS (GC-MS), liquid chromatography-tandem MS (LC-MS/MS), and capillary electrophoresis-MS (CE-MS) metabolomics assays. These findings may make provision for a new set of non-invasive biomarkers for IBS diagnosis and management.
Collapse
Affiliation(s)
- Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Peyman Adibi Sedeh
- Gastroenterology and Hepatology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Morteza Pourfarzam
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
18
|
Yan H, Li G, Zhang X, Zhang C, Li M, Qiu Y, Sun W, Dong Y, Li S, Li J. Targeted metabolomics-based understanding of the sleep disturbances in drug-naïve patients with schizophrenia. BMC Psychiatry 2024; 24:355. [PMID: 38741058 DOI: 10.1186/s12888-024-05805-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/30/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Sleep disturbances are a common occurrence in patients with schizophrenia, yet the underlying pathogenesis remain poorly understood. Here, we performed a targeted metabolomics-based approach to explore the potential biological mechanisms contributing to sleep disturbances in schizophrenia. METHODS Plasma samples from 59 drug-naïve patients with schizophrenia and 36 healthy controls were subjected to liquid chromatography-mass spectrometry (LC-MS) targeted metabolomics analysis, allowing for the quantification and profiling of 271 metabolites. Sleep quality and clinical symptoms were assessed using the Pittsburgh Sleep Quality Index (PSQI) and the Positive and Negative Symptom Scale (PANSS), respectively. Partial correlation analysis and orthogonal partial least squares discriminant analysis (OPLS-DA) model were used to identify metabolites specifically associated with sleep disturbances in drug-naïve schizophrenia. RESULTS 16 characteristic metabolites were observed significantly associated with sleep disturbances in drug-naïve patients with schizophrenia. Furthermore, the glycerophospholipid metabolism (Impact: 0.138, p<0.001), the butanoate metabolism (Impact: 0.032, p=0.008), and the sphingolipid metabolism (Impact: 0.270, p=0.104) were identified as metabolic pathways associated with sleep disturbances in drug-naïve patients with schizophrenia. CONCLUSIONS Our study identified 16 characteristic metabolites (mainly lipids) and 3 metabolic pathways related to sleep disturbances in drug-naïve schizophrenia. The detection of these distinct metabolites provide valuable insights into the underlying biological mechanisms associated with sleep disturbances in schizophrenia.
Collapse
Affiliation(s)
- Huiming Yan
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
| | - Gang Li
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
- Chifeng Anding Hospital, NO.18 Gongger Street, Hongshan District, Chifeng City, 024000, Inner Mongolia Autonomous Region, China
| | - Xue Zhang
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
- Chifeng Anding Hospital, NO.18 Gongger Street, Hongshan District, Chifeng City, 024000, Inner Mongolia Autonomous Region, China
| | - Chuhao Zhang
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
| | - Meijuan Li
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
| | - Yuying Qiu
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
| | - Wei Sun
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
| | - Yeqing Dong
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
| | - Shen Li
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China.
| | - Jie Li
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China.
| |
Collapse
|
19
|
Peng N, Li S. Short or long sleep duration was associated with chronic kidney disease in a Chinese nationwide cohort study. Int Urol Nephrol 2024; 56:1695-1701. [PMID: 37934349 DOI: 10.1007/s11255-023-03861-1] [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: 07/24/2023] [Accepted: 10/17/2023] [Indexed: 11/08/2023]
Abstract
OBJECTIVE Sleep duration is an important factor influencing health outcomes. The association between sleep duration and kidney function remains elusive. This study aimed to explore the association between sleep duration and chronic kidney disease (CKD) amongst Chinese adults. METHODS We conducted a cross-sectional study based on the China Health and Nutrition Survey (CHNS) in the wave of 2009. Participants were divided into three groups: ≤ 6 h/day (short sleepers), 7-8 h/day (regular sleepers) and ≥ 9 h/day (long sleepers) according to self-reported sleep duration. CKD was defined as estimated glomerular filtration rate < 60 mL/min/1.73 m2. RESULTS A total of 8096 Chinese adults (45.9% men) with a mean age of 50.6 years were included in the study. Compared with regular sleepers, both short and long regular sleepers had a higher prevalence of CKD. A U-shaped relationship between sleep duration and CKD was displayed by restricted cubic spline curve (P-overall < 0.001, P-nonlinear < 0.001). Multivariate logistic regression models revealed that both short and long sleep duration were clinically associated with higher odds of CKD, after adjustments for covariates [adjusted odds ratio (OR) 1.25 and 1.30; 95% confidence interval (CI) 1.00-1.56 and 1.08-1.54, for short and long sleep duration, respectively]. In subgroup analyses, we found the association was still observed in participants without hypertension or diabetes mellitus. CONCLUSION Short or long sleep duration was associated with CKD in the general population.
Collapse
Affiliation(s)
- Naling Peng
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Shizhen Li
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
20
|
Basu R, Preat T, Plaçais PY. Glial metabolism versatility regulates mushroom body-driven behavioral output in Drosophila. Learn Mem 2024; 31:a053823. [PMID: 38862167 PMCID: PMC11199944 DOI: 10.1101/lm.053823.123] [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: 03/22/2024] [Accepted: 04/23/2024] [Indexed: 06/13/2024]
Abstract
Providing metabolic support to neurons is now recognized as a major function of glial cells that is conserved from invertebrates to vertebrates. However, research in this field has focused for more than two decades on the relevance of lactate and glial glycolysis for neuronal energy metabolism, while overlooking many other facets of glial metabolism and their impact on neuronal physiology, circuit activity, and behavior. Here, we review recent work that has unveiled new features of glial metabolism, especially in Drosophila, in the modulation of behavioral traits involving the mushroom bodies (MBs). These recent findings reveal that spatially and biochemically distinct modes of glucose-derived neuronal fueling are implemented within the MB in a memory type-specific manner. In addition, cortex glia are endowed with several antioxidant functions, whereas astrocytes can serve as pro-oxidant agents that are beneficial to redox signaling underlying long-term memory. Finally, glial fatty acid oxidation seems to play a dual fail-safe role: first, as a mode of energy production upon glucose shortage, and, second, as a factor underlying the clearance of excessive oxidative load during sleep. Altogether, these integrated studies performed in Drosophila indicate that glial metabolism has a deterministic role on behavior.
Collapse
Affiliation(s)
- Ruchira Basu
- Energy & Memory, Brain Plasticity (UMR 8249), CNRS, ESPCI Paris, PSL Research University, 75005 Paris, France
| | - Thomas Preat
- Energy & Memory, Brain Plasticity (UMR 8249), CNRS, ESPCI Paris, PSL Research University, 75005 Paris, France
| | - Pierre-Yves Plaçais
- Energy & Memory, Brain Plasticity (UMR 8249), CNRS, ESPCI Paris, PSL Research University, 75005 Paris, France
| |
Collapse
|
21
|
Hu B, Shen W, Wang Y, Wu Q, Li J, Xu X, Han Y, Xiao L, Yin D. Prevalence and related factors of sleep quality among Chinese undergraduates in Jiangsu Province: multiple models' analysis. Front Psychol 2024; 15:1343186. [PMID: 38659684 PMCID: PMC11040509 DOI: 10.3389/fpsyg.2024.1343186] [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: 11/23/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Background and aims In China, a significant number of undergraduates are experiencing poor sleep quality. This study was designed to investigate the prevalence of poor sleep quality and identify associated factors among undergraduates in Jiangsu Province, China. Methods A total of 8,457 participants were collected in 2022 using whole-group convenience sampling. The factors studied included basic demographics, family and social support, personal lifestyles, physical and mental health, mobile phone addiction index (MPAI), and the Connor-Davidson resilience scale (CD-RISC). The Pittsburgh Sleep Quality Index (PSQI) was used to assess sleep quality. Four models, including weighted multiple linear regression, binary logistic regression, weighted linear mixed model, and logistic regression with random effects, were applied to identify associated factors for sleep quality. Results Of the 8,457 participants analyzed, 26.64% (2,253) were classified into the poor sleep quality group with a PSQI score >7. No significant relationship was found between sleep quality and gender, native place, economic level of family, physical exercise, dormitory light, dormitory hygiene, and amativeness matter. Risk factors for sleep quality identified by the four models included lower CD-RISC, higher MPAI, fourth grade or above, smoking, drinking, greater academic pressure, greater employment pressure, roommate sleeping late, noisy dormitory, poorer physical health status, poorer mental health status, and psychological counseling. Conclusions These findings provide valuable insights for university administrators, enabling them to better understand the risk factors associated with poor sleep quality in undergraduates. By identifying these factors, administrators can provide targeted intervention measures and counseling programs to improve students' sleep quality.
Collapse
Affiliation(s)
- Bin Hu
- Key Laboratory of Human Genetics and Environmental Medicine, School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Wen Shen
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, Jiangsu, China
| | - Yun Wang
- Department of Dermatology, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, Jiangsu, China
| | - Qi Wu
- Key Laboratory of Human Genetics and Environmental Medicine, School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Jiali Li
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Xiaozhou Xu
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Yaohui Han
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Lishun Xiao
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, China
- Center for Medical Statistics and Data Analysis, Xuzhou Medical University, Xuzhou, China
| | - Dehui Yin
- Key Laboratory of Human Genetics and Environmental Medicine, School of Public Health, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
22
|
Zhang N, Gao X, Li D, Xu L, Zhou G, Xu M, Peng L, Sun G, Pan F, Li Y, Ren R, Huang R, Yang Y, Wang Z. Sleep deprivation-induced anxiety-like behaviors are associated with alterations in the gut microbiota and metabolites. Microbiol Spectr 2024; 12:e0143723. [PMID: 38421192 PMCID: PMC10986621 DOI: 10.1128/spectrum.01437-23] [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: 04/07/2023] [Accepted: 02/03/2024] [Indexed: 03/02/2024] Open
Abstract
The present study aimed to characterize the gut microbiota and serum metabolome changes associated with sleep deprivation (SD) as well as to explore the potential benefits of multi-probiotic supplementation in alleviating SD-related mental health disorders. Rats were subjected to 7 days of SD, followed by 14 days of multi-probiotics or saline administration. Open-field tests were conducted at baseline, end of SD (day 7), and after 14 days of saline or multi-probiotic gavage (day 21). Metagenomic sequencing was conducted on fecal samples, and serum metabolites were measured by untargeted liquid chromatography tandem-mass spectrometry. At day 7, anxiety-like behaviors, including significant decreases in total movement distance (P = 0.0002) and staying time in the central zone (P = 0.021), were observed. In addition, increased levels of lipopolysaccharide (LPS; P = 0.028) and decreased levels of uridine (P = 0.018) and tryptophan (P = 0.01) were detected in rats after 7 days of SD. After SD, the richness of the gut bacterial community increased, and the levels of Akkermansia muciniphila, Muribaculum intestinale, and Bacteroides caecimuris decreased. The changes in the host metabolism and gut microbiota composition were strongly associated with the anxiety-like behaviors caused by SD. In addition, multi-probiotic supplementation for 14 days modestly improved the anxiety-like behaviors in SD rats but significantly reduced the serum level of LPS (P = 0.045). In conclusion, SD induces changes in the gut microbiota and serum metabolites, which may contribute to the development of chronic inflammatory responses and affect the gut-brain axis, causing anxiety-like behaviors. Probiotic supplementation significantly reduces serum LPS, which may alleviate the influence of chronic inflammation. IMPORTANCE The disturbance in the gut microbiome and serum metabolome induced by SD may be involved in anxiety-like behaviors. Probiotic supplementation decreases serum levels of LPS, but this reduction may be insufficient for alleviating SD-induced anxiety-like behaviors.
Collapse
Affiliation(s)
- Nana Zhang
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Xuefeng Gao
- Shenzhen Key Laboratory of Gastrointestinal Microbiota and Disease, Integrative Microecology Clinical Center, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Clinical Innovation & Research Center, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Donghao Li
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Lijuan Xu
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Guanzhou Zhou
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Mengqi Xu
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Lihua Peng
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Gang Sun
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Fei Pan
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Yan Li
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Rongrong Ren
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Ruolan Huang
- Department of Neurology, Shenzhen University Clinical Research Center for Neurological Diseases, Shenzhen University General Hospital, Shenzhen, China
| | - Yunsheng Yang
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| | - Zikai Wang
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology and Hepatology, The First Centre of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
23
|
Archer SN, Möller-Levet C, Bonmatí-Carrión MÁ, Laing EE, Dijk DJ. Extensive dynamic changes in the human transcriptome and its circadian organization during prolonged bed rest. iScience 2024; 27:109331. [PMID: 38487016 PMCID: PMC10937834 DOI: 10.1016/j.isci.2024.109331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/11/2023] [Accepted: 02/20/2024] [Indexed: 03/17/2024] Open
Abstract
Physiological and molecular processes including the transcriptome change across the 24-h day, driven by molecular circadian clocks and behavioral and systemic factors. It is not known how the temporal organization of the human transcriptome responds to a long-lasting challenge. This may, however, provide insights into adaptation, disease, and recovery. We investigated the human 24-h time series transcriptome in 20 individuals during a 90-day constant bed rest protocol. We show that the protocol affected 91% of the transcriptome with 76% of the transcriptome still affected after 10 days of recovery. Dimensionality-reduction approaches revealed that many affected transcripts were associated with mRNA translation and immune function. The number, amplitude, and phase of rhythmic transcripts, including clock genes, varied significantly across the challenge. These findings of long-lasting changes in the temporal organization of the transcriptome have implications for understanding the mechanisms underlying health consequences of conditions such as microgravity and bed rest.
Collapse
Affiliation(s)
- Simon N. Archer
- Surrey Sleep Research Centre, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
| | - Carla Möller-Levet
- Bioinformatics Core Facility, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
| | - María-Ángeles Bonmatí-Carrión
- Surrey Sleep Research Centre, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
- Chronobiology Laboratory, Department of Physiology, University of Murcia, Murcia, Spain
- Ciber Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Emma E. Laing
- Department of Microbiology, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
| | - Derk-Jan Dijk
- Surrey Sleep Research Centre, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
- UK Dementia Research Institute Care Research & Technology Centre, Imperial College London & University of Surrey, Guildford, UK
| |
Collapse
|
24
|
Jeppe K, Ftouni S, Nijagal B, Grant LK, Lockley SW, Rajaratnam SMW, Phillips AJK, McConville MJ, Tull D, Anderson C. Accurate detection of acute sleep deprivation using a metabolomic biomarker-A machine learning approach. SCIENCE ADVANCES 2024; 10:eadj6834. [PMID: 38457492 PMCID: PMC11094653 DOI: 10.1126/sciadv.adj6834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 02/02/2024] [Indexed: 03/10/2024]
Abstract
Sleep deprivation enhances risk for serious injury and fatality on the roads and in workplaces. To facilitate future management of these risks through advanced detection, we developed and validated a metabolomic biomarker of sleep deprivation in healthy, young participants, across three experiments. Bi-hourly plasma samples from 2 × 40-hour extended wake protocols (for train/test models) and 1 × 40-hour protocol with an 8-hour overnight sleep interval were analyzed by untargeted liquid chromatography-mass spectrometry. Using a knowledge-based machine learning approach, five consistently important variables were used to build predictive models. Sleep deprivation (24 to 38 hours awake) was predicted accurately in classification models [versus well-rested (0 to 16 hours)] (accuracy = 94.7%/AUC 99.2%, 79.3%/AUC 89.1%) and to a lesser extent in regression (R2 = 86.1 and 47.8%) models for within- and between-participant models, respectively. Metabolites were identified for replicability/future deployment. This approach for detecting acute sleep deprivation offers potential to reduce accidents through "fitness for duty" or "post-accident analysis" assessments.
Collapse
Affiliation(s)
- Katherine Jeppe
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
- Cooperative Research Centre for Alertness, Safety and Productivity, Melbourne, Australia
| | - Suzanne Ftouni
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
- Cooperative Research Centre for Alertness, Safety and Productivity, Melbourne, Australia
| | - Brunda Nijagal
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, Parkville, Australia
| | - Leilah K. Grant
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
- Cooperative Research Centre for Alertness, Safety and Productivity, Melbourne, Australia
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Steven W. Lockley
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
- Cooperative Research Centre for Alertness, Safety and Productivity, Melbourne, Australia
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Shantha M. W. Rajaratnam
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
- Cooperative Research Centre for Alertness, Safety and Productivity, Melbourne, Australia
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Andrew J. K. Phillips
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
| | - Malcolm J. McConville
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, Parkville, Australia
| | - Dedreia Tull
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, Parkville, Australia
| | - Clare Anderson
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
- Cooperative Research Centre for Alertness, Safety and Productivity, Melbourne, Australia
- Centre for Human Brain Health, School of Psychology, University of Birmingham, Edgbaston, UK
| |
Collapse
|
25
|
Bonomo R, Canta A, Chiorazzi A, Carozzi VA, Meregalli C, Pozzi E, Alberti P, Frampas CF, Van der Veen DR, Marmiroli P, Skene DJ, Cavaletti G. Effect of age on metabolomic changes in a model of paclitaxel-induced peripheral neurotoxicity. J Peripher Nerv Syst 2024; 29:58-71. [PMID: 38126610 DOI: 10.1111/jns.12609] [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: 09/15/2023] [Revised: 11/22/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND AND AIMS Chemotherapy-induced peripheral neurotoxicity (CIPN) is one of the most common dose-limiting side effects of paclitaxel (PTX) treatment. Many age-related changes have been hypothesized to underlie susceptibility to damage or impaired regeneration/repair after nerve injury. The results of these studies, however, are inconclusive and other potential biomarkers of nerve impairment need to be investigated. METHODS Twenty-four young (2 months) and 24 adult (9 months) Wistar male rats were randomized to either PTX treatment (10 mg/kg i.v. once/week for 4 weeks) or vehicle administration. Neurophysiological and behavioral tests were performed at baseline, after 4 weeks of treatment and 2-week follow-up. Skin biopsies and nerve specimens collected from sacrificed animals were examined for intraepidermal nerve fiber (IENF) density assessment and nerve morphology/morphometry. Blood and liver samples were collected for targeted metabolomics analysis. RESULTS At the end of treatment, the neurophysiological studies revealed a reduction in sensory nerve action potential amplitude (p < .05) in the caudal nerve of young PTX-animals, and in both the digital and caudal nerve of adult PTX-animals (p < .05). A significant decrease in the mechanical threshold was observed only in young PTX-animals (p < .001), but not in adult PTX-ones. Nevertheless, both young and adult PTX-rats had reduced IENF density (p < .0001), which persisted at the end of follow-up period. Targeted metabolomics analysis showed significant differences in the plasma metabolite profiles between PTX-animals developing peripheral neuropathy and age-matched controls, with triglycerides, diglycerides, acylcarnitines, carnosine, long chain ceramides, sphingolipids, and bile acids playing a major role in the response to PTX administration. INTERPRETATION Our study identifies for the first time multiple related metabolic axes involved in PTX-induced peripheral neurotoxicity, and suggests age-related differences in CIPN manifestations and in the metabolic profile.
Collapse
Affiliation(s)
- Roberta Bonomo
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
- School of Medicine and Surgery, Kore University of Enna, Enna, Italy
| | - Annalisa Canta
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Alessia Chiorazzi
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Valentina Alda Carozzi
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Cristina Meregalli
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Eleonora Pozzi
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Department of Neurology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Cecile F Frampas
- Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Daan R Van der Veen
- Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Paola Marmiroli
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Debra J Skene
- Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Guido Cavaletti
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Department of Neurology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
26
|
Zhang Y, Yu B, Qi Q, Azarbarzin A, Chen H, Shah NA, Ramos AR, Zee PC, Cai J, Daviglus ML, Boerwinkle E, Kaplan R, Liu PY, Redline S, Sofer T. Metabolomic profiles of sleep-disordered breathing are associated with hypertension and diabetes mellitus development. Nat Commun 2024; 15:1845. [PMID: 38418471 PMCID: PMC10902315 DOI: 10.1038/s41467-024-46019-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 02/12/2024] [Indexed: 03/01/2024] Open
Abstract
Sleep-disordered breathing (SDB) is a prevalent disorder characterized by recurrent episodic upper airway obstruction. Using data from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL), we apply principal component analysis (PCA) to seven SDB-related measures. We estimate the associations of the top two SDB PCs with serum levels of 617 metabolites, in both single-metabolite analysis, and a joint penalized regression analysis. The discovery analysis includes 3299 individuals, with validation in a separate dataset of 1522 individuals. Five metabolite associations with SDB PCs are discovered and replicated. SDB PC1, characterized by frequent respiratory events common in older and male adults, is associated with pregnanolone and progesterone-related sulfated metabolites. SDB PC2, characterized by short respiratory event length and self-reported restless sleep, enriched in young adults, is associated with sphingomyelins. Metabolite risk scores (MRSs), representing metabolite signatures associated with the two SDB PCs, are associated with 6-year incident hypertension and diabetes. These MRSs have the potential to serve as biomarkers for SDB, guiding risk stratification and treatment decisions.
Collapse
Affiliation(s)
- Ying Zhang
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Bing Yu
- Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Ali Azarbarzin
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham & Women's Hospital & Harvard Medical School, Boston, MA, 02115, USA
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Neomi A Shah
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alberto R Ramos
- Sleep Medicine Program, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Phyllis C Zee
- Division of Sleep Medicine, Department of Neurology, Northwestern University, Chicago, IL, 60611, USA
| | - Jianwen Cai
- Collaborative Studies Coordinating Center, Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Martha L Daviglus
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60612, USA
| | - Eric Boerwinkle
- Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, NY, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Peter Y Liu
- The Institute for Translational Genomics and Population Sciences, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham & Women's Hospital & Harvard Medical School, Boston, MA, 02115, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham & Women's Hospital & Harvard Medical School, Boston, MA, 02115, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, 02115, USA.
| |
Collapse
|
27
|
Moreno-Cortés ML, Meza-Alvarado JE, García-Mena J, Hernández-Rodríguez A. Chronodisruption and Gut Microbiota: Triggering Glycemic Imbalance in People with Type 2 Diabetes. Nutrients 2024; 16:616. [PMID: 38474745 DOI: 10.3390/nu16050616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/27/2024] [Accepted: 01/28/2024] [Indexed: 03/14/2024] Open
Abstract
The desynchronization of physiological and behavioral mechanisms influences the gut microbiota and eating behavior in mammals, as shown in both rodents and humans, leading to the development of pathologies such as Type 2 diabetes (T2D), obesity, and metabolic syndrome. Recent studies propose resynchronization as a key input controlling metabolic cycles and contributing to reducing the risk of suffering some chronic diseases such as diabetes, obesity, or metabolic syndrome. In this analytical review, we present an overview of how desynchronization and its implications for the gut microbiome make people vulnerable to intestinal dysbiosis and consequent chronic diseases. In particular, we explore the eubiosis-dysbiosis phenomenon and, finally, propose some topics aimed at addressing chronotherapy as a key strategy in the prevention of chronic diseases.
Collapse
Affiliation(s)
- María Luisa Moreno-Cortés
- Laboratorio de Biomedicina, Instituto de Investigaciones Biológicas, Universidad Veracruzana, Xalapa 91190, Veracruz, Mexico
| | | | - Jaime García-Mena
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, CDMX 07360, Mexico
| | - Azucena Hernández-Rodríguez
- Laboratorio de Biomedicina, Instituto de Investigaciones Biológicas, Universidad Veracruzana, Xalapa 91190, Veracruz, Mexico
- Facultad de Bioanálisis, Universidad Veracruzana, Xalapa 91010, Veracruz, Mexico
| |
Collapse
|
28
|
Kazemian N, Zhou T, Chalasani N, Narayan A, Cedeño Laurent JG, Olvera Alvarez HA, Pakpour S. Long-Term Impact of Childhood Adversity on the Gut Microbiome of Nursing Students. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:68. [PMID: 38248533 PMCID: PMC10815413 DOI: 10.3390/ijerph21010068] [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: 11/16/2023] [Revised: 12/29/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024]
Abstract
Adverse childhood experiences (ACEs) encompass negative, stressful, and potentially traumatic events during childhood, impacting physical and mental health outcomes in adulthood. Limited studies suggest ACEs can have short-term effects on children's gut microbiomes and adult cognitive performance under stress. Nevertheless, the long-term effects of ACEs experienced during adulthood remain unexplored. Thus, this study aimed to assess the long-term effects of ACEs on the gut microbiota of adult nursing students. We employed a multidimensional approach, combining 16S rRNA sequencing, bioinformatics tools, and machine learning to predict functional capabilities. High-ACE individuals had an increased abundance of Butyricimonas spp. and Prevotella spp. and decreased levels of Clostridiales, and Lachnospira spp. Prevotella abundance correlated negatively with L-glutamate and L-glutamine biosynthesis, potentially impacting intestinal tissue integrity. While nursing students with high ACE reported increased depression, evidence for a direct gut microbiota-depression relationship was inconclusive. High-ACE individuals also experienced a higher prevalence of diarrhea. These findings highlight the long-lasting impact of ACEs on the gut microbiota and its functions in adulthood, particularly among nursing students. Further research is warranted to develop targeted interventions and strategies for healthcare professionals, optimizing overall health outcomes.
Collapse
Affiliation(s)
- Negin Kazemian
- School of Engineering, University of British Columbia, Kelowna, BC V1V 1V7, Canada;
| | - Tony Zhou
- Department of Computer Science, University of British Columbia, Kelowna, BC V1V 1V7, Canada; (T.Z.); (N.C.); (A.N.)
| | - Naveen Chalasani
- Department of Computer Science, University of British Columbia, Kelowna, BC V1V 1V7, Canada; (T.Z.); (N.C.); (A.N.)
| | - Apurva Narayan
- Department of Computer Science, University of British Columbia, Kelowna, BC V1V 1V7, Canada; (T.Z.); (N.C.); (A.N.)
- Department of Computer Science, University of Western Ontario, 1151 Richmond St., London, ON N6A 3K7, Canada
- Department of Electrical and Computer Engineering, University of Western Ontario, 1151 Richmond St., London, ON N6A 3K7, Canada
| | - Jose Guillermo Cedeño Laurent
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
- Department of Environmental and Occupational Health and Justice, Rutgers School of Public Health, Piscataway, NJ 08854, USA
| | | | - Sepideh Pakpour
- School of Engineering, University of British Columbia, Kelowna, BC V1V 1V7, Canada;
| |
Collapse
|
29
|
Sinturel F, Chera S, Brulhart-Meynet MC, Montoya JP, Stenvers DJ, Bisschop PH, Kalsbeek A, Guessous I, Jornayvaz FR, Philippe J, Brown SA, D'Angelo G, Riezman H, Dibner C. Circadian organization of lipid landscape is perturbed in type 2 diabetic patients. Cell Rep Med 2023; 4:101299. [PMID: 38016481 PMCID: PMC10772323 DOI: 10.1016/j.xcrm.2023.101299] [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: 12/20/2022] [Revised: 06/26/2023] [Accepted: 10/30/2023] [Indexed: 11/30/2023]
Abstract
Lipid homeostasis in humans follows a diurnal pattern in muscle and pancreatic islets, altered upon metabolic dysregulation. We employ tandem and liquid-chromatography mass spectrometry to investigate daily regulation of lipid metabolism in subcutaneous white adipose tissue (SAT) and serum of type 2 diabetic (T2D) and non-diabetic (ND) human volunteers (n = 12). Around 8% of ≈440 lipid metabolites exhibit diurnal rhythmicity in serum and SAT from ND and T2D subjects. The spectrum of rhythmic lipids differs between ND and T2D individuals, with the most substantial changes observed early morning, as confirmed by lipidomics in an independent cohort of ND and T2D subjects (n = 32) conducted at a single morning time point. Strikingly, metabolites identified as daily rhythmic in both serum and SAT from T2D subjects exhibit phase differences. Our study reveals massive temporal and tissue-specific alterations of human lipid homeostasis in T2D, providing essential clues for the development of lipid biomarkers in a temporal manner.
Collapse
Affiliation(s)
- Flore Sinturel
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), 1211 Geneva, Switzerland
| | - Simona Chera
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), 1211 Geneva, Switzerland; Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Marie-Claude Brulhart-Meynet
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Jonathan Paz Montoya
- Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Dirk Jan Stenvers
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, Amsterdam, 1105 AZ, the Netherlands
| | - Peter H Bisschop
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, Amsterdam, 1105 AZ, the Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, Amsterdam, 1105 AZ, the Netherlands; Laboratory for Endocrinology, Department of Clinical Chemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Arts and Sciences (KNAW), Amsterdam, 1105 BA, the Netherlands
| | - Idris Guessous
- Department and Division of Primary Care Medicine, University Hospitals of Geneva, 1211 Geneva, Switzerland
| | - François R Jornayvaz
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Division of Endocrinology, Diabetes, Nutrition, and Therapeutic Patient Education, Department of Medicine, University Hospitals of Geneva, 1211 Geneva, Switzerland
| | - Jacques Philippe
- Division of Endocrinology, Diabetes, Nutrition, and Therapeutic Patient Education, Department of Medicine, University Hospitals of Geneva, 1211 Geneva, Switzerland
| | - Steven A Brown
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland
| | - Giovanni D'Angelo
- Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Howard Riezman
- Department of Biochemistry, Faculty of Science, NCCR Chemical Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Charna Dibner
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), 1211 Geneva, Switzerland.
| |
Collapse
|
30
|
Sang D, Lin K, Yang Y, Ran G, Li B, Chen C, Li Q, Ma Y, Lu L, Cui XY, Liu Z, Lv SQ, Luo M, Liu Q, Li Y, Zhang EE. Prolonged sleep deprivation induces a cytokine-storm-like syndrome in mammals. Cell 2023; 186:5500-5516.e21. [PMID: 38016470 DOI: 10.1016/j.cell.2023.10.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 08/17/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023]
Abstract
Most animals require sleep, and sleep loss induces serious pathophysiological consequences, including death. Previous experimental approaches for investigating sleep impacts in mice have been unable to persistently deprive animals of both rapid eye movement sleep (REMS) and non-rapid eye movement sleep (NREMS). Here, we report a "curling prevention by water" paradigm wherein mice remain awake 96% of the time. After 4 days of exposure, mice exhibit severe inflammation, and approximately 80% die. Sleep deprivation increases levels of prostaglandin D2 (PGD2) in the brain, and we found that elevated PGD2 efflux across the blood-brain-barrier-mediated by ATP-binding cassette subfamily C4 transporter-induces both accumulation of circulating neutrophils and a cytokine-storm-like syndrome. Experimental disruption of the PGD2/DP1 axis dramatically reduced sleep-deprivation-induced inflammation. Thus, our study reveals that sleep-related changes in PGD2 in the central nervous system drive profound pathological consequences in the peripheral immune system.
Collapse
Affiliation(s)
- Di Sang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China; National Institute of Biological Sciences, Beijing, China
| | - Keteng Lin
- National Institute of Biological Sciences, Beijing, China; College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yini Yang
- Peking University School of Life Sciences, Beijing, China
| | - Guangdi Ran
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Bohan Li
- Peking-Tsinghua Center for Life Sciences, Beijing, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Chen Chen
- National Institute of Biological Sciences, Beijing, China
| | - Qi Li
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Yan Ma
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Lihui Lu
- National Institute of Biological Sciences, Beijing, China
| | - Xi-Yang Cui
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Zhibo Liu
- Peking-Tsinghua Center for Life Sciences, Beijing, China; Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Sheng-Qing Lv
- Department of Neurosurgery, Xinqiao Hospital, Chongqing, China
| | - Minmin Luo
- National Institute of Biological Sciences, Beijing, China; School of Life Sciences, Tsinghua University, Beijing, China; Chinese Institute for Brain Research, Beijing, China
| | - Qinghua Liu
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Yulong Li
- Peking University School of Life Sciences, Beijing, China; Peking-Tsinghua Center for Life Sciences, Beijing, China; State Key Laboratory of Membrane Biology, Beijing, China; PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Eric Erquan Zhang
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| |
Collapse
|
31
|
Depner CM. Biomarkers linking habitual short sleep duration with risk of cardiometabolic disease: current progress and future directions. FRONTIERS IN SLEEP 2023; 2:1293941. [PMID: 39041043 PMCID: PMC11262587 DOI: 10.3389/frsle.2023.1293941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Approximately one in three adults in the United States sleeps less than the recommended 7 h per night. Decades of epidemiological data and data from experimental sleep restriction studies demonstrate short sleep duration is associated with adverse cardiometabolic risk, including risk of type 2 diabetes and cardiovascular disease. However, the precise mechanisms underlying this risk are not fully elucidated and there is a lack of sleep-based interventions designed to mitigate such risk. One strategy to overcome these limitations is to develop biomarkers that link habitual short sleep duration with adverse cardiometabolic risk. Such biomarkers could inform biochemical mechanisms, identify new targets for interventions, support precision medicine by identifying individuals most likely to benefit from sleep-based interventions, and ultimately lead to improved cardiometabolic health in people with habitual short sleep durations. Early progress demonstrates proof-of-principle that omics-based technologies are a viable approach to create biochemical signatures (biomarkers) of short sleep duration, primarily derived from acute studies of experimental sleep restriction. Yet, much work remains. Notably, studies that translate early findings from experimental sleep restriction to free-living adults with habitual short sleep duration have high potential to advance the field. Such studies also create an exciting opportunity for larger randomized controlled trials that simultaneously identify biomarkers of habitual short sleep duration and evaluate the efficacy of sleep-based interventions. Ultimately, early progress in developing molecular biomarkers of short sleep duration combined with the prior decades of progress in the sleep and metabolism fields provide the foundation for exciting progress in the biomarker development space.
Collapse
Affiliation(s)
- Christopher M. Depner
- Department of Health and Kinesiology, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
32
|
Russell KL, Rodman HR, Pak VM. Sleep insufficiency, circadian rhythms, and metabolomics: the connection between metabolic and sleep disorders. Sleep Breath 2023; 27:2139-2153. [PMID: 37147557 DOI: 10.1007/s11325-023-02828-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/06/2023] [Accepted: 04/05/2023] [Indexed: 05/07/2023]
Abstract
PURPOSE US adults who report experiencing insufficient sleep are more likely to suffer from metabolic disorders such as hyperlipidemia, diabetes, and obesity than those with sufficient sleep. Less is understood about the underlying molecular mechanisms connecting these phenomena. A systematic, qualitative review of metabolomics studies exploring metabolic changes in response to sleep insufficiency, sleep deprivation, or circadian disruption was conducted in accordance with PRISMA guidelines. METHODS An electronic literature review in the PubMed database was performed considering publications through May 2021 and screening and eligibility criteria were applied to articles retrieved. The following keywords were used: "metabolomics" and "sleep disorders" or "sleep deprivation" or "sleep disturbance" or "circadian rhythm." After screening and addition of studies included from reference lists of retrieved studies, 16 records were identified for review. RESULTS Consistent changes in metabolites were observed across studies between individuals experiencing sleep deprivation compared to non-sleep deprived controls. Significant increases in phosphatidylcholines, acylcarnitines, sphingolipids, and other lipids were consistent across studies. Increased levels of amino acids such as tryptophan and phenylalanine were also noted. However, studies were limited to small samples of young, healthy, mostly male participants conducted in short inpatient sessions, limiting generalizability. CONCLUSION Changes in lipid and amino acid metabolites accompanying sleep deprivation and/or circadian rhythms may indicate cellular membrane and protein breakdown underlying the connection between sleep disturbance, hyperlipidemia, and other metabolic disorders. Larger epidemiological studies examining changes in the human metabolome in response to chronic insufficient sleep would help elucidate this relationship.
Collapse
Affiliation(s)
| | | | - Victoria M Pak
- Emory Nell Hodgson School of Nursing, Atlanta, GA, USA.
- Emory Rollins School of Public Health, Atlanta, GA, USA.
| |
Collapse
|
33
|
Gombert M, Reisdorph N, Morton SJ, Wright KP, Depner CM. Insufficient sleep and weekend recovery sleep: classification by a metabolomics-based machine learning ensemble. Sci Rep 2023; 13:21123. [PMID: 38036605 PMCID: PMC10689438 DOI: 10.1038/s41598-023-48208-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023] Open
Abstract
Although weekend recovery sleep is common, the physiological responses to weekend recovery sleep are not fully elucidated. Identifying molecular biomarkers that represent adequate versus insufficient sleep could help advance our understanding of weekend recovery sleep. Here, we identified potential molecular biomarkers of insufficient sleep and defined the impact of weekend recovery sleep on these biomarkers using metabolomics in a randomized controlled trial. Healthy adults (n = 34) were randomized into three groups: control (CON: 9-h sleep opportunities); sleep restriction (SR: 5-h sleep opportunities); or weekend recovery (WR: simulated workweek of 5-h sleep opportunities followed by ad libitum weekend recovery sleep and then 2 days with 5-h sleep opportunities). Blood for metabolomics was collected on the simulated Monday immediately following the weekend. Nine machine learning models, including a machine learning ensemble, were built to classify samples from SR versus CON. Notably, SR showed decreased glycerophospholipids and sphingolipids versus CON. The machine learning ensemble showed the highest G-mean performance and classified 50% of the WR samples as insufficient sleep. Our findings show insufficient sleep and recovery sleep influence the plasma metabolome and suggest more than one weekend of recovery sleep may be necessary for the identified biomarkers to return to healthy adequate sleep levels.
Collapse
Affiliation(s)
- Marie Gombert
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, 46010, Valencia, Spain
- Center for Health Sciences, SRI International, Menlo Park, CA, USA
| | - Nichole Reisdorph
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sarah J Morton
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, 1725 Pleasant Street; Clare Small 114, Boulder, CO, 80309-0354, USA
| | - Kenneth P Wright
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, 1725 Pleasant Street; Clare Small 114, Boulder, CO, 80309-0354, USA.
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - Christopher M Depner
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, 1725 Pleasant Street; Clare Small 114, Boulder, CO, 80309-0354, USA.
- Department of Health and Kinesiology, University of Utah, 250 S 1850 E; HPER North, RM 206, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
34
|
Sengupta A, Tudor JC, Cusmano D, Baur JA, Abel T, Weljie AM. Sleep deprivation and aging are metabolically linked across tissues. Sleep 2023; 46:zsad246. [PMID: 37738102 PMCID: PMC11502955 DOI: 10.1093/sleep/zsad246] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/21/2023] [Indexed: 09/24/2023] Open
Abstract
STUDY OBJECTIVES Insufficient sleep is a concerning hallmark of modern society because sleep deprivation (SD) is a risk factor for neurodegenerative and cardiometabolic disorders. SD imparts an aging-like effect on learning and memory, although little is known about possible common molecular underpinnings of SD and aging. Here, we examine this question by profiling metabolic features across different tissues after acute SD in young adult and aged mice. METHODS Young adult and aged mice were subjected to acute SD for 5 hours. Blood plasma, hippocampus, and liver samples were subjected to UPLC-MS/MS-based metabolic profiling. RESULTS SD preferentially impacts peripheral plasma and liver profiles (e.g. ketone body metabolism) whereas the hippocampus is more impacted by aging. We further demonstrate that aged animals exhibit SD-like metabolic features at baseline. Hepatic alterations include parallel changes in nicotinamide metabolism between aging and SD in young animals. Overall, metabolism in young adult animals is more impacted by SD, which in turn induces aging-like features. A set of nine metabolites was classified (79% correct) based on age and sleep status across all four groups. CONCLUSIONS Our metabolic observations demonstrate striking parallels to previous observations in studies of learning and memory and define a molecular metabolic signature of sleep loss and aging.
Collapse
Affiliation(s)
- Arjun Sengupta
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer C Tudor
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
- Current affiliation: Department of Biology, Saint Joseph’s University, Philadelphia, PA, USA
| | - Danielle Cusmano
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph A Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ted Abel
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
- Current Affiliation: Iowa Neuroscience Institute, Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 2312 PBDB, Iowa City, IA, USA
| | - Aalim M Weljie
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
35
|
Malik DM, Sengupta A, Sehgal A, Weljie AM. Altered Metabolism During the Dark Period in Drosophila Short Sleep Mutants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.30.564858. [PMID: 37961245 PMCID: PMC10634958 DOI: 10.1101/2023.10.30.564858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Sleep is an almost universally required state in biology. Disrupted sleep has been associated with adverse health risks including metabolic perturbations. Sleep is in part regulated via circadian mechanisms, however, metabolic dysfunction at different times of day arising from sleep disruption is unclear. We used targeted liquid chromatography-mass spectrometry to probe metabolic alterations using high-resolution temporal sampling of two Drosophila short sleep mutants, fumin and sleepless, across a circadian day. Discriminant analyses revealed overall distinct metabolic profiles for mutants when compared to a wild type dataset. Altered levels of metabolites involved in nicotinate/nicotinamide, alanine, aspartate, and glutamate, glyoxylate and dicarboxylate metabolism, and the TCA cycle were observed in mutants suggesting increased energetic demands. Furthermore, rhythmicity analyses revealed fewer 24 hr rhythmic metabolites in both mutants. Interestingly, mutants displayed two major peaks in phases while wild type displayed phases that were less concerted. In contrast to 24 hr rhythmic metabolites, an increase in the number of 12 hr rhythmic metabolites was observed in fumin while sleepless displayed a decrease. These results support that decreased sleep alters the overall metabolic profile with short sleep mutants displaying altered metabolite levels associated with a number of pathways in addition to altered neurotransmitter levels.
Collapse
Affiliation(s)
- Dania M. Malik
- Pharmacology Graduate Group
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
| | - Amita Sehgal
- Chronobiology and Sleep Institute
- Howard Hughes Medical Institute
| | - Aalim M. Weljie
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
- Chronobiology and Sleep Institute
| |
Collapse
|
36
|
Isherwood CM, Robertson MD, Skene DJ, Johnston JD. Daily rhythms of diabetogenic factors in men: role of type 2 diabetes and body weight. Endocr Connect 2023; 12:e230064. [PMID: 37855336 PMCID: PMC10620456 DOI: 10.1530/ec-23-0064] [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: 02/27/2023] [Accepted: 09/20/2023] [Indexed: 09/28/2023]
Abstract
Obesity is a major cause of type 2 diabetes. Transition from obesity to type 2 diabetes manifests in the dysregulation of hormones controlling glucose homeostasis and inflammation. As metabolism is a dynamic process that changes across 24 h, we assessed diurnal rhythmicity in a panel of 10 diabetes-related hormones. Plasma hormones were analysed every 2 h over 24 h in a controlled laboratory study with hourly isocaloric drinks during wake. To separate effects of body mass from type 2 diabetes, we recruited three groups of middle-aged men: an overweight (OW) group with type 2 diabetes and two control groups (lean and OW). Average daily concentrations of glucose, triacylglycerol and all the hormones except visfatin were significantly higher in the OW group compared to the lean group (P < 0.001). In type 2 diabetes, glucose, insulin, C-peptide, glucose-dependent insulinotropic peptide and glucagon-like peptide-1 increased further (P < 0.05), whereas triacylglycerol, ghrelin and plasminogen activator inhibitor-1 concentrations were significantly lower compared to the OW group (P < 0.001). Insulin, C-peptide, glucose-dependent insulinotropic peptide and leptin exhibited significant diurnal rhythms in all study groups (P < 0.05). Other hormones were only rhythmic in 1 or 2 groups. In every group, hormones associated with glucose regulation (insulin, C-peptide, glucose-dependent insulinotropic peptide, ghrelin and plasminogen activator inhibitor-1), triacylglycerol and glucose peaked in the afternoon, whereas glucagon and hormones associated with appetite and inflammation peaked at night. Thus being OW with or without type 2 diabetes significantly affected hormone concentrations but did not affect the timing of the hormonal rhythms.
Collapse
Affiliation(s)
- Cheryl M Isherwood
- Section of Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - M Denise Robertson
- Section of Metabolic Medicine, Food and Macronutrients, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Debra J Skene
- Section of Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Jonathan D Johnston
- Section of Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
37
|
Lin L, Huang Y, Wang J, Guo X, Yu F, He D, Wu C, Guo L, Wu B. CRY1/2 regulate rhythmic CYP2A5 in mouse liver through repression of E4BP4. Biochem Pharmacol 2023; 217:115843. [PMID: 37797722 DOI: 10.1016/j.bcp.2023.115843] [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: 08/11/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023]
Abstract
CYP2A5, an enzyme responsible for metabolism of diverse drugs, displays circadian rhythms in its expression and activity. However, the underlying mechanisms are not fully established. Here we aimed to investigate a potential role of CRY1/2 (circadian clock modulators) in circadian regulation of hepatic CYP2A5. Regulatory effects of CRY1/2 on CYP2A5 were determined using Cry1-null and Cry2-null mice, and validated using AML-12, Hepa1-6 and HepG2 cells. CYP2A5 activities both in vivo and in vitro were assessed using coumarin 7-hydroxylation as a probe reaction. mRNA and protein levels were detected by qPCR and western blotting, respectively. Regulatory mechanism was studied using a combination of luciferase reporter assays, chromatin immunoprecipitation (ChIP) and co-immunoprecipitation (Co-IP). We found that ablation of Cry1 or Cry2 in mice reduced hepatic CYP2A5 expression (at both mRNA and protein levels) and blunted its diurnal rhythms. Consistently, these knockouts showed decreased CYP2A5 activity (characterised by coumarin 7-hydroxylation) and a loss of its time-dependency, as well as exacerbated coumarin-induced hepatotoxicity. Cell-based assays confirmed that CRY1/2 positively regulated CYP2A5 expression and rhythms. Based on combined luciferase reporter, ChIP and Co-IP assays, we unraveled that CRY1/2 interacted with E4BP4 protein to repress its inhibitory effect on Cyp2a5 transcription and expression. In conclusion, CRY1/2 regulate rhythmic CYP2A5 in mouse liver through repression of E4BP4. These findings advance our understanding of circadian regulation of drug metabolism and pharmacokinetics.
Collapse
Affiliation(s)
- Luomin Lin
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Yuwei Huang
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinyi Wang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Xiaocao Guo
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Fangjun Yu
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Di He
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Lianxia Guo
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Baojian Wu
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
38
|
Georgopoulos D, Kondili E, Gerardy B, Alexopoulou C, Bolaki M, Younes M. Sleep Architecture Patterns in Critically Ill Patients and Survivors of Critical Illness: A Retrospective Study. Ann Am Thorac Soc 2023; 20:1624-1632. [PMID: 37413661 DOI: 10.1513/annalsats.202301-038oc] [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: 01/17/2023] [Accepted: 07/06/2023] [Indexed: 07/08/2023] Open
Abstract
Rationale: Sleep abnormalities are very frequent in critically ill patients during and after intensive care unit (ICU) stays. Their mechanisms are poorly understood. The odds ratio product (ORP) is a continuous metric (range, 0.0-2.5) of sleep depth measured in 3-second intervals and derived from the relationship of powers of different electroencephalographic frequencies to one another. When expressed as the percentage of epochs within 10 ORP deciles covering the entire ORP range, it provides information about the mechanism(s) of abnormal sleep. Objectives: To determine ORP architecture types in critically ill patients and survivors of critical illness who had previously undergone sleep studies. Methods: Nocturnal polysomnograms from 47 unsedated critically ill patients and 23 survivors of critical illness at hospital discharge were analyzed. Twelve critically ill patients were monitored also during the day, and 15 survivors underwent subsequent polysomnography 6 months after hospital discharge. In all polysomnograms, each 30-second epoch was characterized by the mean ORP of the 10 3-second epochs. The number of 30-second epochs with mean ORP within each of 10 ORP deciles covering the entire ORP range (0.0-2.5) was calculated and expressed as a percentage of total recording time. Thereafter, each polysomnogram was characterized using a two-digit ORP type, with the first digit (range, 1-3) reflecting increasing degrees of deep sleep (ORP < 0.5, deciles 1 and 2) and the second digit (range, 1-3) reflecting increasing degrees of full wakefulness (ORP > 2.25, decile 10). Results from patients were compared with those from 831 age- and gender-matched community dwellers free of sleep disorders. Results: In critically ill patients, types 1,1 and 1,2 (little deep sleep and little or average full wakefulness) dominated (46% of patients). In the community, these types are uncommon (<15%) and seen primarily in disorders that preclude progression to deep sleep (e.g., very severe obstructive sleep apnea). Next in frequency (22%) was type 1,3, consistent with hyperarousal. Day ORP sleep architecture was similar to night results. Survivors had similar patterns, with little improvement after 6 months. Conclusions: Sleep abnormalities in critically ill patients and survivors of critical illness result primarily from stimuli that preclude progression to deep sleep or from the presence of a hyperarousal state.
Collapse
Affiliation(s)
- Dimitris Georgopoulos
- Department of Intensive Care Medicine, University Hospital of Heraklion, Heraklion, Greece
| | - Eumorfia Kondili
- Department of Intensive Care Medicine, University Hospital of Heraklion, Heraklion, Greece
| | | | - Christina Alexopoulou
- Department of Intensive Care Medicine, University Hospital of Heraklion, Heraklion, Greece
| | - Maria Bolaki
- Department of Intensive Care Medicine, University Hospital of Heraklion, Heraklion, Greece
| | - Magdy Younes
- Sleep Disorders Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
39
|
Younes M. New insights and potential clinical implications of the odds ratio product. Front Neurol 2023; 14:1273623. [PMID: 37885480 PMCID: PMC10598615 DOI: 10.3389/fneur.2023.1273623] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 09/11/2023] [Indexed: 10/28/2023] Open
Abstract
The odds ratio product (ORP) is a continuous metric of sleep depth that ranges from 0 (very deep sleep) to 2. 5 (full wakefulness). Its advantage over the conventional method recommended by AASM is that it discloses different levels of stage wake (sleep propensity) and different sleep depths within the same sleep stage. As such, it can be used to identify differences in sleep depth between subjects, and in the same subjects under different circumstances, when differences are not discernible by conventional staging. It also identifies different sleep depths within stage rapid-eye-movement sleep, with possible implications to disorders during this stage. Epoch-by-epoch ORP can be displayed graphically across the night or as average values in conventional sleep stages. In addition, ORP can be reported as % of recording time in specific ORP ranges (e.g., deciles of the total ORP range) where it produces distinct distribution patterns (ORP-architecture) that have been associated with different clinical disorders and outcomes. These patterns offer unique research opportunities to identify different mechanisms and potential therapy for various sleep complaints and disorders. In this review I will discuss how ORP is measured, its validation, differences from delta power, and the various phenotypes, and their postulated mechanisms, identified by ORP architecture and the opportunities for research to advance management of sleep-disordered breathing, insomnia and idiopathic hypersomnia.
Collapse
Affiliation(s)
- Magdy Younes
- Department of Medicine, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
40
|
Ji J, Ye Y, Sheng L, Sun J, Hong Q, Liu C, Ding J, Geng S, Xu D, Zhang Y, Sun X. Sleep Promotion by 3-Hydroxy-4-Iminobutyric Acid in Walnut Diaphragma juglandis Fructus. RESEARCH (WASHINGTON, D.C.) 2023; 6:0216. [PMID: 37732131 PMCID: PMC10508226 DOI: 10.34133/research.0216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 08/08/2023] [Indexed: 09/22/2023]
Abstract
Insufficient sleep can produce a multitude of deleterious repercussions on various domains of human well-being. Concomitantly, the walnut (Juglans mandshurica) confers numerous salutary biological activities pertaining to sleep. Nevertheless, the sedative and hypnotic capacities of walnut's functional constituents remain obscure. In this investigation, we analyzed the sedative and hypnotic components of the walnut Diaphragma juglandis fructus and innovatively discovered a compound, defined as 3-hydroxy-4-iminobutyric acid (HIBA), which disrupts motor activity and enhances sleep duration by regulating the neurotransmitters (GABA, DA, etc.) within the brain and serum of mice. Subsequently, a metabolomics approach of the serum, basal ganglia, hypothalamus, and hippocampus as well as the gut microbiota was undertaken to unravel the underlying molecular mechanisms of sleep promotion. Our data reveal that HIBA can regulate the metabolism of basal ganglia (sphingolipids, acylcarnitines, etc.), possibly in relation to HIBA's influence on the gut microbiome (Muribaculum, Bacteroides, Lactobacillus, etc.). Therefore, we introduce a novel natural product, HIBA, and explicate the modulation of sleep promotion in mice based on the microbiota-gut-brain axis. This study contributes fresh insights toward natural product-based sleep research.
Collapse
Affiliation(s)
- Jian Ji
- State Key Laboratory of Food Science and Technology,
School of Food Science and Technology, National Engineering Research Center for Functional Food, Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Lihu Avenue 1800, Wuxi, Jiangsu 214100, P.R. China
- College of Food Science and Pharmacy, Xinjiang Agricultural University, No. 311 Nongda Dong Road, Ürümqi, Xinjiang, Uygur Autonomous Region 830052, P.R. China
| | - Yongli Ye
- State Key Laboratory of Food Science and Technology,
School of Food Science and Technology, National Engineering Research Center for Functional Food, Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Lihu Avenue 1800, Wuxi, Jiangsu 214100, P.R. China
| | - Lina Sheng
- State Key Laboratory of Food Science and Technology,
School of Food Science and Technology, National Engineering Research Center for Functional Food, Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Lihu Avenue 1800, Wuxi, Jiangsu 214100, P.R. China
| | - Jiadi Sun
- State Key Laboratory of Food Science and Technology,
School of Food Science and Technology, National Engineering Research Center for Functional Food, Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Lihu Avenue 1800, Wuxi, Jiangsu 214100, P.R. China
| | - Qianqian Hong
- State Key Laboratory of Food Science and Technology,
School of Food Science and Technology, National Engineering Research Center for Functional Food, Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Lihu Avenue 1800, Wuxi, Jiangsu 214100, P.R. China
| | - Chang Liu
- State Key Laboratory of Food Science and Technology,
School of Food Science and Technology, National Engineering Research Center for Functional Food, Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Lihu Avenue 1800, Wuxi, Jiangsu 214100, P.R. China
| | - Jun Ding
- Department of Chemistry,
Wuhan University, Wuhan, Hubei 430072, P.R. China
| | - Shuxiang Geng
- Yunnan Academy of Forestry and Grassland, Kunming, Yunnan 650201, P.R. China
| | - Deping Xu
- State Key Laboratory of Food Science and Technology,
School of Food Science and Technology, National Engineering Research Center for Functional Food, Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Lihu Avenue 1800, Wuxi, Jiangsu 214100, P.R. China
| | - Yinzhi Zhang
- State Key Laboratory of Food Science and Technology,
School of Food Science and Technology, National Engineering Research Center for Functional Food, Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Lihu Avenue 1800, Wuxi, Jiangsu 214100, P.R. China
| | - Xiulan Sun
- State Key Laboratory of Food Science and Technology,
School of Food Science and Technology, National Engineering Research Center for Functional Food, Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Lihu Avenue 1800, Wuxi, Jiangsu 214100, P.R. China
| |
Collapse
|
41
|
Zhuang Z, Dong X, Jia J, Liu Z, Huang T, Qi L. Sleep Patterns, Plasma Metabolome, and Risk of Incident Type 2 Diabetes Mellitus. J Clin Endocrinol Metab 2023; 108:e1034-e1043. [PMID: 37084357 DOI: 10.1210/clinem/dgad218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/06/2023] [Accepted: 04/11/2023] [Indexed: 04/23/2023]
Abstract
CONTEXT A healthy sleep pattern has been related to a lower risk of type 2 diabetes mellitus (T2DM). OBJECTIVE We aimed to identify the metabolomic signature for the healthy sleep pattern and assess its potential causality with T2DM. METHODS This study included 78 659 participants with complete phenotypic data (sleep information and metabolomic measurements) from the UK Biobank study. Elastic net regularized regression was applied to calculate a metabolomic signature reflecting overall sleep patterns. We also performed genome-wide association analysis of the metabolomic signature and one-sample mendelian randomization (MR) with T2DM risk. RESULTS During a median of 8.8 years of follow-up, we documented 1489 incident T2DM cases. Compared with individuals who had an unhealthy sleep pattern, those with a healthy sleep pattern had a 49% lower risk of T2DM (multivariable-adjusted hazard ratio [HR], 0.51; 95% CI, 0.40-0.63). We further constructed a metabolomic signature using elastic net regularized regressions that comprised 153 metabolites, and robustly correlated with sleep pattern (r = 0.19; P = 3×10-325). In multivariable Cox regressions, the metabolomic signature showed a statistically significant inverse association with T2DM risk (HR per SD increment in the signature, 0.56; 95% CI, 0.52-0.60). Additionally, MR analyses indicated a significant causal relation between the genetically predicted metabolomic signature and incident T2DM (P for trend < .001). CONCLUSION In this large prospective study, we identified a metabolomic signature for the healthy sleep pattern, and such a signature showed a potential causality with T2DM risk independent of traditional risk factors.
Collapse
Affiliation(s)
- Zhenhuang Zhuang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Xue Dong
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Jinzhu Jia
- Department of Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Zhonghua Liu
- Department of Biostatistics, Columbia University, New York, NY 10027-6902, USA
| | - Tao Huang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, China
- Center for Intelligent Public Health, Institute for Artificial Intelligence, Peking University, Beijing 100191, China
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70118, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
42
|
Li M, Zhang J, Chen W, Liu S, Liu X, Ning Y, Cao Y, Zhao Y. Supraphysiologic doses of 17β-estradiol aggravate depression-like behaviors in ovariectomized mice possibly via regulating microglial responses and brain glycerophospholipid metabolism. J Neuroinflammation 2023; 20:204. [PMID: 37679787 PMCID: PMC10485970 DOI: 10.1186/s12974-023-02889-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND 17β-Estradiol (E2) is generally considered neuroprotective in humans. However, the current clinical use of estrogen replacement therapy (ERT) is based on the physiological dose of E2 to treat menopausal syndrome and has limited therapeutic efficacy. The efficacy and potential toxicity of superphysiological doses of ERT for menopausal neurodegeneration are unknown. METHODS In this study, we investigated the effect of E2 with a supraphysiologic dose (0.5 mg/kg, sE2) on the treatment of menopausal mouse models established by ovariectomy. We performed the open field, Y-maze spontaneous alternation, forced swim tests, and sucrose preference test to investigate behavioral alterations. Subsequently, the status of microglia and neurons was detected by immunohistochemistry, HE staining, and Nissl staining, respectively. Real-time PCR was used to detect neuroinflammatory cytokines in the hippocampus and cerebral cortex. Using mass spectrometry proteomics platform and LC-MS/ MS-based metabolomics platform, proteins and metabolites in brain tissues were extracted and analyzed. BV2 and HT22 cell lines and primary neurons and microglia were used to explore the underlying molecular mechanisms in vitro. RESULTS sE2 aggravated depression-like behavior in ovariectomized mice, caused microglia response, and increased proinflammatory cytokines in the cerebral cortex and hippocampus, as well as neuronal damage and glycerophospholipid metabolism imbalance. Subsequently, we demonstrated that sE2 induced the pro-inflammatory phenotype of microglia through ERα/NF-κB signaling pathway and downregulated the expression of cannabinoid receptor 1 in neuronal cells, which were important in the pathogenesis of depression. CONCLUSION These data suggest that sE2 may be nonhelpful or even detrimental to menopause-related depression, at least partly, by regulating microglial responses and glycerophospholipid metabolism.
Collapse
Affiliation(s)
- Ming Li
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Jing Zhang
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Wendi Chen
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Shuang Liu
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Xin Liu
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yunna Ning
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yongzhi Cao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yueran Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
43
|
Simon L, Admon R. From childhood adversity to latent stress vulnerability in adulthood: the mediating roles of sleep disturbances and HPA axis dysfunction. Neuropsychopharmacology 2023; 48:1425-1435. [PMID: 37391592 PMCID: PMC10425434 DOI: 10.1038/s41386-023-01638-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/29/2023] [Accepted: 06/16/2023] [Indexed: 07/02/2023]
Abstract
Childhood adversity is a prominent predisposing risk factor for latent stress vulnerability, expressed as an elevated likelihood of developing stress-related psychopathology upon subsequent exposure to trauma in adulthood. Sleep disturbances have emerged as one of the most pronounced maladaptive behavioral outcomes of childhood adversity and are also a highly prevalent core feature of stress-related psychopathology, including post-traumatic stress disorder (PTSD). After reviewing the extensive literature supporting these claims, the current review addresses the notion that childhood adversity-induced sleep disturbances may play a causal role in elevating individuals' stress vulnerability in adulthood. Corroborating this, sleep disturbances that predate adult trauma exposure have been associated with an increased likelihood of developing stress-related psychopathology post-exposure. Furthermore, novel empirical evidence suggests that sleep disturbances, including irregularity of the sleep-wake cycle, mediate the link between childhood adversity and stress vulnerability in adulthood. We also discuss cognitive and behavioral mechanisms through which such a cascade may evolve, highlighting the putative role of impaired memory consolidation and fear extinction. Next, we present evidence to support the contribution of the hypothalamic-pituitary-adrenal (HPA) axis to these associations, stemming from its critical role in stress and sleep regulatory pathways. Childhood adversity may yield bi-directional effects within the HPA stress and sleep axes in which sleep disturbances and HPA axis dysfunction reinforce each other, leading to elevated stress vulnerability. To conclude, we postulate a conceptual path model from childhood adversity to latent stress vulnerability in adulthood and discuss the potential clinical implications of these notions, while highlighting directions for future research.
Collapse
Affiliation(s)
- Lisa Simon
- School of Psychological Sciences, University of Haifa, Haifa, Israel
| | - Roee Admon
- School of Psychological Sciences, University of Haifa, Haifa, Israel.
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, Israel.
| |
Collapse
|
44
|
Abstract
Epidemiologic studies have demonstrated that short sleep duration is associated with an increased risk of cardio-metabolic health outcomes including cardiovascular disease mortality, coronary heart disease, type 2 diabetes mellitus, hypertension, and metabolic syndrome. Experimental sleep restriction studies have sought to explain these findings. This review describes the main evidence of these associations and possible mechanisms explaining them. Whether sleep extension reverses these now widely acknowledged adverse health effects and the feasibility of implementing such strategies on a public health level is discussed.
Collapse
Affiliation(s)
- Roo Killick
- Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Lachlan Stranks
- Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, University of Sydney, Sydney, Australia; The University of Adelaide, Faculty of Health and Medical Sciences, Adelaide, Australia
| | - Camilla M Hoyos
- Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, University of Sydney, Sydney, Australia; The University of Sydney, Faculty of Science, School of Psychology and Brain and Mind Centre, Sydney, Australia.
| |
Collapse
|
45
|
Masanneck L, Voth J, Huntemann N, Öztürk M, Schroeter CB, Ruck T, Meuth SG, Pawlitzki M. Introducing electronic monitoring of disease activity in patients with chronic inflammatory demyelinating polyneuropathy (EMDA CIDP): trial protocol of a proof of concept study. Neurol Res Pract 2023; 5:39. [PMID: 37612774 PMCID: PMC10464162 DOI: 10.1186/s42466-023-00267-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 07/07/2023] [Indexed: 08/25/2023] Open
Abstract
INTRODUCTION Chronic inflammatory demyelinating polyneuropathy (CIDP) is one of the most common immune neuropathies leading to severe impairments in daily life. Current treatment options include intravenous immunoglobulins (IVIG), which are administered at intervals of 4-12 weeks. Determination of individual treatment intervals is challenging since existing clinical scores lack sensitivity to objectify small, partially fluctuating deficits in patients. End-of-dose phenomena described by patients, manifested by increased fatigue and worsening of (motor) symptoms, are currently difficult to detect. From a medical and socio-economic point of view, it is necessary to identify and validate new, more sensitive outcome measures for accurate mapping of disease progression and, thus, for interval finding. Digital health technologies such as wearables may be particularly useful for this purpose, as they record real-life data and consequently, in contrast to classic clinical 'snapshots', can continuously depict the disease course. METHODS In this prospective, observational, non-interventional, single-center, investigator-initiated study, CIDP patients treated with IVIG will be continuously monitored over a period of 6 months. Clinical scores and blood analyses will be assessed and collected during three visits (V1, V2, V3). Additionally, activity, sleep, and cardiac parameters will be recorded over the entire period using a wearable device. Further, patients' subjective disease development and quality of life will be recorded at various visits (read-outs). The usability of the smartwatch will be assessed at the end of the study. PERSPECTIVE The study aims to evaluate different digital measurements obtained with the smartwatch and blood-based analyses for monitoring disease activity and progress in CIDP patients. In conjunction, both means of monitoring might offer detailed insights into behavioral and biological patterns associated with treatment-related fluctuations such as end-of-dose phenomena. TRIAL REGISTRATION The study protocol was registered at ClinicalTrials.gov. Identifier: NCT05723848. Initially, the protocol was submitted prospectively on January 10, 2023. The trial was publicly released after formal improvements on February 13, 2023, after first patients were included according to the original protocol.
Collapse
Affiliation(s)
- Lars Masanneck
- Department of Neurology, Medical Faculty University Hospital Düsseldorf, Heinrich-Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
- Hasso Plattner Institute, University of Potsdam, 14482, Potsdam, Germany
| | - Jan Voth
- Department of Neurology, Medical Faculty University Hospital Düsseldorf, Heinrich-Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Niklas Huntemann
- Department of Neurology, Medical Faculty University Hospital Düsseldorf, Heinrich-Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Menekse Öztürk
- Department of Neurology, Medical Faculty University Hospital Düsseldorf, Heinrich-Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Christina B Schroeter
- Department of Neurology, Medical Faculty University Hospital Düsseldorf, Heinrich-Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty University Hospital Düsseldorf, Heinrich-Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty University Hospital Düsseldorf, Heinrich-Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty University Hospital Düsseldorf, Heinrich-Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
46
|
Li X, He J, Sun Q. Sleep Duration and Sarcopenia: An Updated Systematic Review and Meta-Analysis. J Am Med Dir Assoc 2023; 24:1193-1206.e5. [PMID: 37295459 DOI: 10.1016/j.jamda.2023.04.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 06/12/2023]
Abstract
OBJECTIVES In adults, short and long sleep duration has been associated with sarcopenia risk. Studies have shown that various factors, including biological and psychological factors, could be the underlying cause of the association between aberrant sleep duration and sarcopenia risk. In this study, we have qualitatively and quantitatively summarized previously published studies on sleep duration to assess the relationship between sleep duration and sarcopenia risk in adults. This would aid in enhancing our understanding of recent advancements in this field and the association between sleep duration and sarcopenia risk. DESIGN Systematic review and meta-analysis. SETTING AND PARTICIPANTS In this review, we included studies evaluating the association between the duration of sleep and sarcopenia in adults in observational studies. METHODS Five electronic databases (PubMed, EMBASE, Cochrane Library, China National Knowledge Infrastructure, and Web of Science) were searched to April 20, 2023, to identify studies related to sarcopenia and sleep duration. Next, we calculated the odds ratios (ORs) for sarcopenia prevalence based on the adjusted data from individual studies. Statistical analyses were performed using Stata 11.0. RESULTS Sarcopenia prevalence was high (18%) in adults with long sleep duration. Our results showed a significant association between short duration of sleep and high sarcopenia prevalence in older adults (OR 1.2, 95% CI 1.02-1.41, I2 = 56.6%). Furthermore, a significant association was observed between all participants with long-duration sleep and high sarcopenia prevalence (OR 1.53, 95% CI 1.34-1.75, I2 = 56.8%). We also observed significant heterogeneity in the adjusted ORs. CONCLUSIONS AND IMPLICATIONS There was a correlation between sarcopenia and short or long sleep duration, especially in older adults. In adults with a long duration of sleep, sarcopenia prevalence was relatively high.
Collapse
Affiliation(s)
- Xiaoyan Li
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie He
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Qiuhua Sun
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
47
|
Knauert MP, Adekolu O, Xu Z, Deng A, Chu JH, Baldassarri SR, Kushida C, Yaggi HK, Zinchuk A. Morning Chronotype Is Associated with Improved Adherence to Continuous Positive Airway Pressure among Individuals with Obstructive Sleep Apnea. Ann Am Thorac Soc 2023; 20:1182-1191. [PMID: 36917194 PMCID: PMC10405611 DOI: 10.1513/annalsats.202210-885oc] [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: 10/24/2022] [Accepted: 03/13/2023] [Indexed: 03/16/2023] Open
Abstract
Rationale: Poor adherence limits the effectiveness of continuous positive airway pressure (CPAP) therapy for obstructive sleep apnea (OSA). A better understanding of CPAP adherence is needed to develop novel strategies to improve it. Objectives: To determine if the chronotype (morning, evening, or intermediate) of patients with OSA is associated with differences in CPAP adherence. If such an association exists, determine the mechanisms underlying this association. Methods: We performed a secondary analysis of the APPLES (Apnea Positive Pressure Long-term Efficacy Study) clinical trial. We assessed chronotype using the Morningness-Eveningness Questionnaire (MEQ) among participants randomized to the CPAP arm with daily adherence data (n = 469). Evening (MEQ ⩽ 41), intermediate (41 < MEQ < 59), and morning type (MEQ ⩾ 59) categories were the exposures. We modeled daily CPAP use (hours per night) over a 6-month period, using a linear mixed model, adjusted for covariates (e.g., age, sex, marital status). To assess mechanisms of the association, we performed mediation analyses using sleep duration, weekend catch-up sleep, depression, and other factors. Results: Most participants were obese men with severe OSA (body mass index of 32.3 ± 7.3 kg/m2, 65% male, and apnea-hypopnea index 39.8 ± 24.6/h). Participants were 44% morning, 47% intermediate, and 8% evening chronotype. Participants with the morning chronotype reported the shortest sleep duration on weekends (7.3 vs. 7.6 and 7.9 h/night) compared with the intermediate and evening types. Participants with the morning chronotype exhibited a 40-min/night higher CPAP use (P = 0.001) than persons with the intermediate chronotype. This relationship was mildly attenuated (32.8 min/night; P = 0.011) after adjustment for covariates. None of the selected factors (e.g., sleep duration, weekend catch-up sleep) exhibited a significant mediation effect. Conclusions: Morning chronotype is associated with a clinically meaningful increase in CPAP adherence compared with other chronotypes. Mechanisms of this association require further study. Chronotype may be a novel predictor of CPAP adherence. Clinical trial registered with www.clinicaltrials.gov (NCT00051363).
Collapse
Affiliation(s)
- Melissa P. Knauert
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Olurotimi Adekolu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Zhichao Xu
- Department of Biostatistics, Yale University School of Public Health, New Haven, Connecticut
| | - Annan Deng
- Department of Biostatistics, Yale University School of Public Health, New Haven, Connecticut
| | - Jen-hwa Chu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Department of Biostatistics, Yale University School of Public Health, New Haven, Connecticut
| | - Stephen R. Baldassarri
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Clete Kushida
- Stanford University Sleep Clinic and Center for Human Sleep Research, Redwood City, California; and
| | - H. Klar Yaggi
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Clinical Epidemiology Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Andrey Zinchuk
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
48
|
Edelbo BL, Andreassen SN, Steffensen AB, MacAulay N. Day-night fluctuations in choroid plexus transcriptomics and cerebrospinal fluid metabolomics. PNAS NEXUS 2023; 2:pgad262. [PMID: 37614671 PMCID: PMC10443925 DOI: 10.1093/pnasnexus/pgad262] [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: 04/13/2023] [Revised: 07/06/2023] [Accepted: 07/31/2023] [Indexed: 08/25/2023]
Abstract
The cerebrospinal fluid (CSF) provides mechanical protection for the brain and serves as a brain dispersion route for nutrients, hormones, and metabolic waste. The CSF secretion rate is elevated in the dark phase in both humans and rats, which could support the CSF flow along the paravascular spaces that may be implicated in waste clearance. The similar diurnal CSF dynamics pattern observed in the day-active human and the nocturnal rat suggests a circadian regulation of this physiological variable, rather than sleep itself. To obtain a catalog of potential molecular drivers that could provide the day-night-associated modulation of the CSF secretion rate, we determined the diurnal fluctuation in the rat choroid plexus transcriptomic profile with RNA-seq and in the CSF metabolomics with ultraperformance liquid chromatography combined with mass spectrometry. We detected significant fluctuation of 19 CSF metabolites and differential expression of 2,778 choroid plexus genes between the light and the dark phase, the latter of which encompassed circadian rhythm-related genes and several choroid plexus transport mechanisms. The fluctuating components were organized with joint pathway analysis, of which several pathways demonstrated diurnal regulation. Our results illustrate substantial transcriptional and metabolic light-dark phase-mediated changes taking place in the rat choroid plexus and its encircling CSF. The combined data provide directions toward future identification of the molecular pathways governing the fluctuation of this physiological process and could potentially be harnessed to modulate the CSF dynamics in pathology.
Collapse
Affiliation(s)
| | | | | | - Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
49
|
Soliz-Rueda JR, López-Fernández-Sobrino R, Torres-Fuentes C, Bravo FI, Suárez M, Mulero M, Muguerza B. Metabolism disturbance by light/dark cycle switching depends on the rat health status: the role of grape seed flavanols. Food Funct 2023; 14:6443-6454. [PMID: 37377055 DOI: 10.1039/d3fo00260h] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Changes in light/dark cycles and obesogenic diets are related to the disruption of circadian rhythms and metabolic disorders. Grape seed flavanols have shown beneficial effects on metabolic diseases and, recently, a circadian system modulation has been suggested to mediate their health-enhancing properties. Therefore, the aim of this study was to evaluate the grape seed (poly)phenol extract (GSPE) effects in healthy and obese rats after a light/dark cycle disruption. Forty-eight rats were fed a standard (STD) or cafeteria (CAF) diet for 6 weeks under STD conditions of a light/dark cycle (12 h light per day, L12). Then, animals were switched to a long (18 h light per day, L18) or short (6 h light per day, L6) photoperiod and administered a vehicle (VH) or GSPE (25 mg kg-1) for 1 week. The results showed changes in serum lipids and insulin and metabolomic profiles dependent on the photoperiod and animal health status. GSPE administration improved serum parameters and increased the Nampt gene expression in CAF rats and modified the metabolomic profile in a photoperiod-dependent manner. Metabolic effects of light/dark disturbance depend on the health status of the rats, with diet-induced CAF-induced obese rats being more affected. Grape seed flavanols improve the metabolic status in a photoperiod-dependent manner and their effects on the circadian system suggest that part of their metabolic effects could be mediated by their action on biological rhythms.
Collapse
Affiliation(s)
- Jorge R Soliz-Rueda
- University Rovira i Virgili, Biochemistry and Biotechnology Department, Nutrigenomics Research Group, Tarragona, 43007, Spain.
- Pere Virgili Institute for Health Research (IISPV), Tarragona, 43007, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), University Rovira i Virgili, 43007 Tarragona, Spain
| | - Raúl López-Fernández-Sobrino
- University Rovira i Virgili, Biochemistry and Biotechnology Department, Nutrigenomics Research Group, Tarragona, 43007, Spain.
| | - Cristina Torres-Fuentes
- University Rovira i Virgili, Biochemistry and Biotechnology Department, Nutrigenomics Research Group, Tarragona, 43007, Spain.
- Pere Virgili Institute for Health Research (IISPV), Tarragona, 43007, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), University Rovira i Virgili, 43007 Tarragona, Spain
| | - Francisca I Bravo
- University Rovira i Virgili, Biochemistry and Biotechnology Department, Nutrigenomics Research Group, Tarragona, 43007, Spain.
- Pere Virgili Institute for Health Research (IISPV), Tarragona, 43007, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), University Rovira i Virgili, 43007 Tarragona, Spain
| | - Manuel Suárez
- University Rovira i Virgili, Biochemistry and Biotechnology Department, Nutrigenomics Research Group, Tarragona, 43007, Spain.
- Pere Virgili Institute for Health Research (IISPV), Tarragona, 43007, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), University Rovira i Virgili, 43007 Tarragona, Spain
| | - Miquel Mulero
- University Rovira i Virgili, Biochemistry and Biotechnology Department, Nutrigenomics Research Group, Tarragona, 43007, Spain.
- Pere Virgili Institute for Health Research (IISPV), Tarragona, 43007, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), University Rovira i Virgili, 43007 Tarragona, Spain
| | - Begoña Muguerza
- University Rovira i Virgili, Biochemistry and Biotechnology Department, Nutrigenomics Research Group, Tarragona, 43007, Spain.
- Pere Virgili Institute for Health Research (IISPV), Tarragona, 43007, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), University Rovira i Virgili, 43007 Tarragona, Spain
| |
Collapse
|
50
|
Xu Y, Qu B, Liu F, Gong Z, Zhang Y, Xu D. Sleep Deprivation and Heart Rate Variability in Healthy Volunteers: Effects of REM and SWS Sleep Deprivation. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2023; 2023:7121295. [PMID: 37469834 PMCID: PMC10353901 DOI: 10.1155/2023/7121295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 07/21/2023]
Abstract
Objective Using PSG-guided acute selective REM/SWS sleep deprivation in volunteers, this study examined the effects of sleep deprivation on the cardiovascular and autonomic nervous systems, as well as the relationship between cardiac neuromodulation homeostasis and cardiovascular disease. Methods An experiment was conducted using 30 healthy volunteers (male : female = 1 : 1, aged 26.33 ± 4.5 years) divided into groups for sleep deprivation of SWS and REM sleep, and then, each group was crossed over for normal sleep (2 days) and repeated sleep deprivation (1 day, 3 times). During the study period, PSG and ELECTRO ECG monitoring were conducted, and five-minute frequency domain parameters and blood pressure values were measured before and after sleep deprivation. Results Changes in VLF, LFnu, LF/HF, HF, and HFnu after SWS sleep deprivation were statistically significant (P < 0.05), but not LF (P = 0.063). Changes in VLF, LF, HF, LF/HF, LFnu, and HFnu after REM sleep deprivation were not statistically significant (P > 0.05). Conclusions An increase in sympathetic nerve activity results from sleep deprivation and sudden awakening from SWS sleep is associated with a greater risk of cardiovascular disease.
Collapse
Affiliation(s)
- YaHui Xu
- Department of Respiratory and Critical Care Medicine, The Affiliated Central Hospital of Qingdao University, Qingdao, Shandong, China
| | - BinBin Qu
- Department of Respiratory and Critical Care Medicine, The Affiliated Central Hospital of Qingdao University, Qingdao, Shandong, China
| | - FengJuan Liu
- Clinical Trial Research Center, The Affiliated Central Hospital of Qingdao University, Qingdao, Shandong, China
| | - ZhiHua Gong
- Electrocardiogram Department, The Affiliated Central Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yi Zhang
- Department of Respiratory and Critical Care Medicine, Beijing China-Japan Friendship Hospital, China
| | - DeXiang Xu
- Department of Respiratory and Critical Care Medicine, The Affiliated Central Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|