1
|
Gachon F, Bugianesi E, Castelnuovo G, Oster H, Pendergast JS, Montagnese S. Potential bidirectional communication between the liver and the central circadian clock in MASLD. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:15. [PMID: 40225783 PMCID: PMC11981938 DOI: 10.1038/s44324-025-00058-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/25/2025] [Indexed: 04/15/2025]
Abstract
Most aspects of physiology and behaviour fluctuate every 24 h in mammals. These circadian rhythms are orchestrated by an autonomous central clock located in the suprachiasmatic nuclei that coordinates the timing of cellular clocks in tissues throughout the body. The critical role of this circadian system is emphasized by increasing evidence associating disruption of circadian rhythms with diverse pathologies. Accordingly, mounting evidence suggests a bidirectional relationship where disruption of rhythms by circadian misalignment may contribute to liver diseases while liver diseases alter the central clock and circadian rhythms in other tissues. Therefore, liver pathophysiology may broadly impact the circadian system and may provide a mechanistic framework for understanding and targeting metabolic diseases and adjust metabolic setpoints.
Collapse
Affiliation(s)
- Frédéric Gachon
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus, Denmark
| | | | | | - Henrik Oster
- Institute of Neurobiology, Center of Brain, Behavior & Metabolism, University of Lübeck, Lübeck, Germany
| | | | - Sara Montagnese
- Department of Medicine, University of Padova, Padova, Italy
- Chronobiology Section, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
2
|
Smith HA, Templeman I, Davis M, Slater T, Clayton DJ, Varley I, James LJ, Middleton B, Johnston JD, Karagounis LG, Tsintzas K, Thompson D, Gonzalez JT, Walhin JP, Betts JA. Characterizing 24-Hour Skeletal Muscle Gene Expression Alongside Metabolic and Endocrine Responses Under Diurnal Conditions. J Clin Endocrinol Metab 2025; 110:e1017-e1030. [PMID: 38779872 PMCID: PMC11913097 DOI: 10.1210/clinem/dgae350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
CONTEXT Skeletal muscle plays a central role in the storage, synthesis, and breakdown of nutrients, yet little research has explored temporal responses of this human tissue, especially with concurrent measures of systemic biomarkers of metabolism. OBJECTIVE To characterize temporal profiles in skeletal muscle expression of genes involved in carbohydrate metabolism, lipid metabolism, circadian clocks, and autophagy and descriptively relate them to systemic metabolites and hormones during a controlled laboratory protocol. METHODS Ten healthy adults (9M/1F, [mean ± SD] age 30 ± 10 years; BMI 24.1 ± 2.7 kg·m-2) rested in the laboratory for 37 hours with all data collected during the final 24 hours (08:00-08:00 hours). Participants ingested hourly isocaloric liquid meal replacements alongside appetite assessments during waking before a sleep opportunity from 22:00 to 07:00 hours. Blood samples were collected hourly for endocrine and metabolite analyses, with muscle biopsies occurring every 4 hours from 12:00 to 08:00 hours the following day to quantify gene expression. RESULTS Plasma insulin displayed diurnal rhythmicity peaking at 18:04 hours. Expression of skeletal muscle genes involved in carbohydrate metabolism (Name, Acrophase [hours]: GLUT4, 14:40; PPARGC1A, 16:13; HK2, 18:24) and lipid metabolism (FABP3, 12:37; PDK4, 05:30; CPT1B, 12:58) displayed 24-hour rhythmicity that reflected the temporal rhythm of insulin. Equally, circulating glucose (00:19 hours), nonesterified fatty acids (04:56), glycerol (04:32), triglyceride (23:14), urea (00:46), C-terminal telopeptide (05:07), and cortisol (22:50) concentrations also all displayed diurnal rhythmicity. CONCLUSION Diurnal rhythms were present in human skeletal muscle gene expression as well systemic metabolites and hormones under controlled diurnal conditions. The temporal patterns of genes relating to carbohydrate and lipid metabolism alongside circulating insulin are consistent with diurnal rhythms being driven in part by the diurnal influence of cyclic feeding and fasting.
Collapse
Affiliation(s)
- Harry A Smith
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK, BA2 7AY
| | - Iain Templeman
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK, BA2 7AY
| | - Max Davis
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK, BA2 7AY
| | - Tommy Slater
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK, NG1 4FQ
| | - David J Clayton
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK, NG1 4FQ
| | - Ian Varley
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK, NG1 4FQ
| | - Lewis J James
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK, LE11 3TU
| | - Benita Middleton
- Section of Chronobiology, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK, GU2 7XH
| | - Jonathan D Johnston
- Section of Chronobiology, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK, GU2 7XH
| | - Leonidas G Karagounis
- Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland
- Mary MacKillop Institute for Health Research (MMIHR), Australian Catholic University (ACU), Melbourne, VIC 3000, Australia
| | - Kostas Tsintzas
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK, NG7 2UH
| | - Dylan Thompson
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK, BA2 7AY
| | - Javier T Gonzalez
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK, BA2 7AY
| | - Jean-Philippe Walhin
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK, BA2 7AY
| | - James A Betts
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK, BA2 7AY
| |
Collapse
|
3
|
Barker DH, Carskadon MA, Gredvig-Ardito C, Hart CN, Raynor HA, Scheer FAJL. Independent effects of the human circadian system and sleep/eating cycles on caloric intake in adolescents vary by weight status. Proc Natl Acad Sci U S A 2025; 122:e2407907122. [PMID: 39964717 PMCID: PMC11874483 DOI: 10.1073/pnas.2407907122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/16/2024] [Indexed: 02/20/2025] Open
Abstract
Late-day eating is linked to increased obesity risk; however, whether the endogenous circadian system independently influences caloric intake and if this control differs among individuals based on weight status is unknown. Here, we investigated in adolescents the independent roles of the endogenous circadian system and of the behavioral sleep/wake cycle (sleep/wake, fasting/eating, rest/activity, dark/dim light, social interaction, posture, etc.) on self-selected caloric intake using a Forced Desynchrony protocol. Fifty-one male and female adolescents across three weight status categories (24 with healthy weight, 13 with overweight, and 14 with obesity) completed the protocol where participants lived on seven 28-h sleep/wake cycles in dim light during wake and complete darkness during sleep. Results suggest that the circadian system and the behavioral cycle each affected caloric intake, with a decrease across the wake episode and an increase from circadian morning to circadian evening in caloric intake. The endogenous circadian rhythm in caloric intake showed a circadian peak-to-trough difference of 196 [CI 95% 164, 226] kcal per meal with peak timing of 296° [288°, 304°; equivalent to ~17:30 in these participants]. In those with overweight/obesity, more calories were consumed later in the waking episode and later in the circadian cycle, and with blunted amplitudes compared to those with healthy weight. Results implicate both the endogenous circadian system and the behavioral cycle in shaping the daily rhythm of food intake. Furthermore, these results help explain the increased drive for caloric intake toward the evening, especially in those at risk for obesity.
Collapse
Grants
- P20GM139743 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- R01HL153969 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL153969 NHLBI NIH HHS
- R01HL140574 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL167746 NHLBI NIH HHS
- P20 GM139743 NIGMS NIH HHS
- R01 HL140574 NHLBI NIH HHS
- R01HL164454 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL167746 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 DK101046 NIDDK NIH HHS
- R01DK101046 HHS | NIH | NIDDK | Division of Diabetes, Endocrinology, and Metabolic Diseases (DEM)
- R01 HL164454 NHLBI NIH HHS
Collapse
Affiliation(s)
- David H. Barker
- Department of Psychiatry and Human Behavior, Bradley Hospital Sleep Research Laboratory, Alpert Medical School of Brown University, Providence, RI02906
- Department of Psychiatry and Human Behavior, The Warren Alpert Medical School of Brown University, Providence, RI02906
| | - Mary A. Carskadon
- Department of Psychiatry and Human Behavior, Bradley Hospital Sleep Research Laboratory, Alpert Medical School of Brown University, Providence, RI02906
- Department of Psychiatry and Human Behavior, The Warren Alpert Medical School of Brown University, Providence, RI02906
| | - Caroline Gredvig-Ardito
- Department of Psychiatry and Human Behavior, Bradley Hospital Sleep Research Laboratory, Alpert Medical School of Brown University, Providence, RI02906
| | - Chantelle N. Hart
- Department of Social and Behavioral Sciences and Center for Obesity Research and Education, College of Public Health, Temple University, Philadelphia, PA19140
| | - Hollie A. Raynor
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN37996
| | - Frank A. J. L. Scheer
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Division of Sleep and Circadian Disorders, Department of Neurology, Brigham and Women’s Hospital, Boston, MA02115
- Division of Sleep Medicine, Harvard Medical School, Boston, MA02115
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
| |
Collapse
|
4
|
Daneshzad E, Janmohammadi P, Basirat V, Qorbani M, Azadbakht L. Egg consumption, sleep, and mental health status among women with type II diabetes. Sci Rep 2025; 15:1368. [PMID: 39779825 PMCID: PMC11711463 DOI: 10.1038/s41598-025-85347-x] [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: 04/13/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
To evaluate if egg consumption is associated with sleep quality and psychological health (depression, anxiety, and stress) in women with type 2 diabetes. A cross-sectional study was conducted on women with type 2 diabetes (n = 230). Weight, height, waist circumference (WC), and blood pressure were measured. A Food Frequency Questionnaire (FFQ) was used to obtain dietary intake data and estimate total egg consumption, which was presented in tertiles. The Pittsburgh Sleep Quality Index (PSQI) and Depression, Anxiety, and Stress Scale (DASS-21) were used to evaluate sleep and mental health outcomes, respectively. Dietary intake of carbohydrates, sodium, saturated fatty acids, and cholesterol was greater in the highest tertile of egg consumption (P < 0.05). Dietary intake of fat, monounsaturated, and polyunsaturated fatty acids was lower in the highest tertile of egg consumption (P < 0.05). WC was greater in the highest tertile of egg consumption in the crude model (p = 0.03), however, there was no evidence of this association in the adjusted model. There was no evidence of an association between egg consumption and the odds of poor psychological health or sleep quality in unadjusted or adjusted models. There was no association between egg consumption and poor sleep or mental disorders. Future studies are needed to confirm these findings and to identify the mechanism of action.
Collapse
Affiliation(s)
- Elnaz Daneshzad
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Parisa Janmohammadi
- Kuopio Musculoskeletal Research Unit, University of Eastern Finland, Kuopio, Finland
| | - Vahid Basirat
- Department of Gastroenterology, School of Medicine, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran
| | - Mostafa Qorbani
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Department of Epidemiology, Non-Communicable Diseases Research Center, Endocrinology, and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Azadbakht
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, P. O. Box: 1416643931, Tehran, Iran.
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Hannemann J, Laing A, Middleton B, Schwedhelm E, Marx N, Federici M, Kastner M, Skene DJ, Böger R. Effect of oral melatonin treatment on insulin resistance and diurnal blood pressure variability in night shift workers. A double-blind, randomized, placebo-controlled study. Pharmacol Res 2024; 199:107011. [PMID: 38029806 DOI: 10.1016/j.phrs.2023.107011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/21/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Night shift work is associated with sleep disturbances, obesity, and cardiometabolic diseases. Disruption of the circadian clock system has been suggested to be an independent cause of type 2 diabetes and cardiovascular disease in shift workers. We aimed to improve alignment of circadian timing with social and environmental factors with administration of melatonin. METHODS In a randomized, placebo-controlled, prospective study, we analysed the effects of 2 mg of sustained-release melatonin versus placebo on glucose tolerance, insulin resistance indices, sleep quality, circadian profiles of plasma melatonin and cortisol, and diurnal blood pressure profiles in 24 rotating night shift workers during 12 weeks of treatment, followed by 12 weeks of wash-out. In a novel design, the time of melatonin administration (at night or in the morning) depended upon the shift schedule. We also compared the baseline profiles of the night shift (NS) workers with 12 healthy non-night shift (NNS)-working controls. RESULTS We found significantly impaired indices of insulin resistance at baseline in NS versus NNS (p < 0.05), but no differences in oral glucose tolerance tests nor in the diurnal profiles of melatonin, cortisol, or blood pressure. Twelve weeks of melatonin treatment did not significantly improve insulin resistance, nor did it significantly affect diurnal blood pressure or melatonin and cortisol profiles. Melatonin administration, however, caused a significant improvement in sleep quality which was significantly impaired in NS versus NNS at baseline (p < 0.001). CONCLUSIONS Rotating night shift work causes mild-to-moderate impairment of sleep quality and insulin resistance. Melatonin treatment at bedtime improves sleep quality, but does not significantly affect insulin resistance in rotating night shift workers after 12 weeks of administration.
Collapse
Affiliation(s)
- Juliane Hannemann
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anika Laing
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benita Middleton
- Chronobiology, University of Surrey, Guildford, Surrey, England, UK
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nikolaus Marx
- Department of Cardiology, University Medical Center Aachen, Aachen, Germany
| | - Massimo Federici
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Mariola Kastner
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Debra J Skene
- Chronobiology, University of Surrey, Guildford, Surrey, England, UK
| | - Rainer Böger
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
6
|
Gubin D, Danilenko K, Stefani O, Kolomeichuk S, Markov A, Petrov I, Voronin K, Mezhakova M, Borisenkov M, Shigabaeva A, Yuzhakova N, Lobkina S, Weinert D, Cornelissen G. Blue Light and Temperature Actigraphy Measures Predicting Metabolic Health Are Linked to Melatonin Receptor Polymorphism. BIOLOGY 2023; 13:22. [PMID: 38248453 PMCID: PMC10813279 DOI: 10.3390/biology13010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/29/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024]
Abstract
This study explores the relationship between the light features of the Arctic spring equinox and circadian rhythms, sleep and metabolic health. Residents (N = 62) provided week-long actigraphy measures, including light exposure, which were related to body mass index (BMI), leptin and cortisol. Lower wrist temperature (wT) and higher evening blue light exposure (BLE), expressed as a novel index, the nocturnal excess index (NEIbl), were the most sensitive actigraphy measures associated with BMI. A higher BMI was linked to nocturnal BLE within distinct time windows. These associations were present specifically in carriers of the MTNR1B rs10830963 G-allele. A larger wake-after-sleep onset (WASO), smaller 24 h amplitude and earlier phase of the activity rhythm were associated with higher leptin. Higher cortisol was associated with an earlier M10 onset of BLE and with our other novel index, the Daylight Deficit Index of blue light, DDIbl. We also found sex-, age- and population-dependent differences in the parametric and non-parametric indices of BLE, wT and physical activity, while there were no differences in any sleep characteristics. Overall, this study determined sensitive actigraphy markers of light exposure and wT predictive of metabolic health and showed that these markers are linked to melatonin receptor polymorphism.
Collapse
Affiliation(s)
- Denis Gubin
- Department of Biology, Tyumen Medical University, 625023 Tyumen, Russia
- Laboratory for Chronobiology and Chronomedicine, Research Institute of Biomedicine and Biomedical Technologies, Tyumen Medical University, 625023 Tyumen, Russia; (K.D.); (A.S.)
- Tyumen Cardiology Research Center, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Konstantin Danilenko
- Laboratory for Chronobiology and Chronomedicine, Research Institute of Biomedicine and Biomedical Technologies, Tyumen Medical University, 625023 Tyumen, Russia; (K.D.); (A.S.)
- Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia
| | - Oliver Stefani
- Department Engineering and Architecture, Institute of Building Technology and Energy, Lucerne University of Applied Sciences and Arts, 6048 Horw, Switzerland;
| | - Sergey Kolomeichuk
- Laboratory for Genomics, Proteomics, and Metabolomics, Research Institute of Biomedicine and Biomedical Technologies, Medical University, 625023 Tyumen, Russia; (S.K.); (A.M.); (K.V.); (N.Y.)
- Laboratory of Genetics, Institute of Biology of the Karelian Science Center, Russian Academy of Sciences, 185910 Petrozavodsk, Russia
| | - Alexander Markov
- Laboratory for Genomics, Proteomics, and Metabolomics, Research Institute of Biomedicine and Biomedical Technologies, Medical University, 625023 Tyumen, Russia; (S.K.); (A.M.); (K.V.); (N.Y.)
| | - Ivan Petrov
- Department of Biological & Medical Physics UNESCO, Medical University, 625023 Tyumen, Russia
| | - Kirill Voronin
- Laboratory for Genomics, Proteomics, and Metabolomics, Research Institute of Biomedicine and Biomedical Technologies, Medical University, 625023 Tyumen, Russia; (S.K.); (A.M.); (K.V.); (N.Y.)
| | - Marina Mezhakova
- Laboratory for Genomics, Proteomics, and Metabolomics, Research Institute of Biomedicine and Biomedical Technologies, Medical University, 625023 Tyumen, Russia; (S.K.); (A.M.); (K.V.); (N.Y.)
| | - Mikhail Borisenkov
- Department of Molecular Immunology and Biotechnology, Institute of Physiology of the Federal Research Centre Komi Science Centre, Ural Branch of the Russian Academy of Sciences, 167982 Syktyvkar, Russia;
| | - Aislu Shigabaeva
- Laboratory for Chronobiology and Chronomedicine, Research Institute of Biomedicine and Biomedical Technologies, Tyumen Medical University, 625023 Tyumen, Russia; (K.D.); (A.S.)
| | - Natalya Yuzhakova
- Laboratory for Genomics, Proteomics, and Metabolomics, Research Institute of Biomedicine and Biomedical Technologies, Medical University, 625023 Tyumen, Russia; (S.K.); (A.M.); (K.V.); (N.Y.)
| | - Svetlana Lobkina
- Healthcare Institution of Yamalo-Nenets Autonomous Okrug “Tarko-Sale Central District Hospital”, 629850 Urengoy, Russia;
| | - Dietmar Weinert
- Institute of Biology/Zoology, Martin Luther University, 06108 Halle-Wittenberg, Germany;
| | - Germaine Cornelissen
- Department of Integrated Biology and Physiology, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
7
|
Potes Y, Díaz-Luis A, Bermejo-Millo JC, Pérez-Martínez Z, de Luxán-Delgado B, Rubio-González A, Menéndez-Valle I, Gutiérrez-Rodríguez J, Solano JJ, Caballero B, Vega-Naredo I, Coto-Montes A. Melatonin Alleviates the Impairment of Muscle Bioenergetics and Protein Quality Control Systems in Leptin-Deficiency-Induced Obesity. Antioxidants (Basel) 2023; 12:1962. [PMID: 38001815 PMCID: PMC10669624 DOI: 10.3390/antiox12111962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Leptin is critically compromised in the major common forms of obesity. Skeletal muscle is the main effector tissue for energy modification that occurs as a result of the effect of endocrine axes, such as leptin signaling. Our study was carried out using skeletal muscle from a leptin-deficient animal model, in order to ascertain the importance of this hormone and to identify the major skeletal muscle mechanisms affected. We also examined the therapeutic role of melatonin against leptin-induced muscle wasting. Here, we report that leptin deficiency stimulates fatty acid β-oxidation, which results in mitochondrial uncoupling and the suppression of mitochondrial oxidative damage; however, it increases cytosolic oxidative damage. Thus, different nutrient-sensing pathways are disrupted, impairing proteostasis and promoting lipid anabolism, which induces myofiber degeneration and drives oxidative type I fiber conversion. Melatonin treatment plays a significant role in reducing cellular oxidative damage and regulating energy homeostasis and fuel utilization. Melatonin is able to improve both glucose and mitochondrial metabolism and partially restore proteostasis. Taken together, our study demonstrates melatonin to be a decisive mitochondrial function-fate regulator in skeletal muscle, with implications for resembling physiological energy requirements and targeting glycolytic type II fiber recovery.
Collapse
Affiliation(s)
- Yaiza Potes
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Institute of Neurosciences of the Principality of Asturias (INEUROPA), 33006 Oviedo, Spain
| | - Andrea Díaz-Luis
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
| | - Juan C. Bermejo-Millo
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Institute of Neurosciences of the Principality of Asturias (INEUROPA), 33006 Oviedo, Spain
| | - Zulema Pérez-Martínez
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Microbiology Service, Central University Hospital of Asturias, 33011 Oviedo, Spain
| | - Beatriz de Luxán-Delgado
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
| | - Adrian Rubio-González
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
| | - Iván Menéndez-Valle
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Institute of Neurosciences of the Principality of Asturias (INEUROPA), 33006 Oviedo, Spain
- Immunology Service, Central University Hospital of Asturias, 33011 Oviedo, Spain
| | - José Gutiérrez-Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Geriatric Service, Monte Naranco Hospital, 33012 Oviedo, Spain
| | - Juan J. Solano
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Geriatric Service, Monte Naranco Hospital, 33012 Oviedo, Spain
| | - Beatriz Caballero
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Institute of Neurosciences of the Principality of Asturias (INEUROPA), 33006 Oviedo, Spain
| | - Ignacio Vega-Naredo
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Institute of Neurosciences of the Principality of Asturias (INEUROPA), 33006 Oviedo, Spain
| | - Ana Coto-Montes
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Institute of Neurosciences of the Principality of Asturias (INEUROPA), 33006 Oviedo, Spain
| |
Collapse
|
8
|
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
|
9
|
Li FI, Spence RJ, de Laat MA, Harris PA, Sonntag J, Menzies-Gow NJ, Durham AE, Bailey SR, Sillence MN. Association between insulin dysregulation and adrenocorticotropic hormone in aged horses and ponies with no clinical signs of pituitary pars intermedia dysfunction. Equine Vet J 2023; 55:1003-1011. [PMID: 36641787 DOI: 10.1111/evj.13925] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 01/05/2023] [Indexed: 01/16/2023]
Abstract
BACKGROUND High concentrations of adrenocorticotropic hormone (ACTH) are used as an indicator of pituitary pars intermedia dysfunction (PPID), but other factors that may influence ACTH need to be understood, if diagnostic reference ranges for ACTH are to be used with confidence. Insulin dysregulation (ID) could be one such factor, as insulin affects pituitary hormones in other species. OBJECTIVES To test the hypothesis that a relationship exists between high insulin and high ACTH in aged (>15-year-old) animals with no clinical signs of PPID. STUDY DESIGN A cohort study. METHODS Thirteen horses and eleven ponies (17-25 years-old; mares and geldings) were clinically examined for signs of PPID in the spring (November 2020) and autumn (April 2021). On the same day, blood samples were taken before and 2 h after an oral glucose test (OGT). Concentrations of insulin, glucose, ACTH and cortisol were measured. RESULTS There was no association between ACTH and cortisol. However, there was a positive linear correlation between ACTH and post-OGT (insulin in the autumn (r = 0.427, p = 0.04). Two horses and six ponies had ACTH above the cut-off value for PPID diagnosis, and of these eight animals, six also had insulin concentrations above the cut-off value for ID. MAIN LIMITATIONS The cohort was small and thyrotropin-releasing hormone (TRH) stimulation tests were not performed. CONCLUSIONS In autumn, high ACTH was associated with ID, when no clinical signs of PPID were present. Because ACTH is used in PPID diagnosis, further work is required to understand this interaction.
Collapse
Affiliation(s)
- Fang I Li
- School of Biology & Environmental Science, Science Faculty, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Robert J Spence
- School of Biology & Environmental Science, Science Faculty, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Melody A de Laat
- School of Biology & Environmental Science, Science Faculty, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Patricia A Harris
- Equine Studies Group, Waltham Petcare Science Institute, Leicestershire, UK
| | - Johanna Sonntag
- Boehringer Ingelheim Vetmedica GmbH, Ingelheim am Rhein, Germany
| | | | | | - Simon R Bailey
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Martin N Sillence
- School of Biology & Environmental Science, Science Faculty, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| |
Collapse
|
10
|
Grosjean E, Simonneaux V, Challet E. Reciprocal Interactions between Circadian Clocks, Food Intake, and Energy Metabolism. BIOLOGY 2023; 12:biology12040539. [PMID: 37106739 PMCID: PMC10136292 DOI: 10.3390/biology12040539] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023]
Abstract
Like other biological functions, food intake and energy metabolism display daily rhythms controlled by the circadian timing system that comprises a main circadian clock and numerous secondary clocks in the brain and peripheral tissues. Each secondary circadian clock delivers local temporal cues based on intracellular transcriptional and translational feedback loops that are tightly interconnected to intracellular nutrient-sensing pathways. Genetic impairment of molecular clocks and alteration in the rhythmic synchronizing cues, such as ambient light at night or mistimed meals, lead to circadian disruption that, in turn, negatively impacts metabolic health. Not all circadian clocks are sensitive to the same synchronizing signals. The master clock in the suprachiasmatic nuclei of the hypothalamus is mostly synchronized by ambient light and, to a lesser extent, by behavioral cues coupled to arousal and exercise. Secondary clocks are generally phase-shifted by timed metabolic cues associated with feeding, exercise, and changes in temperature. Furthermore, both the master and secondary clocks are modulated by calorie restriction and high-fat feeding. Taking into account the regularity of daily meals, the duration of eating periods, chronotype, and sex, chrononutritional strategies may be useful for improving the robustness of daily rhythmicity and maintaining or even restoring the appropriate energy balance.
Collapse
Affiliation(s)
- Emma Grosjean
- Institute of Cellular and Integrative Neurosciences, CNRS UPR3212, University of Strasbourg, 67000 Strasbourg, France
| | - Valérie Simonneaux
- Institute of Cellular and Integrative Neurosciences, CNRS UPR3212, University of Strasbourg, 67000 Strasbourg, France
| | - Etienne Challet
- Institute of Cellular and Integrative Neurosciences, CNRS UPR3212, University of Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
11
|
Ishihara A, Courville AB, Chen KY. The Complex Effects of Light on Metabolism in Humans. Nutrients 2023; 15:nu15061391. [PMID: 36986120 PMCID: PMC10056135 DOI: 10.3390/nu15061391] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/15/2023] Open
Abstract
Light is an essential part of many life forms. The natural light–dark cycle has been the dominant stimulus for circadian rhythms throughout human evolution. Artificial light has restructured human activity and provided opportunities to extend the day without reliance on natural day–night cycles. The increase in light exposure at unwanted times or a reduced dynamic range of light between the daytime and nighttime has introduced negative consequences for human health. Light exposure is closely linked to sleep–wake regulation, activity and eating patterns, body temperature, and energy metabolism. Disruptions to these areas due to light are linked to metabolic abnormalities such as an increased risk of obesity and diabetes. Research has revealed that various properties of light influence metabolism. This review will highlight the complex role of light in human physiology, with a specific emphasis on metabolic regulation from the perspective of four main properties of light (intensity, duration, timing of exposure, and wavelength). We also discuss the potential influence of the key circadian hormone melatonin on sleep and metabolic physiology. We explore the relationship between light and metabolism through circadian physiology in various populations to understand the optimal use of light to mitigate short and long-term health consequences.
Collapse
|
12
|
Souissi A, Dergaa I, Romdhani M, Ghram A, Irandoust K, Chamari K, Ben Saad H. Can melatonin reduce the severity of post-COVID-19 syndrome? EXCLI JOURNAL 2023; 22:173-187. [PMID: 36998709 PMCID: PMC10043401 DOI: 10.17179/excli2023-5864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 04/01/2023]
Abstract
This short review aimed at (i) providing an update on the health benefits associated with melatonin supplementation, while (ii) considering future potential research directions concerning melatonin supplementation use relative to Coronavirus disease of 2019 (COVID-19). A narrative review of the literature was undertaken to ascertain the effect of exogenous melatonin administration on humans. Night-time melatonin administration has a positive impact on human physiology and mental health. Indeed, melatonin (i) modulates the circadian components of the sleep-wake cycle; (ii) improves sleep efficiency and mood status; (iii) improves insulin sensitivity; and (iv) reduces inflammatory markers and oxidative stress. Melatonin has also remarkable neuroprotective and cardioprotective effects and may therefore prevent deterioration caused by COVID-19. We suggest that melatonin could be used as a potential therapy in the post-COVID-19 syndrome, and therefore call for action the research community to investigate on the potential use of exogenous melatonin to enhance the quality of life in patients with post-COVID-19 syndrome. See also Figure 1(Fig. 1).
Collapse
Affiliation(s)
- Amine Souissi
- Université de Sousse, Faculté de Médecine de Sousse, Hôpital Farhat HACHED, Laboratoire de Recherche (Insuffisance Cardiaque, LR12SP09), Sousse, Tunisie
- *To whom correspondence should be addressed: Amine Souissi, Université de Sousse, Faculté de Médecine de Sousse, Hôpital Farhat HACHED, Laboratoire de Recherche (Insuffisance Cardiaque, LR12SP09), Sousse, Tunisie, E-mail:
| | - Ismail Dergaa
- Primary Health Care Corporation (PHCC), Doha, P.O. Box 26555, Qatar
| | - Mohamed Romdhani
- Research Unit: Physical Activity, Sport, and Health, UR18JS01, National Observatory of Sport, Tunis, Tunisia
- Motricité-Interactions-Performance, MIP, UR4334, Le Mans Université, Le Mans, France
| | - Amine Ghram
- Université de Sousse, Faculté de Médecine de Sousse, Hôpital Farhat HACHED, Laboratoire de Recherche (Insuffisance Cardiaque, LR12SP09), Sousse, Tunisie
| | - Khadijeh Irandoust
- Department of Sport Sciences, Imam Khomeini International University, Qazvin, Iran
| | - Karim Chamari
- Aspetar, Orthopedic and Sports Medicine Hospital, FIFA Medical Center of Excellence, Doha, Qatar
| | - Helmi Ben Saad
- Université de Sousse, Faculté de Médecine de Sousse, Hôpital Farhat HACHED, Laboratoire de Recherche (Insuffisance Cardiaque, LR12SP09), Sousse, Tunisie
| |
Collapse
|
13
|
Circadian mechanism disruption is associated with dysregulation of inflammatory and immune responses: a systematic review. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2022. [DOI: 10.1186/s43088-022-00290-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractThe circadian rhythms are regulated by the circadian clock which is under the control of suprachiasmatic nucleus of hypothalamus. The central and peripheral clocks on different tissue together synchronize to form circadian system. Factors disrupt the circadian rhythm, such as irregular eating patterns, sleep/wake time, night shift work and temperature. Due to the misalignment of central clock components, it has been recognized as the pathophysiology of lifestyle-related diseases mediated by the inflammation such as diabetes, obesity, neurological disorder and hormonal imbalance. Also we discuss the therapeutic effect of time-restricted feeding over diabetes and obesity caused by miscommunication between central and peripheral clock. The genetic and epigenetic changes involve due to the deregulation of circadian system. The aim of the present review is to discuss the circadian mechanisms that are involved in the complex interaction between host and external factors and its disruption is associated with deregulation of inflammatory and immune responses. Hence, we need to understand the mechanism of functioning of our biological clocks so that it helps us treat health-related problems such as jet lags, sleep disorders due to night-time shift work, obesity and mental disturbances. We hope minimal cost behavioural and lifestyle changes can improve circadian rhythms and presumably provide a better health.
Collapse
|
14
|
The melatonergic agonist agomelatine ameliorates high fat diet-induced obesity in mice through the modulation of the gut microbiome. Biomed Pharmacother 2022; 153:113445. [PMID: 36076560 DOI: 10.1016/j.biopha.2022.113445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 07/11/2022] [Accepted: 07/18/2022] [Indexed: 11/21/2022] Open
|
15
|
Pivonello C, Negri M, Patalano R, Amatrudo F, Montò T, Liccardi A, Graziadio C, Muscogiuri G, Pivonello R, Colao A. The role of melatonin in the molecular mechanisms underlying metaflammation and infections in obesity: A narrative review. Obes Rev 2022; 23:e13390. [PMID: 34861097 PMCID: PMC9285339 DOI: 10.1111/obr.13390] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 12/20/2022]
Abstract
Obesity is a chronic condition whose management is a critical challenge for physicians. The scientific community has increased its focus on the molecular mechanisms involved in obesity etiopathogenesis to better manage patients with obesity and its associated complications. The tight connection between adipose tissue and the immune system has been demonstrated to play a crucial role in inflammation, and melatonin is important for circadian rhythm regulation and metabolic homeostasis, in which it orchestrates several molecular mechanisms involved in obesity and associated inflammation. Melatonin also regulates innate and adaptive immunity; its antioxidant properties are linked to reduced predisposition to infection and weight gain in patients with obesity through the modulation of the immune response, which has a significant beneficial effect on inflammation and, consequently, on the metabolic state. Low melatonin levels have been linked to obesity, and melatonin supplementation can reduce body weight, improve metabolic profile, and ameliorate immune responses and pro-inflammatory stimuli. The role of melatonin in obesity is mainly related to improved oxidative stress signaling, modulation of adipokine secretion, and a switching from white-to-brown adipose tissue phenotype and activity. Moreover, the role of melatonin in obesity modulation by controlling circadian rhythm has recently emerged as a pivotal mechanism for lipid and glucose metabolism dysfunction in adipose, muscle, and liver tissues. Melatonin may also regulate the immune system by acting directly on thymus morphology and activity as well as by modulating oxidative stress and inflammatory states during infections. The tight association between melatonin and immune response regulation is coordinated by Toll-like receptors, which are rhythmically expressed during the day. Their expression may be strongly modulated by melatonin as their signaling is highly inhibited by melatonin. The current review summarizes studies of melatonin-induced mechanisms involved in infection regulation, particularly the modulation of obesity-associated inflammation and systemic complications.
Collapse
Affiliation(s)
- Claudia Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Mariarosaria Negri
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Roberta Patalano
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Feliciana Amatrudo
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Tatiana Montò
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Alessia Liccardi
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Chiara Graziadio
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Giovanna Muscogiuri
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy.,Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy.,UNESCO Chair for Health Education and Sustainable Development, Federico II University, Naples, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy.,UNESCO Chair for Health Education and Sustainable Development, Federico II University, Naples, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy.,Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy.,UNESCO Chair for Health Education and Sustainable Development, Federico II University, Naples, Italy
| |
Collapse
|
16
|
Melatonin ameliorates cardiac remodelling in fructose-induced metabolic syndrome rat model by using genes encoding cardiac potassium ion channels. Mol Biol Rep 2021; 48:5811-5819. [PMID: 34347240 DOI: 10.1007/s11033-021-06526-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/27/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Metabolic syndrome comprises a group of disorders, including cardiac abnormalities. Ventricular arrhythmias observed in metabolic syndrome are due to the impaired ventricular repolarization. This study aims to determine the effects of melatonin on cardiac ventricular repolarization in metabolic syndrome rat model. METHODS AND RESULTS Sprague-Dawley rats were divided into control (n = 8), melatonin (n = 8), metabolic syndrome (n = 8) and metabolic syndrome + melatonin (n = 8) groups. Fructose (200 g/lt/day) was added into the drinking water during 8 weeks of rats to induce metabolic syndrome model. In the last two weeks, melatonin (20 mg/kg/day) was administered via oral gavage. Blood pressure measurements and ECG recordings were taken at three different times. Blood and left ventricular tissue samples were harvested and the KCNQ1,3 and KCNH2 gene expressions were analysed by qRT-PCR method. We observed insulin resistance, hyperglycemia, dyslipidemia and higher systolic blood pressure in metabolic syndrome group (p < 0.01, for all). Prolonged QT interval was observed in metabolic syndrome group (p < 0.001). The expression levels of the KCNQ genes encoding the Kv7 channel was significantly reduced, however KCNH2 gene which encodes Kv11.1 channel was increased in metabolic syndrome group compared to control group (p < 0.05, p < 0.001, respectively). Melatonin significantly normalised the prolongation on QT interval in metabolic syndrome group (p < 0.001) and the expressions of the KCNQ (p < 0.002) and KCNH2 genes (p = 0.003). CONCLUSIONS The present study revealed that melatonin had ameliorative effects on ventricular repolarization by improving the prolonged QT duration in rats with metabolic syndrome and this effect was generated by the KCNQ and KCNH2 gene families.
Collapse
|
17
|
Sides MB, Johnston SL, Sirek A, Lee PH, Blue RS, Antonsen EL, Basner M, Douglas GL, Epstein A, Flynn-Evans EE, Gallagher MB, Hayes J, Lee SMC, Lockley SW, Monseur B, Nelson NG, Sargsyan A, Smith SM, Stenger MB, Stepanek J, Zwart SR. Bellagio II Report: Terrestrial Applications of Space Medicine Research. Aerosp Med Hum Perform 2021; 92:650-669. [PMID: 34503618 DOI: 10.3357/amhp.5843.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
AbstractINTRODUCTION: For over 50 yr, investigators have studied the physiological adaptations of the human system during short- and long-duration spaceflight exposures. Much of the knowledge gained in developing health countermeasures for astronauts onboard the International Space Station demonstrate terrestrial applications. To date, a systematic process for translating these space applications to terrestrial human health has yet to be defined.METHODS: In the summer of 2017, a team of 38 international scientists launched the Bellagio ll Summit Initiative. The goals of the Summit were: 1) To identify space medicine findings and countermeasures with highest probability for future terrestrial applications; and 2) To develop a roadmap for translation of these countermeasures to future terrestrial application. The team reviewed public domain literature, NASA databases, and evidence books within the framework of the five-stage National Institutes of Health (NIH) translation science model, and the NASA two-stage translation model. Teams then analyzed and discussed interdisciplinary findings to determine the most significant evidence-based countermeasures sufficiently developed for terrestrial application.RESULTS: Teams identified published human spaceflight research and applied translational science models to define mature products for terrestrial clinical practice.CONCLUSIONS: The Bellagio ll Summit identified a snapshot of space medicine research and mature science with the highest probability of translation and developed a Roadmap of terrestrial application from space medicine-derived countermeasures. These evidence-based findings can provide guidance regarding the terrestrial applications of best practices, countermeasures, and clinical protocols currently used in spaceflight.Sides MB, Johnston SL III, Sirek A, Lee PH, Blue RS, Antonsen EL, Basner M, Douglas GL, Epstein A, Flynn-Evans EE, Gallagher MB, Hayes J, Lee SMC, Lockley SW, Monseur B, Nelson NG, Sargsyan A, Smith SM, Stenger MB, Stepanek J, Zwart SR; Bellagio II Team. Bellagio II report: terrestrial applications of space medicine research. Aerosp Med Hum Perform. 2021; 92(8):650669.
Collapse
|
18
|
Cherngwelling R, Pengrattanachot N, Swe MT, Thongnak L, Promsan S, Phengpol N, Sutthasupha P, Lungkaphin A. Agomelatine protects against obesity-induced renal injury by inhibiting endoplasmic reticulum stress/apoptosis pathway in rats. Toxicol Appl Pharmacol 2021; 425:115601. [PMID: 34081941 DOI: 10.1016/j.taap.2021.115601] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/18/2021] [Accepted: 05/28/2021] [Indexed: 12/18/2022]
Abstract
Obesity is recognized as a risk for the development of chronic kidney disease. Excessive fat accumulation in obesity is associated with the overproduction of reactive oxygen species with the underproduction of antioxidant mechanisms generating oxidative stress together with chronic low-grade inflammation which subsequently leads to the development of several obesity-related complications. It has been suggested that the abnormal lipid accumulation can induce endoplasmic reticulum (ER) stress and cellular apoptosis in several tissue types. Agomelatine is a relatively new antidepressant which is a synthetic agonist of melatonin. Previous study reported the antioxidant and anti-inflammatory effects of agomelatine. In this study, we investigated the therapeutic effects of agomelatine in obesity-related renal injury. Male Wistar rats were fed with normal diet or high-fat diet (HF) for 16 weeks. After that, vehicle or agomelatine or vildagliptin was orally administered to HF rats for 4 weeks. Our results indicated that HF rats demonstrated insulin resistance which was accompanied by an impairment of renal function and renal organic anion transporter 3 (Oat3) function as well as renal oxidative stress, ER stress, and apoptosis. Interestingly, agomelatine treatment not only improved the metabolic parameters, renal function and renal Oat3 function but also attenuated renal oxidative stress, ER stress and subsequent apoptosis. Therefore, agomelatine exerted renoprotective effects in obese insulin-resistant condition. These results suggested that agomelatine could be used as a drug to improve metabolic disturbance and prevent kidney dysfunction in obese condition.
Collapse
Affiliation(s)
- Rada Cherngwelling
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Myat Theingi Swe
- Department of Physiology, University of Medicine 2, Yangon, Myanmar
| | - Laongdao Thongnak
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sasivimon Promsan
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nichakorn Phengpol
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Prempree Sutthasupha
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Anusorn Lungkaphin
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
19
|
Templeman I, Smith HA, Walhin JP, Middleton B, Gonzalez JT, Karagounis LG, Johnston JD, Betts JA. Unacylated ghrelin, leptin, and appetite display diurnal rhythmicity in lean adults. J Appl Physiol (1985) 2021; 130:1534-1543. [PMID: 33703941 DOI: 10.1152/japplphysiol.00920.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Constant routine and forced desynchrony protocols typically remove the effects of behavioral/environmental cues to examine endogenous circadian rhythms, yet this may not reflect rhythms of appetite regulation in the real world. It is therefore important to understand these rhythms within the same subjects under controlled diurnal conditions of light, sleep, and feeding. Ten healthy adults (9 M/1 F, means ±SD: age, 30 ± 10 yr; body mass index, 24.1 ± 2.7 kg·m-2) rested supine in the laboratory for 37 h. All data were collected during the final 24 h of this period (i.e., 0800-0800 h). Participants were fed hourly isocaloric liquid meal replacements alongside appetite assessments during waking before a sleep opportunity from 2200 to 0700 h. Hourly blood samples were collected throughout the 24-h period. Dim light melatonin onset occurred at 2318 ± 46 min. A diurnal rhythm in mean plasma unacylated ghrelin concentration was identified (P = 0.04), with the acrophase occurring shortly after waking (0819), falling to a nadir in the evening with a relative amplitude of 9%. Plasma leptin concentration also exhibited a diurnal rhythm (P < 0.01), with the acrophase occurring shortly after lights-out (0032 h) and the lowest concentrations at midday. The amplitude for this rhythm was 25%. Diurnal rhythms were established in all dimensions of appetite except for sweet preference (P = 0.29), with both hunger (2103 h) and prospective food consumption (1955 h) reaching their peak in the evening before falling to their nadir shortly after waking. Under controlled diurnal conditions, simultaneous measurement of leptin, unacylated ghrelin, and subjective appetite over a 24-h period revealed rhythmicity in appetite regulation in lean, healthy humans.NEW & NOTEWORTHY Simultaneous assessment of subjective appetite, unacylated ghrelin, and leptin was carried out over a continuous 37-h protocol for the first time under conditions of controlled light, sleep, and feeding in healthy, lean adults. Rhythms were observed in unacylated ghrelin, leptin, and components of subjective appetite, such as hunger, prospective consumption, and fullness. Concurrent measurement of rhythms in these variables is important to fully understand the temporal relationships between components of appetite as well as the influence of diurnal factors such as sleep, light, and feeding.
Collapse
Affiliation(s)
- Iain Templeman
- Department for Health, Centre for Nutrition, Exercise and Metabolism, University of Bath, Bath, United Kingdom
| | - Harry A Smith
- Department for Health, Centre for Nutrition, Exercise and Metabolism, University of Bath, Bath, United Kingdom
| | - Jean-Philippe Walhin
- Department for Health, Centre for Nutrition, Exercise and Metabolism, University of Bath, Bath, United Kingdom
| | - Benita Middleton
- Section of Chronobiology, Faculty of Health and Medical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Javier T Gonzalez
- Department for Health, Centre for Nutrition, Exercise and Metabolism, University of Bath, Bath, United Kingdom
| | - Leonidas G Karagounis
- Nestlé Health Science, Translation Research, Vevey, Switzerland.,Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Jonathan D Johnston
- Section of Chronobiology, Faculty of Health and Medical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - James A Betts
- Department for Health, Centre for Nutrition, Exercise and Metabolism, University of Bath, Bath, United Kingdom
| |
Collapse
|
20
|
Arellanes-Licea EDC, Pérez-Mendoza M, Carmona-Castro A, Díaz-Muñoz M, Miranda-Anaya M. Obese Neotomodon alstoni mice exhibit sexual dimorphism in the daily profile of circulating melatonin and clock proteins PER1 and BMAL1 in the hypothalamus and peripheral oscillators. Chronobiol Int 2021; 38:584-597. [PMID: 33393371 DOI: 10.1080/07420528.2020.1860999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022]
Abstract
Obesity is a global health threat and a risk factor for several metabolic conditions. Though circadian dysfunction has been considered among the multiple causes of obesity, little work has been done to explore the relationship between obesity, circadian dysfunction, and sexual dimorphism. The Neotomodon alstoni mouse is a suitable model for such research. This study employed N. alstoni mice in a chronobiological analysis to determine whether there is circadian desynchronization of relative PER1 and BMAL1 protein levels in the hypothalamus, liver, visceral white adipose tissue, kidney, and heart. It also compared differences between sexes and lean and obese N. alstoni adult mice, by recording behavior and daily circulating serum melatonin as markers of circadian output. We found that obese mice display reduced locomotor activity. Additionally, Cosinor analyses of the relative expression of PER1 and BMAL1 show differences between lean and obese mice in a sex-linked manner. The PER1 24 h rhythm was absent in all tissues of obese males and significant in the tissues of obese females. The BMAL1 24 h rhythm also was significant in most of the tissues tested in lean males, whereas it was significant and shifted the acrophase (peak time of rhythm) in most of the tissues in obese females. Both lean male and female mice showed a rhythmic 24 h pattern of circulating serum melatonin. This daily profile was not only absent in obese mice of both sexes but showed sexual dimorphism. Obese male mice showed lower circulating levels of melatonin compared to lean male mice, but they were higher in obese females compared to lean females. Our results suggest that obesity in N. alstoni is associated with an internal circadian desynchronization in a sex-dependent manner. Overall, this study reinforces the need for further research on the neuroendocrinology of obesity and circadian rhythms using this biological model.
Collapse
Affiliation(s)
- Elvira Del Carmen Arellanes-Licea
- Unidad Multidisciplinaria de Docencia e Investigación, Facultad de Ciencias, Universidad Nacional Autónoma de México, Querétaro, México
| | - Moisés Pérez-Mendoza
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Querétaro, México
| | - Agustín Carmona-Castro
- Departamento de Biología Celular, Facultad de Ciencias, Ciudad Universitaria, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Mauricio Díaz-Muñoz
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Manuel Miranda-Anaya
- Unidad Multidisciplinaria de Docencia e Investigación, Facultad de Ciencias, Universidad Nacional Autónoma de México, Querétaro, México
| |
Collapse
|
21
|
Affiliation(s)
- David J Kennaway
- Robinson Research Institute and Adelaide School of Medicine, University of Adelaide, Adelaide, Australia
| |
Collapse
|
22
|
Yuan RK, Zitting K, Wang W, Buxton OM, Williams JS, Duffy JF, Czeisler CA. Fasting blood triglycerides vary with circadian phase in both young and older people. Physiol Rep 2020; 8:e14453. [PMID: 32519460 PMCID: PMC7283043 DOI: 10.14814/phy2.14453] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/31/2022] Open
Abstract
Daily rhythms in several physiological processes are important for cardiometabolic health. The lipid panel is used clinically to assess cardiovascular disease risk, but previous attempts to demonstrate circadian variation in lipids have failed to uncouple the endogenous circadian rhythm from the effects of meals and wake duration. Changes in basal lipid levels and dampening of circadian rhythms have been reported with aging, but it is unknown whether aging is also associated with changes in the rhythmic variation of lipids. We measured fasting lipid panels (triglycerides, total cholesterol, high-density lipoprotein, and low-density lipoprotein) in blood at wake time in 21 healthy adults using a specialized laboratory protocol that uncouples sleep-wake and activity-related effects from the endogenous circadian rhythm. Young and older adults exhibited endogenous circadian variations in fasting triglycerides, with both groups peaking in the early biological morning. Young adults also exhibited significant circadian rhythmicity in total cholesterol and low-density lipoprotein, while older adults did not exhibit circadian rhythmicity in any other lipids. These results reveal that triglyceride metabolism may be regulated by the central circadian pacemaker. Moreover, our findings may have clinical implications in assessing cardiovascular risk in shift workers and younger adults, since routine measurement of morning/fasting lipids may not fully and reliably assess triglyceride- and other lipid-related biomarkers of cardiovascular disease risk in these groups.
Collapse
Affiliation(s)
- Robin K. Yuan
- Division of Sleep and Circadian DisordersDepartments of Medicine and NeurologyBrigham and Women's HospitalBostonMAUSA
- Division of Sleep MedicineHarvard Medical SchoolBostonMAUSA
| | - Kirsi‐Marja Zitting
- Division of Sleep and Circadian DisordersDepartments of Medicine and NeurologyBrigham and Women's HospitalBostonMAUSA
- Division of Sleep MedicineHarvard Medical SchoolBostonMAUSA
| | - Wei Wang
- Division of Sleep and Circadian DisordersDepartments of Medicine and NeurologyBrigham and Women's HospitalBostonMAUSA
- Division of Sleep MedicineHarvard Medical SchoolBostonMAUSA
| | - Orfeu M. Buxton
- Division of Sleep and Circadian DisordersDepartments of Medicine and NeurologyBrigham and Women's HospitalBostonMAUSA
- Division of Sleep MedicineHarvard Medical SchoolBostonMAUSA
- Department of Biobehavioral HealthPennsylvania State UniversityUniversity ParkPAUSA
| | - Jonathan S. Williams
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMAUSA
| | - Jeanne F. Duffy
- Division of Sleep and Circadian DisordersDepartments of Medicine and NeurologyBrigham and Women's HospitalBostonMAUSA
- Division of Sleep MedicineHarvard Medical SchoolBostonMAUSA
| | - Charles A. Czeisler
- Division of Sleep and Circadian DisordersDepartments of Medicine and NeurologyBrigham and Women's HospitalBostonMAUSA
- Division of Sleep MedicineHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
23
|
Hernández Morante JJ, Díaz Soler I, Muñoz JSG, Sánchez HP, Barberá Ortega MDC, Martínez CM, Morillas Ruiz JM. Moderate Weight Loss Modifies Leptin and Ghrelin Synthesis Rhythms but Not the Subjective Sensations of Appetite in Obesity Patients. Nutrients 2020; 12:E916. [PMID: 32230732 PMCID: PMC7230904 DOI: 10.3390/nu12040916] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/21/2020] [Accepted: 03/25/2020] [Indexed: 12/13/2022] Open
Abstract
Obesity is characterized by a resistance to appetite-regulating hormones, leading to a misalignment between the physiological signals and the perceived hunger/satiety signal. A disruption of the synthesis rhythm may explain this situation. The aim of this study was to evaluate the effect of dietary-induced weight loss on the daily rhythms of leptin and ghrelin and its influence on the daily variability of the appetite sensations of patients with obesity. Twenty subjects with obesity underwent a hypocaloric dietary intervention for 12 weeks. Plasma leptin and ghrelin were analyzed at baseline and at the end of the intervention and in 13 normal-weight controls. Appetite ratings were analyzed. Weight loss decreased leptin synthesis (pauc < 0.001) but not the rhythm characteristics, except the mean variability value (pmesor = 0.020). By contrast, the mean ghrelin level increased after weight loss. The rhythm characteristics were also modified until a rhythm similar to the normal-weight subjects was reached. The amount of variability of leptin and ghrelin was correlated with the effectiveness of the dietary intervention (p < 0.020 and p < 0.001, respectively). Losing weight partially restores the daily rhythms of leptin and modifies the ghrelin rhythms, but appetite sensations are barely modified, thus confirming that these hormones cannot exercise their physiological function properly.
Collapse
Affiliation(s)
- Juan José Hernández Morante
- Eating Disorders Research Unit, Catholic University of Murcia (UCAM), 30107 Murcia, Spain; (I.D.S.); (M.d.C.B.O.)
| | - Inmaculada Díaz Soler
- Eating Disorders Research Unit, Catholic University of Murcia (UCAM), 30107 Murcia, Spain; (I.D.S.); (M.d.C.B.O.)
| | | | - Horacio Pérez Sánchez
- Bioinformatics and High Performance Computing Research Group (BIO-HPC), Computer Engineering Department, Catholic University of Murcia (UCAM), 30107 Murcia, Spain;
| | - Mª del Carmen Barberá Ortega
- Eating Disorders Research Unit, Catholic University of Murcia (UCAM), 30107 Murcia, Spain; (I.D.S.); (M.d.C.B.O.)
| | | | - Juana Mª Morillas Ruiz
- Food Technology & Nutrition Dept., Catholic University of Murcia (UCAM), 30107 Murcia, Spain;
| |
Collapse
|
24
|
Sinturel F, Petrenko V, Dibner C. Circadian Clocks Make Metabolism Run. J Mol Biol 2020; 432:3680-3699. [PMID: 31996313 DOI: 10.1016/j.jmb.2020.01.018] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/16/2020] [Accepted: 01/16/2020] [Indexed: 12/12/2022]
Abstract
Most organisms adapt to the 24-h cycle of the Earth's rotation by anticipating the time of the day through light-dark cycles. The internal time-keeping system of the circadian clocks has been developed to ensure this anticipation. The circadian system governs the rhythmicity of nearly all physiological and behavioral processes in mammals. In this review, we summarize current knowledge stemming from rodent and human studies on the tight interconnection between the circadian system and metabolism in the body. In particular, we highlight recent advances emphasizing the roles of the peripheral clocks located in the metabolic organs in regulating glucose, lipid, and protein homeostasis at the organismal and cellular levels. Experimental disruption of circadian system in rodents is associated with various metabolic disturbance phenotypes. Similarly, perturbation of the clockwork in humans is linked to the development of metabolic diseases. We discuss recent studies that reveal roles of the circadian system in the temporal coordination of metabolism under physiological conditions and in the development of human pathologies.
Collapse
Affiliation(s)
- Flore Sinturel
- Department of Medicine, Division of Endocrinology, Diabetes, Hypertension and Nutrition, Faculty of Medicine, University of Geneva, Rue Michel-Servet, 1, CH-1211, Geneva, 14, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, Geneva, Switzerland.
| | - Volodymyr Petrenko
- Department of Medicine, Division of Endocrinology, Diabetes, Hypertension and Nutrition, Faculty of Medicine, University of Geneva, Rue Michel-Servet, 1, CH-1211, Geneva, 14, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, Geneva, Switzerland
| | - Charna Dibner
- Department of Medicine, Division of Endocrinology, Diabetes, Hypertension and Nutrition, Faculty of Medicine, University of Geneva, Rue Michel-Servet, 1, CH-1211, Geneva, 14, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, Geneva, Switzerland.
| |
Collapse
|
25
|
Adverse Events Associated with Melatonin for the Treatment of Primary or Secondary Sleep Disorders: A Systematic Review. CNS Drugs 2019; 33:1167-1186. [PMID: 31722088 DOI: 10.1007/s40263-019-00680-w] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Melatonin is widely available either on prescription for the treatment of sleep disorders or as an over-the-counter dietary supplement. Melatonin has also recently been licensed in the UK for the short-term treatment of jetlag. Little is known about the potential for adverse events (AEs), in particular AEs resulting from long-term use. Concern has been raised over the possible risks of exposure in certain populations including pre-adolescent children and patients with epilepsy or asthma. OBJECTIVES The aim of this systematic review was to assess the evidence for AEs associated with short-term and longer-term melatonin treatment for sleep disorders. METHODS A literature search of the PubMed/Medline database and Google Scholar was conducted to identify randomised, placebo-controlled trials (RCTs) of exogenous melatonin administered for primary or secondary sleep disorders. Studies were included if they reported on both the types and frequencies of AEs. Studies of pre-term infants, studies of < 1 week in duration or involving single doses of melatonin and studies in languages other than English were excluded. Findings from open-label studies that raised concerns relating to AE reports in patients were also examined. Studies were assessed for quality of reporting against the Consolidated Standards of Reporting Trials (CONSORT) checklist and for risk of bias against the Cochrane Collaboration risk-of-bias criteria. RESULTS 37 RCTs met criteria for inclusion. Daily melatonin doses ranged from 0.15 mg to 12 mg. Subjects were monitored for up to 29 weeks, but most studies were of much shorter duration (4 weeks or less). The most frequently reported AEs were daytime sleepiness (1.66%), headache (0.74%), other sleep-related AEs (0.74%), dizziness (0.74%) and hypothermia (0.62%). Very few AEs considered to be serious or of clinical significance were reported. These included agitation, fatigue, mood swings, nightmares, skin irritation and palpitations. Most AEs either resolved spontaneously within a few days with no adjustment in melatonin, or immediately upon withdrawal of treatment. Melatonin was generally regarded as safe and well tolerated. Many studies predated publication of the CONSORT checklist and consequently did not conform closely to the guidelines. Similarly, only eight studies were judged 'good' overall with respect to the Cochrane risk-of-bias criteria. Of the remaining papers, 16 were considered 'fair' and 13 'poor' but publication of almost half of the papers preceded that of the earliest version of the guidelines. CONCLUSION Few, generally mild to moderate, AEs were associated with exogenous melatonin. No AEs that were life threatening or of major clinical significance were identified. The scarcity of evidence from long-term RCTs, however, limits the conclusions regarding the safety of continuous melatonin therapy over extended periods. There are insufficient robust data to allow a meaningful appraisal of concerns that melatonin may result in more clinically significant adverse effects in potentially at-risk populations. Future studies should be designed to comply with appropriate quality standards for RCTs, which most past studies have not.
Collapse
|
26
|
Dumpala S, Zele AJ, Feigl B. Outer Retinal Structure and Function Deficits Contribute to Circadian Disruption in Patients With Type 2 Diabetes. Invest Ophthalmol Vis Sci 2019; 60:1870-1878. [PMID: 31042793 DOI: 10.1167/iovs.18-26297] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Light transmitted by retinal photoreceptors provides the input for circadian photoentrainment. In diabetes, there is a high prevalence of circadian and sleep disruption but the underlying causes are not well understood. Patients with diabetes can exhibit dysfunctional photoreceptors but their role in circadian health is not known. Here we quantify photoreceptor function and contributions to circadian health and sleep in patients with diabetes without diabetic retinopathy and healthy controls. Methods Rod, cone, and melanopsin function was derived using chromatic pupillometry in 47 participants including 23 patients with type 2 diabetes and 24 age-matched healthy controls after an ophthalmic examination including retinal thickness assessment using optical coherence tomography. Circadian health was determined using dim light melatonin onset (DLMO) and sleep questionnaires; light exposure was measured using actigraphy. Results Compared with the control group, the patients with diabetes had a significantly earlier DLMO (1 hour) (P = 0.008), higher subjective sleep scores (P < 0.05), a reduction in pupil constriction amplitude for red stimuli (P = 0.039) and for the early postillumination pupil response (PIPR) for blue (P = 0.024) stimuli. There were no between-group differences in the light exposure pattern, activity levels, and intrinsic melanopsin-mediated PIPR amplitude (P > 0.05). A significant correlation was evident between outer retinal thickness and DLMO (r = -0.65, P = 0.03) and the pupil constriction amplitude (r = 0.63, P = 0.03); patients with thinner retina had earlier DLMO and lower pupil amplitudes. Conclusions We infer that the observed changes in circadian function in patients with no diabetic retinopathy are due to structural and functional outer retinal rod photoreceptor deficits at early stage of diabetic eye disease.
Collapse
Affiliation(s)
- Sunila Dumpala
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,School of Optometry and Vision Sciences, Queensland University of Technology, Brisbane, Australia
| | - Andrew J Zele
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,School of Optometry and Vision Sciences, Queensland University of Technology, Brisbane, Australia
| | - Beatrix Feigl
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia.,Queensland Eye Institute, Brisbane, Australia
| |
Collapse
|
27
|
Christou S, Wehrens SMT, Isherwood C, Möller-Levet CS, Wu H, Revell VL, Bucca G, Skene DJ, Laing EE, Archer SN, Johnston JD. Circadian regulation in human white adipose tissue revealed by transcriptome and metabolic network analysis. Sci Rep 2019; 9:2641. [PMID: 30804433 PMCID: PMC6389935 DOI: 10.1038/s41598-019-39668-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 01/15/2019] [Indexed: 01/28/2023] Open
Abstract
Studying circadian rhythms in most human tissues is hampered by difficulty in collecting serial samples. Here we reveal circadian rhythms in the transcriptome and metabolic pathways of human white adipose tissue. Subcutaneous adipose tissue was taken from seven healthy males under highly controlled 'constant routine' conditions. Five biopsies per participant were taken at six-hourly intervals for microarray analysis and in silico integrative metabolic modelling. We identified 837 transcripts exhibiting circadian expression profiles (2% of 41619 transcript targeting probes on the array), with clear separation of transcripts peaking in the morning (258 probes) and evening (579 probes). There was only partial overlap of our rhythmic transcripts with published animal adipose and human blood transcriptome data. Morning-peaking transcripts associated with regulation of gene expression, nitrogen compound metabolism, and nucleic acid biology; evening-peaking transcripts associated with organic acid metabolism, cofactor metabolism and redox activity. In silico pathway analysis further indicated circadian regulation of lipid and nucleic acid metabolism; it also predicted circadian variation in key metabolic pathways such as the citric acid cycle and branched chain amino acid degradation. In summary, in vivo circadian rhythms exist in multiple adipose metabolic pathways, including those involved in lipid metabolism, and core aspects of cellular biochemistry.
Collapse
Affiliation(s)
- Skevoulla Christou
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Sophie M T Wehrens
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Cheryl Isherwood
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK.,Department of Medicine, Brigham and Women's Hospital, Boston, USA
| | - Carla S Möller-Levet
- Bioinformatics Facility, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Huihai Wu
- Bioinformatics Facility, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Victoria L Revell
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Giselda Bucca
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK.,School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Debra J Skene
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Emma E Laing
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Simon N Archer
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Jonathan D Johnston
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK.
| |
Collapse
|
28
|
Agbaria S, Haim A, Fares F, Zubidat AE. Epigenetic modification in 4T1 mouse breast cancer model by artificial light at night and melatonin - the role of DNA-methyltransferase. Chronobiol Int 2019; 36:629-643. [PMID: 30746962 DOI: 10.1080/07420528.2019.1574265] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Currently, one of the most disputed hypotheses regarding breast cancer (BC) development is exposure to short wavelength artificial light at night (ALAN) as multiple studies suggest a possible link between them. This link is suggested to be mediated by nocturnal melatonin suppression that plays an integral role in circadian regulations including cell division. The objective of the research was to evaluate effects of 1 × 30 min/midnight ALAN (134 µ Wcm-2, 460 nm) with or without nocturnal melatonin supplement on tumor development and epigenetic responses in 4T1 tumor-bearing BALB/c mice. Mice were monitored for body mass (Wb) and tumor volume for 3 weeks and thereafter urine samples were collected at regular intervals for determining daily rhythms of 6-sulfatoxymelatonin (6-SMT). Finally, mice were sacrificed and the tumor, lungs, liver, and spleen were excised for analyzing the total activity of DNA methyltransferases (DNMT) and global DNA methylation (GDM) levels. Mice exposed to ALAN significantly reduced 6-SMT levels and increased Wb, tumor volume, and lung metastasis compared with controls. These effects were diminished by melatonin. The DNMT activity and GDM levels showed tissue-specific response. The enzymatic activity and GDM levels were lower in tumor and liver and higher in spleen and lungs under ALAN compared with controls. Our results suggest that ALAN disrupts the melatonin rhythm and potentially leading to increased BC burden by affecting DNMT activity and GDM levels. These data may also be applicable to early detection and management of BC by monitoring melatonin and GDM levels as early biomarker of ALAN circadian disruption.
Collapse
Affiliation(s)
- Sahar Agbaria
- a Department of Human Biology , University of Haifa , Haifa , Israel
| | - Abraham Haim
- b The Israeli Center for Interdisciplinary Research in Chronobiology , University of Haifa , Haifa , Israel
| | - Fuad Fares
- a Department of Human Biology , University of Haifa , Haifa , Israel.,c Department of Molecular Genetics , Carmel Medical Center , Haifa , Israel
| | - Abed E Zubidat
- b The Israeli Center for Interdisciplinary Research in Chronobiology , University of Haifa , Haifa , Israel
| |
Collapse
|
29
|
Abstract
The epidemic of Type 2 diabetes mellitus necessitates development of novel therapeutic and preventative strategies to attenuate expansion of this debilitating disease. Evidence links the circadian system to various aspects of diabetes pathophysiology and treatment. The aim of this review will be to outline the rationale for therapeutic targeting of the circadian system in the treatment and prevention of Type 2 diabetes mellitus and consequent metabolic comorbidities.
Collapse
Affiliation(s)
- Naureen Javeed
- Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota
| | - Aleksey V Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota.,Department of Medicine, Division of Endocrinology, Metabolism, Diabetes, and Nutrition, Mayo Clinic School of Medicine, Mayo Clinic , Rochester, Minnesota
| |
Collapse
|
30
|
Oishi K, Hashimoto C. Short-term time-restricted feeding during the resting phase is sufficient to induce leptin resistance that contributes to development of obesity and metabolic disorders in mice. Chronobiol Int 2018; 35:1576-1594. [PMID: 30084652 DOI: 10.1080/07420528.2018.1496927] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Feeding at unusual times of the day is thought to be associated with obesity and metabolic disorders in both experimental animals and humans. We previously reported that time-imposed feeding during the sleep phase (daytime feeding, DF) induces obesity and metabolic disorders compared with mice fed only during the active phase (nighttime feeding, NF). The present study aimed to determine whether leptin resistance is caused by DF, and whether it is involved in the underlying mechanisms of DF-induced obesity in mice, since leptin plays an essential role in regulating energy expenditure and adiposity in addition to food intake. We compared leptin sensitivity by evaluating the effects of exogenous injected leptin on food intake and body weight in wild-type C57BL/6J mice under NF and DF. The mice were fed with a high-fat high-sucrose diet throughout the study. To determine whether leptin resistance is a cause or a result of DF-induced obesity with metabolic disorders, we restricted the feeding times of leptin resistant db/db mice. We also examined leptin sensitivity in leptin deficient ob/ob mice under NF and DF to elucidate the underlying mechanisms of DF-induced leptin resistance. C57BL/6J mice under DF gained more weight and adiposity compared with mice under NF, and developed hyperleptinemia and hypothermia. We found that six days of DF abolished exogenous leptin-induced hypophagia and reduction in body weight in mice. We also found that the leptin injection significantly suppressed the mRNA expression of lipogenic genes in the liver of NF, but not in DF mice, suggesting that short-term DF was sufficient to induce metabolic leptin resistance. The DF-induced increases in body weight gain, food efficiency, adipose tissue mass, lipogenic gene expression in metabolic tissues, and hepatic lipid accumulation were abolished in db/db mice, suggesting that the leptin resistance is a cause of DF-induced metabolic disorders. DF resulted in deep hypothermia in db/db, as well as in wild-type mice, suggesting that a decrease in energy expenditure was not the main cause of DF-induced obesity. Exogenous leptin reduced the body weight of ob/ob mice under both NF and DF, and the effect was significantly higher in DF- than in NF-ob/ob mice. Therefore, the development of DF-induced leptin resistance requires endogenous leptin, and central leptin sensitivity fluctuates in a circadian manner. The present findings suggest that leptin resistance is responsible for DF-induced obesity and metabolic disorders, and that the circadian fluctuation of central leptin sensitivity might be involved in leptin resistance induced by DF, although further studies are needed to elucidate the mechanisms of metabolic disorders that depend on the time of feeding. Abbreviations: AMPK, adenosine monophosphate-activated protein kinase; ANOVA, analysis of variance; DF, daytime feeding; FFA, free fatty acid; HOMA-IR, homeostasis model assessment of insulin resistance; NEAT, non-exercise activity thermogenesis; NF, nighttime feeding; PI3, phosphatidylinositol 3; RF, restricted feeding; RW, running-wheel; SCN, suprachiasmatic nucleus; SEM, standard error of the mean; STAT3, signal transducer and activator of transcription 3; T-Cho, total cholesterol; TG, triglyceride; WAT, white adipose tissues.
Collapse
Affiliation(s)
- Katsutaka Oishi
- a Biological Clock Research Group, Biomedical Research Institute , National Institute of Advanced Industrial Science and Technology (AIST) , Tsukuba , Ibaraki , Japan.,b Department of Applied Biological Science, Graduate School of Science and Technology , Tokyo University of Science , Noda , Chiba , Japan.,c Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences , The University of Tokyo , Kashiwa , Chiba , Japan
| | - Chiaki Hashimoto
- a Biological Clock Research Group, Biomedical Research Institute , National Institute of Advanced Industrial Science and Technology (AIST) , Tsukuba , Ibaraki , Japan.,b Department of Applied Biological Science, Graduate School of Science and Technology , Tokyo University of Science , Noda , Chiba , Japan
| |
Collapse
|
31
|
Jiki Z, Lecour S, Nduhirabandi F. Cardiovascular Benefits of Dietary Melatonin: A Myth or a Reality? Front Physiol 2018; 9:528. [PMID: 29867569 PMCID: PMC5967231 DOI: 10.3389/fphys.2018.00528] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/24/2018] [Indexed: 12/14/2022] Open
Abstract
The role of the diet as well as the impact of the dietary habits on human health and disease is well established. Apart from its sleep regulatory effect, the indoleamine melatonin is a well-established antioxidant molecule with multiple health benefits. Convincing evidence supports the presence of melatonin in plants and foods with the intake of such foods affecting circulating melatonin levels in humans. While numerous actions of both endogenous melatonin and melatonin supplementation are well described, little is known about the influence of the dietary melatonin intake on human health. In the present review, evidence for the cardiovascular health benefits of melatonin supplementation and dietary melatonin is discussed. Current knowledge on the biological significance as well as the underlying physiological mechanism of action of the dietary melatonin is also summarized. Whether dietary melatonin constitutes an alternative preventive treatment for cardiovascular disease is addressed.
Collapse
Affiliation(s)
- Zukiswa Jiki
- Cardioprotection Group, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Cardioprotection Group, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frederic Nduhirabandi
- Cardioprotection Group, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
32
|
Chu LW, John EM, Yang B, Kurian AW, Zia Y, Yu K, Ingles SA, Stanczyk FZ, Hsing AW. Measuring serum melatonin in postmenopausal women: Implications for epidemiologic studies and breast cancer studies. PLoS One 2018; 13:e0195666. [PMID: 29641614 PMCID: PMC5895067 DOI: 10.1371/journal.pone.0195666] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/27/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Circulating melatonin is a good candidate biomarker for studies of circadian rhythms and circadian disruption. However, epidemiologic studies on circulating melatonin are limited because melatonin is secreted at night, yet most epidemiologic studies collect blood during the day when melatonin levels are very low, and assays are lacking that are ultrasensitive to detect low levels of melatonin reliably. OBJECTIVE To assess the performance of a refined radioimmunoassay in measuring morning melatonin among women. METHODS We used morning serum samples from 47 postmenopausal women ages 48-80 years without a history of breast cancer who participated in the San Francisco Bay Area Breast Cancer Study, including 19 women who had duplicate measurements. The coefficient of variation (CV) and intraclass coefficient (ICC) were estimated using the random effect model. RESULTS Reproducibility for the assay was satisfactory, with a CV of 11.2% and an ICC of 98.9%; correlation between the replicate samples was also high (R = 0.96). In the 47 women, serum melatonin levels ranged from 0.6 to 62.6 pg/ml, with a median of 7.0 pg/ml. CONCLUSION Our results suggest that it is possible to reliably measure melatonin in postmenopausal women in morning serum samples in large epidemiologic studies to evaluate the role of melatonin in cancer etiology or prognosis.
Collapse
Affiliation(s)
- Lisa W. Chu
- Cancer Prevention Institute of California, Fremont, California, United States of America
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Esther M. John
- Cancer Prevention Institute of California, Fremont, California, United States of America
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Health Research and Policy (Epidemiology), Stanford University School of Medicine, Stanford, California, United States of America
| | - Baiyu Yang
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Allison W. Kurian
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Health Research and Policy (Epidemiology), Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yasaman Zia
- Cancer Prevention Institute of California, Fremont, California, United States of America
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sue A. Ingles
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Frank Z. Stanczyk
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Ann W. Hsing
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Health Research and Policy (Epidemiology), Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
33
|
Valenzuela-Melgarejo FJ, Caro-Díaz C, Cabello-Guzmán G. Potential Crosstalk between Fructose and Melatonin: A New Role of Melatonin-Inhibiting the Metabolic Effects of Fructose. Int J Endocrinol 2018; 2018:7515767. [PMID: 30154843 PMCID: PMC6092995 DOI: 10.1155/2018/7515767] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/22/2018] [Accepted: 06/19/2018] [Indexed: 12/13/2022] Open
Abstract
Increased consumption of energy-dense foods such as fructose-rich syrups represents one of the significant, growing concerns related to the alarming trend of overweight, obesity, and metabolic disorders worldwide. Metabolic pathways affected by fructose involve genes related to lipogenesis/lipolysis, beta-oxidation, mitochondrial biogenesis, gluconeogenesis, oxidative phosphorylation pathways, or altering of circadian production of insulin and leptin. Moreover, fructose can be a risk factor during pregnancy elevating the risk of preterm delivery, hypertension, and metabolic impairment of the mother and fetus. Melatonin is a chronobiotic and homeostatic hormone that can modulate the harmful effects of fructose via clock gene expression and metabolic pathways, modulating the expression of PPARγ, SREBF-1 (SREBP-1), hormone-sensitive lipase, C/EBP-α genes, NRF-1, PGC1α, and uncoupling protein-1. Moreover, this hormone has the capacity in the rat of reverting the harmful effects of fructose, increasing the body weight and weight ratio of the liver, and increasing the body weight and restoring the glycemia from mothers exposed to fructose. The aim of this review is to show the potential crosstalk between fructose and melatonin and their potential role during pregnancy.
Collapse
Affiliation(s)
| | - Claudia Caro-Díaz
- Laboratory of Molecular Cell Biology, Department of Basic Sciences, Universidad del Bío-Bío, Campus Fernando May, Chillán, Chile
| | - Gerardo Cabello-Guzmán
- Laboratory of Molecular Cell Biology, Department of Basic Sciences, Universidad del Bío-Bío, Campus Fernando May, Chillán, Chile
| |
Collapse
|
34
|
Cardinali DP, Vigo DE. Melatonin, mitochondria, and the metabolic syndrome. Cell Mol Life Sci 2017; 74:3941-3954. [PMID: 28819865 PMCID: PMC11107716 DOI: 10.1007/s00018-017-2611-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 08/03/2017] [Indexed: 12/12/2022]
Abstract
A number of risk factors for cardiovascular disease including hyperinsulinemia, glucose intolerance, dyslipidemia, obesity, and elevated blood pressure are collectively known as metabolic syndrome (MS). Since mitochondrial activity is modulated by the availability of energy in cells, the disruption of key regulators of metabolism in MS not only affects the activity of mitochondria but also their dynamics and turnover. Therefore, a link of MS with mitochondrial dysfunction has been suspected since long. As a chronobiotic/cytoprotective agent, melatonin has a special place in prevention and treatment of MS. Melatonin levels are reduced in diseases associated with insulin resistance like MS. Melatonin improves sleep efficiency and has antioxidant and anti-inflammatory properties, partly for its role as a metabolic regulator and mitochondrial protector. We discuss in the present review the several cytoprotective melatonin actions that attenuate inflammatory responses in MS. The clinical data that support the potential therapeutical value of melatonin in human MS are reviewed.
Collapse
Affiliation(s)
- Daniel P Cardinali
- BIOMED-UCA-CONICET and Department of Teaching and Research, Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Av. Alicia Moreau de Justo 1500, 4o piso, 1107, Buenos Aires, Argentina.
| | - Daniel E Vigo
- BIOMED-UCA-CONICET and Department of Teaching and Research, Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Av. Alicia Moreau de Justo 1500, 4o piso, 1107, Buenos Aires, Argentina
| |
Collapse
|
35
|
Isherwood CM, Van der Veen DR, Johnston JD, Skene DJ. Twenty-four-hour rhythmicity of circulating metabolites: effect of body mass and type 2 diabetes. FASEB J 2017; 31:5557-5567. [PMID: 28821636 PMCID: PMC5690388 DOI: 10.1096/fj.201700323r] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/07/2017] [Indexed: 12/16/2022]
Abstract
Metabolic profiling of individuals with type 2 diabetes mellitus (T2DM) has previously been limited to single-time-point samples, ignoring time-of-day variation. Here, we tested our hypothesis that body mass and T2DM affect daily rhythmicity and concentrations of circulating metabolites across a 24-h day in 3 age-matched, male groups-lean, overweight/obese (OW/OB), and OW/OB with T2DM-in controlled laboratory conditions, which were not confounded by large meals. By using targeted liquid chromatography/mass spectrometry metabolomics, we quantified 130 plasma metabolites every 2 h over 24 h, and we show that average metabolite concentrations were significantly altered by increased body mass (90 of 130) and T2DM (56 of 130). Thirty-eight percent of metabolites exhibited daily rhythms in at least 1 study group, and where a metabolite was rhythmic in >1 group, its peak time was comparable. The optimal time of day was assessed to provide discriminating biomarkers. This differed between metabolite classes and study groups-for example, phospholipids showed maximal difference at 5:00 AM (lean vs. OW/OB) and at 5:00 PM (OW/OB vs. T2DM). Metabolites that were identified with both robust 24-h rhythms and significant concentration differences between study groups emphasize the importance of controlling the time of day for diagnosis and biomarker discovery, offering a significant improvement over current single sampling.-Isherwood, C. M., Van der Veen, D. R., Johnston, J. D., Skene, D. J. Twenty-four-hour rhythmicity of circulating metabolites: effect of body mass and type 2 diabetes.
Collapse
Affiliation(s)
- Cheryl M Isherwood
- Chronobiology Section, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Daan R Van der Veen
- Chronobiology Section, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Jonathan D Johnston
- Chronobiology Section, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Debra J Skene
- Chronobiology Section, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
36
|
Qian J, Thomas AP, Schroeder AM, Rakshit K, Colwell CS, Matveyenko AV. Development of diabetes does not alter behavioral and molecular circadian rhythms in a transgenic rat model of type 2 diabetes mellitus. Am J Physiol Endocrinol Metab 2017; 313:E213-E221. [PMID: 28465284 PMCID: PMC5582890 DOI: 10.1152/ajpendo.00406.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 04/27/2017] [Accepted: 04/27/2017] [Indexed: 01/09/2023]
Abstract
Metabolic state and circadian clock function exhibit a complex bidirectional relationship. Circadian disruption increases propensity for metabolic dysfunction, whereas common metabolic disorders such as obesity and type 2 diabetes (T2DM) are associated with impaired circadian rhythms. Specifically, alterations in glucose availability and glucose metabolism have been shown to modulate clock gene expression and function in vitro; however, to date, it is unknown whether development of diabetes imparts deleterious effects on the suprachiasmatic nucleus (SCN) circadian clock and SCN-driven outputs in vivo. To address this question, we undertook studies in aged diabetic rats transgenic for human islet amyloid polypeptide, an established nonobese model of T2DM (HIP rat), which develops metabolic defects closely recapitulating those present in patients with T2DM. HIP rats were also cross-bred with a clock gene reporter rat model (Per1:luciferase transgenic rat) to permit assessment of the SCN and the peripheral molecular clock function ex vivo. Utilizing these animal models, we examined effects of diabetes on 1) behavioral circadian rhythms, 2) photic entrainment of circadian activity, 3) SCN and peripheral tissue molecular clock function, and 4) melatonin secretion. We report that circadian activity, light-induced entrainment, molecular clockwork, as well as melatonin secretion are preserved in the HIP rat model of T2DM. These results suggest that despite the well-characterized ability of glucose to modulate circadian clock gene expression acutely in vitro, SCN clock function and key behavioral and physiological outputs appear to be preserved under chronic diabetic conditions characteristic of nonobese T2DM.
Collapse
MESH Headings
- Animals
- Behavior, Animal/physiology
- Circadian Rhythm/genetics
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/physiopathology
- Disease Models, Animal
- Disease Progression
- Islet Amyloid Polypeptide/genetics
- Islet Amyloid Polypeptide/metabolism
- Light
- Male
- Period Circadian Proteins/metabolism
- Rats
- Rats, Sprague-Dawley
- Rats, Transgenic
- Suprachiasmatic Nucleus/metabolism
- Suprachiasmatic Nucleus/pathology
Collapse
Affiliation(s)
- Jingyi Qian
- Departments of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California
| | - Anthony P Thomas
- Department of Medicine, University of California Los Angeles, Los Angeles, California; and
| | - Analyne M Schroeder
- Departments of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California
| | - Kuntol Rakshit
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Christopher S Colwell
- Departments of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California
| | - Aleksey V Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, Minnesota;
| |
Collapse
|
37
|
Gubin DG, Nelaeva AA, Uzhakova AE, Hasanova YV, Cornelissen G, Weinert D. Disrupted circadian rhythms of body temperature, heart rate and fasting blood glucose in prediabetes and type 2 diabetes mellitus. Chronobiol Int 2017; 34:1136-1148. [PMID: 28759269 DOI: 10.1080/07420528.2017.1347670] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We report a progressive disruption of 24-h rhythms in fasting blood glucose (FBG), body temperature (BT) and heart rate (HR) associated with metabolic dysfunction and the development of prediabetes (PD) and type 2 diabetes mellitus (T2DM) in overweight middle-aged (40-69 years old) humans. Increasing BT and HR mean values and declining 24-h BT and HR amplitudes accompany adverse changes in metabolic state. Increased nocturnal BT and a phase delay of the 24-h BT rhythm, deviant 24-h HR profile and a phase advance of the 24-h HR and FBG rhythms are early signs of the PD metabolic state. In T2DM, the 24-h FBG rhythm is no longer detectable, and the 24-h amplitudes of BT and HR are greatly diminished. In addition, lepton and creatinine values were lowered in T2DM. Moreover, positive correlations between FBG and body mass index, BMI, and negative correlations between the 24-h amplitude of FBG and BMI indicate that overweight is an additional factor causing disruption of the circadian rhythms. Further studies on circadian disruption as a consequence of metabolic dysfunction are necessary. The quantitative analysis of changing circadian BT and HR rhythms may provide prognostic markers of T2DM and therapeutic targets for its prevention and correction.
Collapse
Affiliation(s)
- D G Gubin
- a Department of Biology , Medical University , Tyumen , Russia
| | - A A Nelaeva
- b Department of Endocrinology , Medical University , Tyumen , Russia
| | - A E Uzhakova
- b Department of Endocrinology , Medical University , Tyumen , Russia
| | - Y V Hasanova
- b Department of Endocrinology , Medical University , Tyumen , Russia
| | - G Cornelissen
- c Halberg Chronobiology Center , University of Minnesota , Minneapolis , MN , USA
| | - D Weinert
- d Institute of Biology/Zoology , Martin Luther University , Halle-Wittenberg , Germany
| |
Collapse
|
38
|
Dyar KA, Eckel-Mahan KL. Circadian Metabolomics in Time and Space. Front Neurosci 2017; 11:369. [PMID: 28744188 PMCID: PMC5504240 DOI: 10.3389/fnins.2017.00369] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 06/13/2017] [Indexed: 12/11/2022] Open
Abstract
Circadian rhythms are widely known to govern human health and disease, but specific pathogenic mechanisms linking circadian disruption to metabolic diseases are just beginning to come to light. This is thanks in part to the development and application of various "omics"-based tools in biology and medicine. Current high-throughput technologies allow for the simultaneous monitoring of multiple dynamic cellular events over time, ranging from gene expression to metabolite abundance and sub-cellular localization. These fundamental temporal and spatial perspectives have allowed for a more comprehensive understanding of how various dynamic cellular events and biochemical processes are related in health and disease. With advances in technology, metabolomics has become a more routine "omics" approach for studying metabolism, and "circadian metabolomics" (i.e., studying the 24-h metabolome) has recently been undertaken by several groups. To date, circadian metabolomes have been reported for human serum, saliva, breath, and urine, as well as tissues from several species under specific disease or mutagenesis conditions. Importantly, these studies have consistently revealed that 24-h rhythms are prevalent in almost every tissue and metabolic pathway. Furthermore, these circadian rhythms in tissue metabolism are ultimately linked to and directed by internal 24-h biological clocks. In this review, we will attempt to put these data-rich circadian metabolomics experiments into perspective to find out what they can tell us about metabolic health and disease, and what additional biomarker potential they may reveal.
Collapse
Affiliation(s)
- Kenneth A Dyar
- Molecular Endocrinology, Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD)Munich, Germany
| | - Kristin L Eckel-Mahan
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at HoustonHouston, TX, United States
| |
Collapse
|
39
|
Wehrens SMT, Christou S, Isherwood C, Middleton B, Gibbs MA, Archer SN, Skene DJ, Johnston JD. Meal Timing Regulates the Human Circadian System. Curr Biol 2017; 27:1768-1775.e3. [PMID: 28578930 PMCID: PMC5483233 DOI: 10.1016/j.cub.2017.04.059] [Citation(s) in RCA: 364] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 04/09/2017] [Accepted: 04/27/2017] [Indexed: 12/28/2022]
Abstract
Circadian rhythms, metabolism, and nutrition are intimately linked [1, 2], although effects of meal timing on the human circadian system are poorly understood. We investigated the effect of a 5-hr delay in meals on markers of the human master clock and multiple peripheral circadian rhythms. Ten healthy young men undertook a 13-day laboratory protocol. Three meals (breakfast, lunch, dinner) were given at 5-hr intervals, beginning either 0.5 (early) or 5.5 (late) hr after wake. Participants were acclimated to early meals and then switched to late meals for 6 days. After each meal schedule, participants' circadian rhythms were measured in a 37-hr constant routine that removes sleep and environmental rhythms while replacing meals with hourly isocaloric snacks. Meal timing did not alter actigraphic sleep parameters before circadian rhythm measurement. In constant routines, meal timing did not affect rhythms of subjective hunger and sleepiness, master clock markers (plasma melatonin and cortisol), plasma triglycerides, or clock gene expression in whole blood. Following late meals, however, plasma glucose rhythms were delayed by 5.69 ± 1.29 hr (p < 0.001), and average glucose concentration decreased by 0.27 ± 0.05 mM (p < 0.001). In adipose tissue, PER2 mRNA rhythms were delayed by 0.97 ± 0.29 hr (p < 0.01), indicating that human molecular clocks may be regulated by feeding time and could underpin plasma glucose changes. Timed meals therefore play a role in synchronizing peripheral circadian rhythms in humans and may have particular relevance for patients with circadian rhythm disorders, shift workers, and transmeridian travelers.
Collapse
Affiliation(s)
- Sophie M T Wehrens
- Faculty of Health and Medical Science, University of Surrey, Stag Hill Campus, Guildford, Surrey GU2 7XH, UK
| | - Skevoulla Christou
- Faculty of Health and Medical Science, University of Surrey, Stag Hill Campus, Guildford, Surrey GU2 7XH, UK
| | - Cheryl Isherwood
- Faculty of Health and Medical Science, University of Surrey, Stag Hill Campus, Guildford, Surrey GU2 7XH, UK
| | - Benita Middleton
- Faculty of Health and Medical Science, University of Surrey, Stag Hill Campus, Guildford, Surrey GU2 7XH, UK
| | - Michelle A Gibbs
- Faculty of Health and Medical Science, University of Surrey, Stag Hill Campus, Guildford, Surrey GU2 7XH, UK
| | - Simon N Archer
- Faculty of Health and Medical Science, University of Surrey, Stag Hill Campus, Guildford, Surrey GU2 7XH, UK
| | - Debra J Skene
- Faculty of Health and Medical Science, University of Surrey, Stag Hill Campus, Guildford, Surrey GU2 7XH, UK
| | - Jonathan D Johnston
- Faculty of Health and Medical Science, University of Surrey, Stag Hill Campus, Guildford, Surrey GU2 7XH, UK.
| |
Collapse
|
40
|
Forrestel AC, Miedlich SU, Yurcheshen M, Wittlin SD, Sellix MT. Chronomedicine and type 2 diabetes: shining some light on melatonin. Diabetologia 2017; 60:808-822. [PMID: 27981356 DOI: 10.1007/s00125-016-4175-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/18/2016] [Indexed: 12/19/2022]
Abstract
In mammals, the circadian timing system drives rhythms of physiology and behaviour, including the daily rhythms of feeding and activity. The timing system coordinates temporal variation in the biochemical landscape with changes in nutrient intake in order to optimise energy balance and maintain metabolic homeostasis. Circadian disruption (e.g. as a result of shift work or jet lag) can disturb this continuity and increase the risk of cardiometabolic disease. Obesity and metabolic disease can also disturb the timing and amplitude of the clock in multiple organ systems, further exacerbating disease progression. As our understanding of the synergy between the timing system and metabolism has grown, an interest has emerged in the development of novel clock-targeting pharmaceuticals or nutraceuticals for the treatment of metabolic dysfunction. Recently, the pineal hormone melatonin has received some attention as a potential chronotherapeutic drug for metabolic disease. Melatonin is well known for its sleep-promoting effects and putative activity as a chronobiotic drug, stimulating coordination of biochemical oscillations through targeting the internal timing system. Melatonin affects the insulin secretory activity of the pancreatic beta cell, hepatic glucose metabolism and insulin sensitivity. Individuals with type 2 diabetes mellitus have lower night-time serum melatonin levels and increased risk of comorbid sleep disturbances compared with healthy individuals. Further, reduced melatonin levels, and mutations and/or genetic polymorphisms of the melatonin receptors are associated with an increased risk of developing type 2 diabetes. Herein we review our understanding of molecular clock control of glucose homeostasis, detail the influence of circadian disruption on glucose metabolism in critical peripheral tissues, explore the contribution of melatonin signalling to the aetiology of type 2 diabetes, and discuss the pros and cons of melatonin chronopharmacotherapy in disease management.
Collapse
Affiliation(s)
- Andrew C Forrestel
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 693, Rochester, NY, 14642, USA
| | - Susanne U Miedlich
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 693, Rochester, NY, 14642, USA
| | - Michael Yurcheshen
- UR Medicine Sleep Center, Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Steven D Wittlin
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 693, Rochester, NY, 14642, USA
| | - Michael T Sellix
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 693, Rochester, NY, 14642, USA.
| |
Collapse
|
41
|
Reutrakul S, Siwasaranond N, Nimitphong H, Saetung S, Chirakalwasan N, Chailurkit LO, Srijaruskul K, Ongphiphadhanakul B, Thakkinstian A. Associations between nocturnal urinary 6-sulfatoxymelatonin, obstructive sleep apnea severity and glycemic control in type 2 diabetes. Chronobiol Int 2017; 34:382-392. [PMID: 28128991 DOI: 10.1080/07420528.2016.1278382] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Reduced nocturnal secretion of melatonin, a pineal hormone under circadian control, and obstructive sleep apnea have been both identified as risk factors for the development of type 2 diabetes mellitus. Whether they interact to impact glycemic control in patients with existing type 2 diabetes is not known. Therefore, this study explores the relationships between obstructive sleep apnea, melatonin and glycemic control in type 2 diabetes. As diabetic retinopathy may affect melatonin secretion, we also explore the relationship between retinopathy, melatonin and glycemic control. Fifty-six non-shift workers with type 2 diabetes, who were not using beta-blockers, participated. Most recent hemoglobin A1c (HbA1c) levels and the results of ophthalmologic examinations were obtained from medical records. Obstructive sleep apnea was diagnosed using an ambulatory device. Sleep duration and fragmentation were recorded by 7-day wrist actigraphy. The urinary 6-sulfatoxymelatonin/creatinine ratio, an indicator of nocturnal melatonin secretion, was measured in an overnight urine sample. Mediation analyses were applied to explore whether low nocturnal urinary 6-sulfatoxymelatonin/creatinine ratio could be a causal link between increasing obstructive sleep apnea severity [as measured by an Apnea Hypopnea Index (AHI)] and poorer glycemic control, and between the presence of retinopathy and glycemic control. AHI and HbA1c were log-scale (ln) transformed. Obstructive sleep apnea was found in 76.8%, and 25.5% had diabetic retinopathy. The median (interquartile range) of urinary 6-sulfatoxymelatonin/creatinine ratio was 12.3 (6.0, 20.1) ng/mg. Higher lnHbA1c significantly correlated with lower 6-sulfatoxymelatonin/creatinine ratio (p = 0.04) but was not directly associated with OSA severity. More severe obstructive sleep apnea (lnAHI, p = 0.01), longer diabetes duration (p = 0.02), retinopathy (p = 0.01) and insulin use (p = 0.03) correlated with lower urinary 6-sulfatoxymelatonin/creatinine ratio, while habitual sleep duration and fragmentation did not. A mediation analysis revealed that lnAHI negatively correlated with urinary 6-sulfatoxymelatonin/creatinine ratio (coefficient = -2.413, p = 0.03), and urinary 6-sulfatoxymelatonin/creatinine negatively associated with lnHbA1c (coefficient = -0.005, p = 0.02), after adjusting for covariates. Mediation analysis indicated that the effect of lnAHI on lnHbA1c was indirectly mediated by urinary 6-sulfatoxymelatonin/creatinine ratio (B = 0.013, 95% CI: 0.0006, 0.0505). In addition, having retinopathy was significantly associated with reduced nocturnal urinary 6-sulfatoxymelatonin/creatinine ratio, and an increase in HbA1c by 1.013% of its original value (B = -0.013, 95% CI: -0.038, -0.005). In conclusion, the presence and severity of obstructive sleep apnea as well as the presence of diabetic retinopathy were associated with lower nocturnal melatonin secretion, with an indirect adverse effect on glycemic control. Intervention studies are needed to determine whether melatonin supplementation may be beneficial in type 2 diabetes patients with obstructive sleep apnea.
Collapse
Affiliation(s)
- Sirimon Reutrakul
- a Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital , Mahidol University , Bangkok , Thailand
| | - Nantaporn Siwasaranond
- a Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital , Mahidol University , Bangkok , Thailand
| | - Hataikarn Nimitphong
- a Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital , Mahidol University , Bangkok , Thailand
| | - Sunee Saetung
- a Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital , Mahidol University , Bangkok , Thailand
| | - Naricha Chirakalwasan
- b Division of Pulmonary and Critical Care Medicine, Department of Medicine, Faculty of Medicine , Chulalongkorn University , Bangkok , Thailand.,c Excellence Center for Sleep Disorders , King Chulalongkorn Memorial Hospital, Thai Red Cross Society , Bangkok , Thailand
| | - La-Or Chailurkit
- a Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital , Mahidol University , Bangkok , Thailand
| | - Kriangsuk Srijaruskul
- a Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital , Mahidol University , Bangkok , Thailand
| | - Boonsong Ongphiphadhanakul
- a Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital , Mahidol University , Bangkok , Thailand
| | - Ammarin Thakkinstian
- d Section for Clinical Epidemiology and Biostatistics, Faculty of Medicine , Ramathibodi Hospital, Mahidol University , Bangkok , Thailand
| |
Collapse
|
42
|
Thomas AP, Hoang J, Vongbunyong K, Nguyen A, Rakshit K, Matveyenko AV. Administration of Melatonin and Metformin Prevents Deleterious Effects of Circadian Disruption and Obesity in Male Rats. Endocrinology 2016; 157:4720-4731. [PMID: 27653034 PMCID: PMC5133345 DOI: 10.1210/en.2016-1309] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Circadian disruption and obesity synergize to predispose to development of type 2 diabetes mellitus (T2DM), signifying that therapeutic targeting of both circadian and metabolic dysfunctions should be considered as a potential treatment approach. To address this hypothesis, we studied rats concomitantly exposed to circadian disruption and diet-induced obesity (CDO), a rat model recently shown to recapitulate phenotypical aspects of obese T2DM (eg, circadian disruption, obesity, insulin resistance, and islet failure). CDO rats were subsequently treated daily (for 12 wk) by timed oral gavage with vehicle, melatonin (a known chronobiotic), metformin, or combination treatment of both therapeutics. Melatonin treatment alone improved circadian activity rhythms, attenuated induction of β-cell failure, and enhanced glucose tolerance. Metformin alone did not modify circadian activity but enhanced insulin sensitivity and glucose tolerance. Importantly, the combination of melatonin and metformin had synergistic actions to modify progression of metabolic dysfunction in CDO rats through improved adiposity, circadian activity, insulin sensitivity, and islet cell failure. This study suggests that management of both circadian and metabolic dysfunctions should be considered as a potential preventative and therapeutic option for treatment of obesity and T2DM.
Collapse
Affiliation(s)
- Anthony P Thomas
- Department of Physiology and Biomedical Engineering (K.R., A.V.M.), Mayo Clinic School of Medicine, Mayo Clinic, Rochester, Minnesota 55905; and Department of Medicine (A.P.T., J.H., K.V., A.N., A.V.M.), University of California Los Angeles, Los Angeles, California 90095
| | - Jonathan Hoang
- Department of Physiology and Biomedical Engineering (K.R., A.V.M.), Mayo Clinic School of Medicine, Mayo Clinic, Rochester, Minnesota 55905; and Department of Medicine (A.P.T., J.H., K.V., A.N., A.V.M.), University of California Los Angeles, Los Angeles, California 90095
| | - Kenny Vongbunyong
- Department of Physiology and Biomedical Engineering (K.R., A.V.M.), Mayo Clinic School of Medicine, Mayo Clinic, Rochester, Minnesota 55905; and Department of Medicine (A.P.T., J.H., K.V., A.N., A.V.M.), University of California Los Angeles, Los Angeles, California 90095
| | - Andrew Nguyen
- Department of Physiology and Biomedical Engineering (K.R., A.V.M.), Mayo Clinic School of Medicine, Mayo Clinic, Rochester, Minnesota 55905; and Department of Medicine (A.P.T., J.H., K.V., A.N., A.V.M.), University of California Los Angeles, Los Angeles, California 90095
| | - Kuntol Rakshit
- Department of Physiology and Biomedical Engineering (K.R., A.V.M.), Mayo Clinic School of Medicine, Mayo Clinic, Rochester, Minnesota 55905; and Department of Medicine (A.P.T., J.H., K.V., A.N., A.V.M.), University of California Los Angeles, Los Angeles, California 90095
| | - Aleksey V Matveyenko
- Department of Physiology and Biomedical Engineering (K.R., A.V.M.), Mayo Clinic School of Medicine, Mayo Clinic, Rochester, Minnesota 55905; and Department of Medicine (A.P.T., J.H., K.V., A.N., A.V.M.), University of California Los Angeles, Los Angeles, California 90095
| |
Collapse
|
43
|
McMullan CJ, Rimm EB, Schernhammer ES, Forman JP. A nested case-control study of the association between melatonin secretion and incident myocardial infarction. Heart 2016; 103:694-701. [PMID: 27806988 DOI: 10.1136/heartjnl-2016-310098] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 10/06/2016] [Accepted: 10/09/2016] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVES Low nocturnal melatonin secretion is associated with cardiovascular risk factors, diabetes and hypertension, while individuals with prevalent cardiovascular disease have lower nocturnal melatonin levels. However, the prospective association of melatonin secretion with myocardial infarction (MI) has not been studied. We aimed to study the association between melatonin secretion and the risk of developing MI. METHODS We performed a prospective nested case-control study of participants from the Nurses' Health Study cohorts I and II. A total of 209 incident cases of fatal and non-fatal MI were identified among women who provided first morning voided urine specimens at baseline and were matched to 209 controls. Nocturnal melatonin secretion was assessed using 6-sulfatoxymelatonin concentrations in morning urines normalised to the urines' creatinine concentration. Multivariable conditional logistic regression was used to analyse associations independent of important risk factors. RESULTS Lower melatonin secretion was significantly associated with a higher risk of MI. After conditioning on matching variables, the OR for every one unit lower log-transformed sulfatoxymelatonin/creatinine ratio was 1.51 (95% CI 1.16 to 1.96). In multivariable models controlling for factors included in the American Heart Association Cardiovascular Risk Score plus circadian factors, every one unit lower in the ratio was associated with a significantly increased risk of MI (OR, 1.40; 95% CI 1.02 to 1.93). Women in the highest category had an estimated absolute risk of MI of 84 cases per 100 000 person-years compared with 197 cases per 100 000 person-years in the lowest category. The association was strongly modified by body mass index (BMI) (p value for interaction=0.02). CONCLUSIONS Lower melatonin secretion was significantly associated with a greater risk of incident MI in women with increased BMI. Melatonin may be a novel and modifiable risk factor for MI among such women.
Collapse
Affiliation(s)
- Ciaran J McMullan
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Eric B Rimm
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Eva S Schernhammer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - John P Forman
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
44
|
Abstract
Use of artificial light resulted in relative independence from the natural light-dark (LD) cycle, allowing human subjects to shift the timing of food intake and work to convenient times. However, the increase in artificial light exposure parallels the increase in obesity prevalence. Light is the dominant Zeitgeber for the central circadian clock, which resides within the hypothalamic suprachiasmatic nucleus, and coordinates daily rhythm in feeding behaviour and metabolism. Eating during inappropriate light conditions may result in metabolic disease via changes in the biological clock. In this review, we describe the physiological role of light in the circadian timing system and explore the interaction between the circadian timing system and metabolism. Furthermore, we discuss the acute and chronic effects of artificial light exposure on food intake and energy metabolism in animals and human subjects. We propose that living in synchrony with the natural daily LD cycle promotes metabolic health and increased exposure to artificial light at inappropriate times of day has adverse effects on metabolism, feeding behaviour and body weight regulation. Reducing the negative side effects of the extensive use of artificial light in human subjects might be useful in the prevention of metabolic disease.
Collapse
|
45
|
Szewczyk-Golec K, Woźniak A, Reiter RJ. Inter-relationships of the chronobiotic, melatonin, with leptin and adiponectin: implications for obesity. J Pineal Res 2015; 59:277-91. [PMID: 26103557 DOI: 10.1111/jpi.12257] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 06/19/2015] [Indexed: 12/15/2022]
Abstract
Obesity and its medical complications represent a significant problem throughout the world. In recent decades, mechanisms underlying the progression of obesity have been intensively examined. The involvement of both the behavioral aspects, such as calorie-rich diet, low physical activity and sleep deprivation, and the intrinsic factors, including adipose tissue deregulation, chronic inflammation, oxidative stress, and chronodisruption, has been identified. The circadian disturbances of the adipose tissue endocrine function have been correlated with obesity. Leptin and adiponectin are adipokines strongly associated with glucose and lipid metabolism and with energy balance. Their synthesis and secretion display circadian rhythms that are disturbed in the obese state. Hyperleptinemia resulting in leptin resistance, and hypo-adiponectinemia have been linked to the pathophysiology of the obesity-related disorders. A deficiency of melatonin, one of the consequences of sleep deprivation, has also been demonstrated to correlate with obesity. Melatonin is a pineal secretory product involved in numerous actions, such as regulation of internal biological clocks and energy metabolism, and it functions as an antioxidant and as an anti-inflammatory agent. There exists a substantial amount of evidence supporting the beneficial effects of melatonin supplementation on obesity and its complications. In the current review, the results of studies related to the interactions between melatonin, and both leptin and adiponectin are discussed. Despite the existence of some inconsistencies, melatonin has been found to normalize the expression and secretion patterns of both adipokines. These results support the concept of melatonin as a potential therapeutic agent for obesity and related disorders.
Collapse
Affiliation(s)
- Karolina Szewczyk-Golec
- The Chair of Medical Biology, Nicolaus Copernicus University, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| | - Alina Woźniak
- The Chair of Medical Biology, Nicolaus Copernicus University, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
46
|
Johnston JD, Skene DJ. 60 YEARS OF NEUROENDOCRINOLOGY: Regulation of mammalian neuroendocrine physiology and rhythms by melatonin. J Endocrinol 2015; 226:T187-98. [PMID: 26101375 DOI: 10.1530/joe-15-0119] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2015] [Indexed: 12/15/2022]
Abstract
The isolation of melatonin was first reported in 1958. Since the demonstration that pineal melatonin synthesis reflects both daily and seasonal time, melatonin has become a key element of chronobiology research. In mammals, pineal melatonin is essential for transducing day-length information into seasonal physiological responses. Due to its lipophilic nature, melatonin is able to cross the placenta and is believed to regulate multiple aspects of perinatal physiology. The endogenous daily melatonin rhythm is also likely to play a role in the maintenance of synchrony between circadian clocks throughout the adult body. Pharmacological doses of melatonin are effective in resetting circadian rhythms if taken at an appropriate time of day, and can acutely regulate factors such as body temperature and alertness, especially when taken during the day. Despite the extensive literature on melatonin physiology, some key questions remain unanswered. In particular, the amplitude of melatonin rhythms has been recently associated with diseases such as type 2 diabetes mellitus but understanding of the physiological significance of melatonin rhythm amplitude remains poorly understood.
Collapse
Affiliation(s)
- Jonathan D Johnston
- Faculty of Health and Medical SciencesUniversity of Surrey, Guildford, Surrey GU2 7XH, UK
| | - Debra J Skene
- Faculty of Health and Medical SciencesUniversity of Surrey, Guildford, Surrey GU2 7XH, UK
| |
Collapse
|
47
|
Abstract
Circadian rhythms, metabolism and nutrition are closely interlinked. A great deal of recent research has investigated not only how aspects of metabolic physiology are driven by circadian clocks, but also how these circadian clocks are themselves sensitive to metabolic change. At the cellular level, novel feedback loops have been identified that couple circadian 'clock genes' and their proteins to expression of nuclear receptors, regulation of redox state and other major pathways. Using targeted disruption of circadian clocks, mouse models are providing novel insight into the role of tissue-specific clocks in glucose homeostasis and body weight regulation. The relationship between circadian rhythms and obesity appears complex, with variable alteration of rhythms in obese individuals. However, it is clear from animal studies that the timing and nutritional composition of meals can regulate circadian rhythms, particularly in peripheral tissues. Translation of these findings to human physiology now represents an important goal.
Collapse
Affiliation(s)
- Jonathan D Johnston
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| |
Collapse
|
48
|
Saini C, Brown SA, Dibner C. Human peripheral clocks: applications for studying circadian phenotypes in physiology and pathophysiology. Front Neurol 2015; 6:95. [PMID: 26029154 PMCID: PMC4429585 DOI: 10.3389/fneur.2015.00095] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 04/20/2015] [Indexed: 01/26/2023] Open
Abstract
Most light-sensitive organisms on earth have acquired an internal system of circadian clocks allowing the anticipation of light or darkness. In humans, the circadian system governs nearly all aspects of physiology and behavior. Circadian phenotypes, including chronotype, vary dramatically among individuals and over individual lifespan. Recent studies have revealed that the characteristics of human skin fibroblast clocks correlate with donor chronotype. Given the complexity of circadian phenotype assessment in humans, the opportunity to study oscillator properties by using cultured primary cells has the potential to uncover molecular details difficult to assess directly in humans. Since altered properties of the circadian oscillator have been associated with many diseases including metabolic disorders and cancer, clock characteristics assessed in additional primary cell types using similar technologies might represent an important tool for exploring the connection between chronotype and disease, and for diagnostic purposes. Here, we review implications of this approach for gathering insights into human circadian rhythms and their function in health and disease.
Collapse
Affiliation(s)
- Camille Saini
- Department of Medical Specialties, Faculty of Medicine, University of Geneva , Geneva , Switzerland
| | - Steven A Brown
- Institute of Pharmacology and Toxicology, University of Zürich , Zürich , Switzerland
| | - Charna Dibner
- Department of Medical Specialties, Faculty of Medicine, University of Geneva , Geneva , Switzerland
| |
Collapse
|
49
|
Abstract
Most living beings, including humans, must adapt to rhythmically occurring daily changes in their environment that are generated by the Earth's rotation. In the course of evolution, these organisms have acquired an internal circadian timing system that can anticipate environmental oscillations and thereby govern their rhythmic physiology in a proactive manner. In mammals, the circadian timing system coordinates virtually all physiological processes encompassing vigilance states, metabolism, endocrine functions and cardiovascular activity. Research performed during the past two decades has established that almost every cell in the body possesses its own circadian timekeeper. The resulting clock network is organized in a hierarchical manner. A master pacemaker, located in the suprachiasmatic nucleus (SCN) of the hypothalamus, is synchronized every day to the photoperiod. In turn, the SCN determines the phase of the cellular clocks in peripheral organs through a wide variety of signalling pathways dependent on feeding cycles, body temperature rhythms, oscillating bloodborne signals and, in some organs, inputs of the peripheral nervous system. A major purpose of circadian clocks in peripheral tissues is the temporal orchestration of key metabolic processes, including food processing (metabolism and xenobiotic detoxification). Here, we review some recent findings regarding the molecular and cellular composition of the circadian timing system and discuss its implications for the temporal coordination of metabolism in health and disease. We focus primarily on metabolic disorders such as obesity and type 2 diabetes, although circadian misalignments (shiftwork or 'social jet lag') have also been associated with the aetiology of human malignancies.
Collapse
Affiliation(s)
- C Dibner
- Department of Endocrinology, Diabetes, Nutrition and Hypertension, University Hospital of Geneva, Geneva, Switzerland
| | | |
Collapse
|
50
|
Costes S, Boss M, Thomas AP, Matveyenko AV. Activation of Melatonin Signaling Promotes β-Cell Survival and Function. Mol Endocrinol 2015; 29:682-92. [PMID: 25695910 DOI: 10.1210/me.2014-1293] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by pancreatic islet failure due to loss of β-cell secretory function and mass. Studies have identified a link between a variance in the gene encoding melatonin (MT) receptor 2, T2DM, and impaired insulin secretion. This genetic linkage raises the question whether MT signaling plays a role in regulation of β-cell function and survival in T2DM. To address this postulate, we used INS 832/13 cells to test whether activation of MT signaling attenuates proteotoxicity-induced β-cell apoptosis and through which molecular mechanism. We also used nondiabetic and T2DM human islets to test the potential of MT signaling to attenuate deleterious effects of glucotoxicity and T2DM on β-cell function. MT signaling in β-cells (with duration designed to mimic typical nightly exposure) significantly enhanced activation of the cAMP-dependent signal transduction pathway and attenuated proteotoxicity-induced β-cell apoptosis evidenced by reduced caspase-3 cleavage (∼40%), decreased activation of stress-activated protein kinase/Jun-amino-terminal kinase (∼50%) and diminished oxidative stress response. Activation of MT signaling in human islets was shown to restore glucose-stimulated insulin secretion in islets exposed to chronic hyperglycemia as well as in T2DM islets. Our data suggest that β-cell MT signaling is important for the regulation of β-cell survival and function and implies a preventative and therapeutic potential for preservation of β-cell mass and function in T2DM.
Collapse
Affiliation(s)
- Safia Costes
- Department of Medicine (S.C., M.B., A.P.T., A.V.M.), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095; and Department of Physiology and Biomedical Engineering (A.V.M.), Mayo Clinic School of Medicine, Mayo Clinic Rochester, Minnesota 55905
| | | | | | | |
Collapse
|