1
|
Feeney SP, McCarthy JM, Petruconis CR, Tudor JC. Sleep loss is a metabolic disorder. Sci Signal 2025; 18:eadp9358. [PMID: 40198749 DOI: 10.1126/scisignal.adp9358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 03/12/2025] [Indexed: 04/10/2025]
Abstract
Sleep loss dysregulates cellular metabolism and energy homeostasis. Highly metabolically active cells, such as neurons, enter a catabolic state during periods of sleep loss, which consequently disrupts physiological functioning. Specific to the central nervous system, sleep loss results in impaired synaptogenesis and long-term memory, effects that are also characteristic of neurodegenerative diseases. In this review, we describe how sleep deprivation increases resting energy expenditure, leading to the development of a negative energy balance-a state with insufficient metabolic resources to support energy expenditure-in highly active cells like neurons. This disruption of energetic homeostasis alters the balance of metabolites, including adenosine, lactate, and lipid peroxides, such that energetically costly processes, such as synapse formation, are attenuated. During sleep loss, metabolically active cells shunt energetic resources away from those processes that are not acutely essential, like memory formation, to support cell survival. Ultimately, these findings characterize sleep loss as a metabolic disorder.
Collapse
Affiliation(s)
- Sierra P Feeney
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Jordan M McCarthy
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Cecilia R Petruconis
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Jennifer C Tudor
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| |
Collapse
|
2
|
Deng S, Hu Y, Chen S, Xue Y, Yao D, Sun Q, Nedergaard M, Wang W, Ding F. Chronic sleep fragmentation impairs brain interstitial clearance in young wildtype mice. J Cereb Blood Flow Metab 2024; 44:1515-1531. [PMID: 38639025 PMCID: PMC11418708 DOI: 10.1177/0271678x241230188] [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: 06/27/2023] [Revised: 10/06/2023] [Accepted: 12/27/2023] [Indexed: 04/20/2024]
Abstract
Accumulating evidence shows that most chronic neurological diseases have a link with sleep disturbances, and that patients with chronically poor sleep undergo an accelerated cognitive decline. Indeed, a single-night of sleep deprivation may increase metabolic waste levels in cerebrospinal fluid. However, it remains unknown how chronic sleep disturbances in isolation from an underlying neurological disease may affect the glymphatic system. Clearance of brain interstitial waste by the glymphatic system occurs primarily during sleep, driven by multiple oscillators including arterial pulsatility, and vasomotion. Herein, we induced sleep fragmentation in young wildtype mice and assessed the effects on glymphatic activity and cognitive functions. Chronic sleep fragmentation reduced glymphatic function and impaired cognitive functions in healthy mice. A mechanistic analysis showed that the chronic sleep fragmentation suppressed slow vasomotion, without altering cardiac-driven pulsations. Taken together, results of this study document that chronic sleep fragmentation suppresses brain metabolite clearance and impairs cognition, even in the absence of disease.
Collapse
Affiliation(s)
- Saiyue Deng
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yusi Hu
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Simiao Chen
- Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, 310003, China
| | - Yang Xue
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Di Yao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qian Sun
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Maiken Nedergaard
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Department of Neurology, University of Rochester Medical Center, Rochester, NY, 14642, United States
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fengfei Ding
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
3
|
Gangitano E, Martinez-Sanchez N, Bellini MI, Urciuoli I, Monterisi S, Mariani S, Ray D, Gnessi L. Weight Loss and Sleep, Current Evidence in Animal Models and Humans. Nutrients 2023; 15:3431. [PMID: 37571368 PMCID: PMC10420950 DOI: 10.3390/nu15153431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Sleep is a vital process essential for survival. The trend of reduction in the time dedicated to sleep has increased in industrialized countries, together with the dramatic increase in the prevalence of obesity and diabetes. Short sleep may increase the risk of obesity, diabetes and cardiovascular disease, and on the other hand, obesity is associated with sleep disorders, such as obstructive apnea disease, insomnia and excessive daytime sleepiness. Sleep and metabolic disorders are linked; therefore, identifying the physiological and molecular pathways involved in sleep regulation and metabolic homeostasis can play a major role in ameliorating the metabolic health of the individual. Approaches aimed at reducing body weight could provide benefits for both cardiometabolic risk and sleep quality, which indirectly, in turn, may determine an amelioration of the cardiometabolic phenotype of individuals. We revised the literature on weight loss and sleep, focusing on the mechanisms and the molecules that may subtend this relationship in humans as in animal models.
Collapse
Affiliation(s)
- Elena Gangitano
- OCDEM Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Noelia Martinez-Sanchez
- OCDEM Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
| | | | - Irene Urciuoli
- Department of Surgery, Sapienza University of Rome, 00161 Rome, Italy
| | - Stefania Monterisi
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Stefania Mariani
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - David Ray
- OCDEM Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
| | - Lucio Gnessi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
4
|
Tsereteli N, Vallat R, Fernandez-Tajes J, Delahanty LM, Ordovas JM, Drew DA, Valdes AM, Segata N, Chan AT, Wolf J, Berry SE, Walker MP, Spector TD, Franks PW. Impact of insufficient sleep on dysregulated blood glucose control under standardised meal conditions. Diabetologia 2022; 65:356-365. [PMID: 34845532 PMCID: PMC8741723 DOI: 10.1007/s00125-021-05608-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/03/2021] [Indexed: 12/01/2022]
Abstract
AIMS/HYPOTHESIS Sleep, diet and exercise are fundamental to metabolic homeostasis. In this secondary analysis of a repeated measures, nutritional intervention study, we tested whether an individual's sleep quality, duration and timing impact glycaemic response to a breakfast meal the following morning. METHODS Healthy adults' data (N = 953 [41% twins]) were analysed from the PREDICT dietary intervention trial. Participants consumed isoenergetic standardised meals over 2 weeks in the clinic and at home. Actigraphy was used to assess sleep variables (duration, efficiency, timing) and continuous glucose monitors were used to measure glycaemic variation (>8000 meals). RESULTS Sleep variables were significantly associated with postprandial glycaemic control (2 h incremental AUC), at both between- and within-person levels. Sleep period time interacted with meal type, with a smaller effect of poor sleep on postprandial blood glucose levels when high-carbohydrate (low fat/protein) (pinteraction = 0.02) and high-fat (pinteraction = 0.03) breakfasts were consumed compared with a reference 75 g OGTT. Within-person sleep period time had a similar interaction (high carbohydrate: pinteraction = 0.001, high fat: pinteraction = 0.02). Within- and between-person sleep efficiency were significantly associated with lower postprandial blood glucose levels irrespective of meal type (both p < 0.03). Later sleep midpoint (time deviation from midnight) was found to be significantly associated with higher postprandial glucose, in both between-person and within-person comparisons (p = 0.035 and p = 0.051, respectively). CONCLUSIONS/INTERPRETATION Poor sleep efficiency and later bedtime routines are associated with more pronounced postprandial glycaemic responses to breakfast the following morning. A person's deviation from their usual sleep pattern was also associated with poorer postprandial glycaemic control. These findings underscore sleep as a modifiable, non-pharmacological therapeutic target for the optimal regulation of human metabolic health. Trial registration ClinicalTrials.gov NCT03479866.
Collapse
Affiliation(s)
- Neli Tsereteli
- Genetic & Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Raphael Vallat
- Center for Human Sleep Science, Department of Psychology, University of California, Berkeley, CA, USA
| | - Juan Fernandez-Tajes
- Genetic & Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Linda M Delahanty
- Diabetes Center, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jose M Ordovas
- JM-USDA-Human Nutrition Research Diabetes Center on Aging at Tufts University, Boston, MA, USA
- IMDEA-Food, Madrid, Spain
| | - David A Drew
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ana M Valdes
- NIHR Nottingham BRC at the Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
| | - Nicola Segata
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
- European Institute of Oncology Scientific Institute for Research, Hospitalization and Healthcare, Milan, Italy
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Sarah E Berry
- Department of Nutritional Research, Kings College London, London, UK
| | - Matthew P Walker
- Center for Human Sleep Science, Department of Psychology, University of California, Berkeley, CA, USA
| | | | - Paul W Franks
- Genetic & Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, Sweden.
- Harvard Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
5
|
Liew SC, Aung T. Sleep deprivation and its association with diseases- a review. Sleep Med 2020; 77:192-204. [PMID: 32951993 DOI: 10.1016/j.sleep.2020.07.048] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/08/2020] [Accepted: 07/27/2020] [Indexed: 01/02/2023]
Abstract
Sleep deprivation, a consequence of multiple health problems or a cause of many major health risks, is a significant public health concern in this era. In the recent years, numerous reports have been added to the literature to provide explanation and to answer previously unanswered questions on this important topic but comprehensive updates and reviews in this aspect remain scarce. The present study identified 135 papers that investigated the association between sleep deprivation and health risks, including cardiovascular, respiratory, neurological, gastrointestinal, immunology, dermatology, endocrine, and reproductive health. In this review, we aimed to provide insight into the association between sleep deprivation and the development of diseases. We reviewed the latest updates available in the literature and particular attention was paid to reports that detailed all possible causal relationships involving both extrinsic and intrinsic factors that may be relevant to this topic. Various mechanisms by which sleep deprivation may affect health were presented and discussed, and this review hopes to serve as a platform for ideas generation for future research.
Collapse
Affiliation(s)
- Siaw Cheok Liew
- Department of Clinical Competence, Perdana University-Royal College of Surgeons in Ireland, Kuala Lumpur, Malaysia.
| | - Thidar Aung
- Department of Biochemistry, Perdana University-Royal College of Surgeons in Ireland, Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Triplett J, Ellis D, Braddock A, Roberts E, Ingram K, Perez E, Short A, Brown D, Hutzley V, Webb C, Soto A, Chan V. Temporal and region-specific effects of sleep fragmentation on gut microbiota and intestinal morphology in Sprague Dawley rats. Gut Microbes 2020; 11:706-720. [PMID: 31924109 PMCID: PMC7524289 DOI: 10.1080/19490976.2019.1701352] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Sleep is a fundamental biological process, that when repeatedly disrupted, can result in severe health consequences. Recent studies suggest that both sleep fragmentation (SF) and dysbiosis of the gut microbiome can lead to metabolic disorders, though the underlying mechanisms are largely unclear. To better understand the consequences of SF, we investigated the effects of acute (6 days) and chronic (6 weeks) SF on rats by examining taxonomic profiles of microbiota in the distal ileum, cecum and proximal colon, as well as assessing structural and functional integrity of the gastrointestinal barrier. We further assayed the impact of SF on a host function by evaluating inflammation and immune response. Both acute and chronic SF induced microbial dysbiosis, more dramatically in the distal ileum (compared to other two regions studied), as noted by significant perturbations in alpha- and beta-diversity; though, specific microbial populations were significantly altered throughout each of the three regions. Furthermore, chronic SF resulted in increased crypt depth in the distal ileum and an increase in the number of villi lining both the cecum and proximal colon. Additional changes were noted with chronic SF, including: decreased microbial adhesion and penetration in the distal ileum and cecum, elevation in serum levels of the cytokine KC/GRO, and depressed levels of corticotropin. Importantly, our data show that perturbations to microbial ecology and intestinal morphology intensify in response to prolonged SF and these changes are habitat specific. Together, these results reveal consequences to gut microbiota homeostasis and host response following acute and chronic SF in rats.
Collapse
Affiliation(s)
- Judy Triplett
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - David Ellis
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Amber Braddock
- Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF), Wright-Patterson AFB, OH, USA
| | - Erin Roberts
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Katherine Ingram
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Eric Perez
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Amanda Short
- Air Force Research Laboratory, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Dominique Brown
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| | - Victoria Hutzley
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| | - Chelsey Webb
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| | - Armando Soto
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| | - Victor Chan
- Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA,CONTACT Victor Chan Molecular Mechanisms Branch, Human Centered ISR Division, Airman Systems Directorate, 711 Human Performance Wing, Air Force Research Laboratory (711 HPW/RHXJ), Wright-Patterson AFB, OH, USA
| |
Collapse
|
7
|
Fatima N, Rana S. Metabolic implications of circadian disruption. Pflugers Arch 2020; 472:513-526. [PMID: 32363530 DOI: 10.1007/s00424-020-02381-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 01/20/2023]
Abstract
Circadian rhythms are generated by the circadian clock, a self-sustained internal timing system that exhibits 24-h rhythms in the body. In mammals, circadian rhythms are driven by a central clock located in suprachiasmatic nucleus and various peripheral clocks located in different tissues and organs of the body. Many cellular, behavioral, and physiological processes are regulated by the circadian clock in coordination with environmental cues. The process of metabolism is also under circadian regulation. Loss of synchronization between the internal clock and environmental zeitgebers results in disruption of the circadian rhythms that seriously impacts metabolic homeostasis leading to changed eating behavior, altered glucose and lipid metabolism, and weight gain. This in turn augments the risk of having various cardio-metabolic disorders such as obesity, diabetes, metabolic syndrome, and cardiovascular disease. This review sheds light on circadian rhythms and their role in metabolism with the identification of gaps in the current knowledge that remain to be explored in these fields. In this review, the molecular mechanisms underlying circadian rhythms have been elaborated first. Then, the focus has been kept on explaining the physiological significance of circadian rhythms in regulating metabolism. Finally, the implications for metabolism when these rhythms are disrupted due to genetic mutations or social and occupational needs enforced by modern lifestyle have been discussed.
Collapse
Affiliation(s)
- Narjis Fatima
- Molecular Biology and Human Genetics Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan
| | - Sobia Rana
- Molecular Biology and Human Genetics Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
8
|
Meningeal Lymphangiogenesis and Enhanced Glymphatic Activity in Mice with Chronically Implanted EEG Electrodes. J Neurosci 2020; 40:2371-2380. [PMID: 32047056 DOI: 10.1523/jneurosci.2223-19.2020] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 12/27/2019] [Accepted: 01/22/2020] [Indexed: 12/26/2022] Open
Abstract
Chronic electroencephalography (EEG) is a widely used tool for monitoring cortical electrical activity in experimental animals. Although chronic implants allow for high-quality, long-term recordings in preclinical studies, the electrodes are foreign objects and might therefore be expected to induce a local inflammatory response. We here analyzed the effects of chronic cranial electrode implantation on glymphatic fluid transport and in provoking structural changes in the meninges and cerebral cortex of male and female mice. Immunohistochemical analysis of brain tissue and dura revealed reactive gliosis in the cortex underlying the electrodes and extensive meningeal lymphangiogenesis in the surrounding dura. Meningeal lymphangiogenesis was also evident in mice prepared with the commonly used chronic cranial window. Glymphatic influx of a CSF tracer was significantly enhanced at 30 d postsurgery in both awake and ketamine-xylazine anesthetized mice with electrodes, supporting the concept that glymphatic influx and intracranial lymphatic drainage are interconnected. Altogether, the experimental results provide clear evidence that chronic implantation of EEG electrodes is associated with significant changes in the brain's fluid transport system. Future studies involving EEG recordings and chronic cranial windows must consider the physiological consequences of cranial implants, which include glial scarring, meningeal lymphangiogenesis, and increased glymphatic activity.SIGNIFICANCE STATEMENT This study shows that implantation of extradural electrodes provokes meningeal lymphangiogenesis, enhanced glymphatic influx of CSF, and reactive gliosis. The analysis based on CSF tracer injection in combination with immunohistochemistry showed that chronically implanted electroencephalography electrodes were surrounded by lymphatic sprouts originating from lymphatic vasculature along the dural sinuses and the middle meningeal artery. Likewise, chronic cranial windows provoked lymphatic sprouting. Tracer influx assessed in coronal slices was increased in agreement with previous reports identifying a close association between glymphatic activity and the meningeal lymphatic vasculature. Lymphangiogenesis in the meninges and altered glymphatic fluid transport after electrode implantation have not previously been described and adds new insights to the foreign body response of the CNS.
Collapse
|
9
|
Gozal D, Khalyfa A, Qiao Z, Akbarpour M, Maccari R, Ottanà R. Protein-Tyrosine Phosphatase-1B Mediates Sleep Fragmentation-Induced Insulin Resistance and Visceral Adipose Tissue Inflammation in Mice. Sleep 2018. [PMID: 28651353 DOI: 10.1093/sleep/zsx111] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Study Objectives Sleep fragmentation (SF) is highly prevalent and has emerged as an important contributing factor to obesity and metabolic syndrome. We hypothesized that SF-induced increases in protein tyrosine phosphatase-1B (PTP-1B) expression and activity underlie increased food intake, inflammation, and leptin and insulin resistance. Methods Wild-type (WT) and ObR-PTP-1b-/- mice (Tg) were exposed to SF and control sleep (SC), and food intake was monitored. WT mice received a PTP-1B inhibitor (RO-7d; Tx) or vehicle (Veh). Upon completion of exposures, systemic insulin and leptin sensitivity tests were performed as well as assessment of visceral white adipose tissue (vWAT) insulin receptor sensitivity and macrophages (ATM) polarity. Results SF increased food intake in either untreated or Veh-treated WT mice. Leptin-induced hypothalamic STAT3 phosphorylation was decreased, PTP-1B activity was increased, and reduced insulin sensitivity emerged both systemic and in vWAT, with the latter displaying proinflammatory ATM polarity changes. All of the SF-induced effects were abrogated following PTP-1B inhibitor treatment and in Tg mice. Conclusions SF induces increased food intake, reduced leptin signaling in hypothalamus, systemic insulin resistance, and reduced vWAT insulin sensitivity and inflammation that are mediated by increased PTP-1B activity. Thus, PTP-1B may represent a viable therapeutic target in the context of SF-induced weight gain and metabolic dysfunction.
Collapse
Affiliation(s)
- David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Abdelnaby Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Zhuanghong Qiao
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Mahzad Akbarpour
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Rosanna Maccari
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, PoloAnnunziata, V.le SS. Annunziata, Messina, Italy
| | - Rosaria Ottanà
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, PoloAnnunziata, V.le SS. Annunziata, Messina, Italy
| |
Collapse
|
10
|
Oishi K, Ohyama S, Higo-Yamamoto S. Chronic sleep disorder induced by psychophysiological stress induces glucose intolerance without adipose inflammation in mice. Biochem Biophys Res Commun 2018; 495:2616-2621. [PMID: 29288667 DOI: 10.1016/j.bbrc.2017.12.158] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 12/26/2017] [Indexed: 01/11/2023]
Abstract
Sleep disturbances are associated with various metabolic diseases such as hypertension and diabetes. We had previously established a mouse model of a psychophysiological stress-induced chronic sleep disorder (CSD) characterized by disrupted circadian rhythms of wheel-running activity, core body temperature, and sleep-wake cycles. To evaluate the underlying mechanisms of metabolic disorders induced by CSD, we created mice with CSD for six weeks and fed them with a high-fat diet. Glucose intolerance with hyperglycemia resulted, although plasma insulin levels and body weight increases were identical between control and CSD mice. Gluconeogenesis and glycolysis were enhanced and suppressed, respectively, in the livers of CSD mice, because the mRNA expression of Pck1 was significantly increased, whereas that of Gck and Pklr were significantly decreased in the CSD mice. Adipose inflammation induced by the high-fat diet seemed suppressed by the CSD, because the mRNA expression levels of Adgre1, Ccl2, and Tnf were significantly downregulated in the adipose tissues of CSD mice. These findings suggest that CSD impair glucose tolerance by inducing gluconeogenesis and suppressing glycolysis. Hyperphasia with hypoleptinemia, hypercorticosteronemia, and increased plasma free fatty acids might be involved in the impaired glucose metabolism under a CSD. Further studies are needed to elucidate the endocrine and molecular mechanisms underlying the associations between sleep disorders and impaired glucose homeostasis that consequently causes diabetes.
Collapse
Affiliation(s)
- Katsutaka Oishi
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan; Department of Applied Biological Science, Graduate School of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan; Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan.
| | - Sumika Ohyama
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Sayaka Higo-Yamamoto
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| |
Collapse
|
11
|
Shift-work: is time of eating determining metabolic health? Evidence from animal models. Proc Nutr Soc 2018; 77:199-215. [DOI: 10.1017/s0029665117004128] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The circadian disruption in shift-workers is suggested to be a risk factor to develop overweight and metabolic dysfunction. The conflicting time signals given by shifted activity, shifted food intake and exposure to light at night occurring in the shift-worker are proposed to be the cause for the loss of internal synchrony and the consequent adverse effects on body weight and metabolism. Because food elicited signals have proven to be potent entraining signals for peripheral oscillations, here we review the findings from experimental models of shift-work and verify whether they provide evidence about the causal association between shifted feeding schedules, circadian disruption and altered metabolism. We found mainly four experimental models that mimic the conditions of shift-work: protocols of forced sleep deprivation, of forced activity during the normal rest phase, exposure to light at night and shifted food timing. A big variability in the intensity and duration of the protocols was observed, which led to a diversity of effects. A common result was the disruption of temporal patterns of activity; however, not all studies explored the temporal patterns of food intake. According to studies that evaluate time of food intake as an experimental model of shift-work and studies that evaluate shifted food consumption, time of food intake may be a determining factor for the loss of balance at the circadian and metabolic level.
Collapse
|
12
|
Ho JM, Ducich NH, Nguyen NQK, Opp MR. Acute sleep disruption- and high-fat diet-induced hypothalamic inflammation are not related to glucose tolerance in mice. Neurobiol Sleep Circadian Rhythms 2018; 4:1-9. [PMID: 29732438 PMCID: PMC5931726 DOI: 10.1016/j.nbscr.2017.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Chronic insufficient sleep is a major societal problem and is associated with increased risk of metabolic disease. Hypothalamic inflammation contributes to hyperphagia and weight gain in diet-induced obesity, but insufficient sleep-induced neuroinflammation has yet to be examined in relation to metabolic function. We therefore fragmented sleep of adult male C57BL/6J mice for 18 h daily for 9 days to determine whether sleep disruption elicits inflammatory responses in brain regions that regulate energy balance and whether this relates to glycemic control. To additionally test the hypothesis that exposure to multiple inflammatory factors exacerbates metabolic outcomes, responses were compared in mice exposed to sleep fragmentation (SF), high-fat diet (HFD), both SF and HFD, or control conditions. Three or 9 days of high-fat feeding reduced glucose tolerance but SF alone did not. Transient loss of body mass in SF mice may have affected outcomes. Comparisons of pro-inflammatory cytokine concentrations among central and peripheral metabolic tissues indicate that patterns of liver interleukin-1β concentrations best reflects observed changes in glucose tolerance. However, we demonstrate that SF rapidly and potently increases Iba1 immunoreactivity (-ir), a marker of microglia. After 9 days of manipulations, Iba1-ir remains elevated only in mice exposed to both SF and HFD, indicating a novel interaction between sleep and diet on microglial activation that warrants further investigation.
Collapse
Affiliation(s)
- Jacqueline M. Ho
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - Nicole H. Ducich
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - Nhat-Quynh K. Nguyen
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - Mark R. Opp
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
- Program in Neurobiology and Behavior, University of Washington, Seattle, Washington, USA
- Corresponding author. Present addrss: Department of Integrative Physiology, University of Colorado, UCB 354, 2860 Wilderness Place, 201K, Boulder, CO 80301, USA.
| |
Collapse
|
13
|
Espitia-Bautista E, Velasco-Ramos M, Osnaya-Ramírez I, Ángeles-Castellanos M, Buijs RM, Escobar C. Social jet-lag potentiates obesity and metabolic syndrome when combined with cafeteria diet in rats. Metabolism 2017. [PMID: 28641787 DOI: 10.1016/j.metabol.2017.04.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND/OBJECTIVES Modern lifestyle promotes shifted sleep onset and shifted wake up time between weekdays and weekends, producing a condition termed "social-jet lag." Disrupted sleep promotes increased appetite for carbohydrate and fat-rich food, which in long term leads to overweight, obesity and metabolic syndrome. In order to mimic the human situation we produced an experimental model of social-jet lag (Sj-l). With this model, we explored the link between shifted sleep time with consumption of a cafeteria diet (CafD) and the development of obesity and metabolic syndrome. SUBJECTS/METHODS The first experiment was designed to create and confirm the model of Sj-l. Rats (n=8-10/group) were exposed to a shifted sleep time protocol achieved by placing the rats in slow rotating wheels from Monday to Friday during the first 4h of the light period, while on weekends they were left undisturbed. The second experiment (n=8-12/group) explored the combined effect of Sj-l with the opportunity to ingest CafD. All protocols lasted 12weeks. We evaluated the development of overweight and indicators of metabolic syndrome. The statistical significance for all variables was set at P<0.05. RESULTS Sj-l alone did not affect body weight gain but induced significant changes in cholesterol in metabolic variables representing a risk factor for metabolic syndrome. Daily restricted access to CafD in the day or night induced glucose intolerance and only CafD during the day led to overweight. Sj-l combined with CafD induced overconsumption of the diet, potentiated body weight gain (16%) and promoted 5 of the criteria for metabolic syndrome including high insulin and dislipidemia. CONCLUSION Present data provide an experimental model of social-jet lag that combined with overconsumption of CafD, and maximized the development of obesity and metabolic syndrome. Importantly, access to CafD during the night did not lead to overweight nor metabolic syndrome.
Collapse
Affiliation(s)
- Estefania Espitia-Bautista
- Facultad de Medicina, Departamento de Anatomía, Universidad Nacional Autónoma de México, México, DF 04510, Mexico
| | - Mario Velasco-Ramos
- Facultad de Medicina, Departamento de Anatomía, Universidad Nacional Autónoma de México, México, DF 04510, Mexico; Departamento de Biología Molecular, Instituto Nacional de Cardiología, 14080, México, DF, Mexico
| | - Iván Osnaya-Ramírez
- Facultad de Medicina, Departamento de Anatomía, Universidad Nacional Autónoma de México, México, DF 04510, Mexico
| | - Manuel Ángeles-Castellanos
- Facultad de Medicina, Departamento de Anatomía, Universidad Nacional Autónoma de México, México, DF 04510, Mexico
| | - Ruud M Buijs
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, DF 04510, Mexico
| | - Carolina Escobar
- Facultad de Medicina, Departamento de Anatomía, Universidad Nacional Autónoma de México, México, DF 04510, Mexico.
| |
Collapse
|