1
|
Sugiura C, Sato M, Tanaka S, Okumura N, Terao A. Royal jelly reduced non-rapid eye movement sleep fragmentation and restored sleep stability in diet-induced obese mice. Exp Anim 2025; 74:300-309. [PMID: 39805626 PMCID: PMC12044360 DOI: 10.1538/expanim.24-0128] [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: 09/29/2024] [Accepted: 12/25/2024] [Indexed: 01/16/2025] Open
Abstract
Royal jelly (RJ) is recognized due to its high nutritional value and potential health benefits. Previous research showed that RJ supplementation decreased fat accumulation, resulting in weight loss and improvements in hyperglycemia and insulin resistance in high-fat diet (HFD)-induced obese mice. To expand the weight-reducing properties of RJ, this study aimed to investigate the effects of RJ supplementation on HFD-induced obese mice with impaired sleep stabilization. Over a 20-week period, the C57BL/6J mice were divided into the following dietary groups: normal diet (ND), ND supplemented with 5% lyophilized RJ powder (ND+RJ), HFD, and HFD supplemented with 5% lyophilized RJ powder (HFD+RJ) groups. Compared with the HFD group, the HFD+RJ group exhibited a significant reduction in body weight via a decrease in fat mass. Moreover, much like the ND group, the HFD+RJ group demonstrated improvements in the fragmentation of non-rapid eye movement (NREM) sleep and wakefulness. These processes contributed to the reestablishment of sleep/wake continuity and restored the overall stability of sleep. In contrast, the ND+RJ and ND groups exhibited a similar sleep/wake architecture. Thus, RJ supplementation in the ND demonstrated no substantial effect on sleep/wake. According to these findings, dietary RJ improves the sleep/wake architecture and restores sleep stability. Hence, RJ is a promising dietary component for addressing obesity and restoring sleep stability.
Collapse
Affiliation(s)
- Chiaki Sugiura
- Department of Biology, School of Biological Sciences, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, Hokkaido 005-8601, Japan
| | - Mao Sato
- Department of Biology, School of Biological Sciences, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, Hokkaido 005-8601, Japan
| | - Shun Tanaka
- Department of Biology, School of Biological Sciences, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, Hokkaido 005-8601, Japan
| | - Nobuaki Okumura
- Institute for Bee Products and Health Science, R&D Department, Yamada Bee Company Inc., 194 Ichiba, Kagamino-cho, Tomata-gun, Okayama 708-0393, Japan
| | - Akira Terao
- Department of Biology, School of Biological Sciences, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, Hokkaido 005-8601, Japan
| |
Collapse
|
2
|
Gou R, Chen L, Cheng Z, Cun J, Li G. Association of dietary intake of saturated fatty acids with obstructive sleep apnea: mediating effects of Life's Crucial 9. Front Nutr 2025; 12:1503815. [PMID: 40034735 PMCID: PMC11872719 DOI: 10.3389/fnut.2025.1503815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Obstructive sleep apnea (OSA) is a global public health issue. Life's Crucial 9 (LC9) is recognized as a powerful tool for assessing cardiovascular health. Although the etiology of OSA remains unclear, saturated fatty acids (SFAs) and cardiovascular health are increasingly regarded as a non-negligible element. This study aims to assess the association between dietary intake of SFAs and the risk of OSA, and the mediating effect of LC9. Methods Based on the National Health and Nutrition Examination Survey (NHANES), dietary questionnaires of participant were collected, and the average values of 24-h dietary recall data over 2 days were obtained. A continuous cross-sectional analysis with dietary energy adjustment was employed. Weighted multivariable logistic regression models were used to estimate the weighted odds ratios (ORs) and their 95% confidence intervals (CIs) for SFAs and OSA. Evaluate the mediating role of LC9 in the relationship between SFAs and OSA. Results A total of 13,563 participants aged 20 years and above were included in this study. The intakes of Sfa 4.0 and LC9 among participants with OSA were significantly lower than those in the normal population. After adjusting for confounding factors, total SFAs could increase the risk of OSA [Model 1, Q3, 0.03, 1.49 (1.03, 2.15); Model 2, Q3, 0.04, 1.47 (1.01, 2.13)]. It was emphasized that dietary intake of Sfa 12.0, Sfa 14.0, and Sfa 16.0 were protective factors for OSA, especially among participants aged 45-64 years and white individuals. Moreover, Sfa 12.0 exhibited a better protective effect in female participants [Q3, 0.04, 0.66 (0.45, 0.99)]. In addition, the cardiovascular health score - LC9 had a mediating effect in Sfa4.0 on OSA [Proportion of mediation: -0.035, 95% CI: (-0.058, -0.01); p= 0.002]. There was a nonlinear relationship between dietary intake of Sfa 12.0, Sfa 16.0, and Sfa 18.0 and OSA (P-Nonlinear = 0.013). Discussion These findings suggest that dietary mixtures of saturated fatty acids increase the risk of OSA. Among them, SFA 4:0 can increase the risk of OSA through the level of cardiovascular health. However, contrary to traditional beliefs, long-chain saturated fatty acids can reduce the risk of OSA.
Collapse
Affiliation(s)
- Ruoyu Gou
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Lili Chen
- Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Zeyi Cheng
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiawei Cun
- School of First Clinical Medical, Ningxia, Medical University, Yinchuan, Ningxia, China
| | - Guanghua Li
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
3
|
Wang J, Jiang N, Liu F, Wang C, Zhou W. Uncovering the intricacies of O-GlcNAc modification in cognitive impairment: New insights from regulation to therapeutic targeting. Pharmacol Ther 2025; 266:108761. [PMID: 39603350 DOI: 10.1016/j.pharmthera.2024.108761] [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: 05/08/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024]
Abstract
O-linked β-N-acetylglucosamine (O-GlcNAc) represents a post-translational modification that occurs on serine or threonine residues on various proteins. This conserved modification interacts with vital cellular pathways. Although O-GlcNAc is widely distributed throughout the body, it is particularly enriched in the brain, where most proteins are O-GlcNAcylated. Recent studies have established a causal link between O-GlcNAc regulation in the brain and alterations in neurophysiological function. Alterations in O-GlcNAc levels in the brain are associated with the pathogenesis of several neurogenic diseases that can lead to cognitive impairment. Remarkably, manipulation of O-GlcNAc levels demonstrated a protective effect on cognitive function. Although the precise molecular mechanism of O-GlcNAc modification in the nervous system remains elusive, its regulation is fundamental to multiple neural and cognitive functions, fluctuating levels during normal and pathological cognitive processes. In this review, we highlight the significant functional importance of O-GlcNAc modification in pathological cognitive impairments and the potential application of O-GlcNAc as a promising target for the intervention or amelioration of cognitive impairments.
Collapse
Affiliation(s)
- Jianhui Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Medicines, Beijing 100850, China
| | - Ning Jiang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Medicines, Beijing 100850, China
| | - Feng Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Medicines, Beijing 100850, China
| | - Chenran Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Medicines, Beijing 100850, China
| | - Wenxia Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Medicines, Beijing 100850, China.
| |
Collapse
|
4
|
Sato M, Sugiura C, Okumura N, Terao A. Melinjo (Gnetum gnemon L.) seed extract for treatment of sleep/wake fragmentation in diet-induced obese mice. Exp Anim 2025; 74:114-121. [PMID: 39245578 PMCID: PMC11742479 DOI: 10.1538/expanim.24-0050] [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: 05/11/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024] Open
Abstract
Dietary supplementation with melinjo (Gnetum gnemon L.) seed extract (MSE) has been an integral part of an anti-obesity therapeutic regimen. To examine the relationship between anti-obesity and sleep, we explored the effect of MSE on sleep structure in high-fat diet (HFD)-induced obese mice. Although HFD did not alter the total amount of daily sleep, it significantly reduced the average duration of non-rapid eye movement (NREM) sleep and wakefulness episodes and significantly increased the number of these episodes. These findings indicate fragmented NREM sleep due to repeated brief awakenings in the HFD-fed mice. When 1% (w/v) MSE was given to HFD-fed mice, their weight or sleep structure were comparable to those of ND-fed mice, proving that dietary MSE completely hindered HFD-induced weight gain and sleep/wake fragmentation. Our data provide compelling evidence that MSE is a novel and promising dietary supplement that restores obesity-induced sleep architecture changes in mice.
Collapse
Affiliation(s)
- Mao Sato
- Department of Biology, School of Biological Sciences, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, Hokkaido 005-8601, Japan
| | - Chiaki Sugiura
- Department of Biology, School of Biological Sciences, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, Hokkaido 005-8601, Japan
| | - Nobuaki Okumura
- Institute for Bee Products and Health Science, R&D Department, Yamada Bee Co. Inc., 194 Ichiba, Kagamino-cho, Tomata-gun, Okayama 708-0393, Japan
| | - Akira Terao
- Department of Biology, School of Biological Sciences, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, Hokkaido 005-8601, Japan
| |
Collapse
|
5
|
Kiss MG, Cohen O, McAlpine CS, Swirski FK. Influence of sleep on physiological systems in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1284-1300. [PMID: 39528718 PMCID: PMC11567060 DOI: 10.1038/s44161-024-00560-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Sleep is a fundamental requirement of life and is integral to health. Deviation from optimal sleep associates with numerous diseases including those of the cardiovascular system. Studies, spanning animal models to humans, show that insufficient, disrupted or inconsistent sleep contribute to poor cardiovascular health by disrupting body systems. Fundamental experiments have begun to uncover the molecular and cellular links between sleep and heart health while large-scale human studies have associated sleep with cardiovascular outcomes in diverse populations. Here, we review preclinical and clinical findings that demonstrate how sleep influences the autonomic nervous, metabolic and immune systems to affect atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Máté G Kiss
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Oren Cohen
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S McAlpine
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Filip K Swirski
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
6
|
O'Hearn LA. Signals of energy availability in sleep: consequences of a fat-based metabolism. Front Nutr 2024; 11:1397185. [PMID: 39267859 PMCID: PMC11390529 DOI: 10.3389/fnut.2024.1397185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 08/05/2024] [Indexed: 09/15/2024] Open
Abstract
Humans can flexibly switch between two primary metabolic modes, usually distinguished by whether substrate supply from glucose can meet energy demands or not. However, it is often overlooked that when glucose use is limited, the remainder of energy needs may still be met more or less effectively with fat and ketone bodies. Hence a fat-based metabolism marked by ketosis is often conflated with starvation and contexts of inadequate energy (including at the cellular level), even when energy itself is in ample supply. Sleep and satiation are regulated by common pathways reflecting energy metabolism. A conceptual analysis that distinguishes signals of inadequate energy in a glucose-dominant metabolism from signals of a fat-based metabolism that may well be energy sufficient allows a reexamination of experimental results in the study of sleep that may shed light on species differences and explain why ketogenic diets have beneficial effects simultaneously in the brain and the periphery. It may also help to distinguish clinically when a failure of a ketogenic diet to resolve symptoms is due to inadequate energy rather than the metabolic state itself.
Collapse
|
7
|
Han Y, Xia G, Harris L, Liu P, Guan D, Wu Q. Reversal of Obesity by Enhancing Slow-wave Sleep via a Prokineticin Receptor Neural Circuit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591948. [PMID: 38746393 PMCID: PMC11092673 DOI: 10.1101/2024.04.30.591948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Obese subjects often exhibit hypersomnia accompanied by severe sleep fragmentation, while emerging evidence suggests that poor sleep quality promotes overeating and exacerbates diet-induced obesity (DIO). However, the neural circuit and signaling mechanism underlying the reciprocal control of appetite and sleep is yet not elucidated. Here, we report a neural circuit where prokineticin receptor 2 (PROKR2)-expressing neurons within the parabrachial nucleus (PBN) of the brainstem received direct projections from neuropeptide Y receptor Y2 (NPY2R)-expressing neurons within the lateral preoptic area (LPO) of the hypothalamus. The RNA-Seq results revealed Prokr2 in the PBN is the most regulated GPCR signaling gene that is responsible for comorbidity of obesity and sleep dysfunction. Furthermore, those NPY2R LPO neurons are minimally active during NREM sleep and maximally active during wakefulness and REM sleep. Activation of the NPY2R LPO →PBN circuit or the postsynaptic PROKR2 PBN neurons suppressed feeding of a high-fat diet and abrogated morbid sleep patterns in DIO mice. Further studies showed that genetic ablation of the PROKR2 signaling within PROKR2 PBN neurons alleviated the hyperphagia and weight gain, and restored sleep dysfunction in DIO mice. We further discovered pterostilbene, a plant-derived stilbenoid, is a powerful anti-obesity and sleep-improving agent, robustly suppressed hyperphagia and promoted reconstruction of a healthier sleep architecture, thereby leading to significant weight loss. Collectively, our results unveil a neural mechanism for the reciprocal control of appetite and sleep, through which pterostilbene, along with a class of similarly structured compounds, may be developed as effective therapeutics for tackling obesity and sleep disorders.
Collapse
|
8
|
Kang J, Park M, Oh CM, Kim T. High-fat diet-induced dopaminergic dysregulation induces REM sleep fragmentation and ADHD-like behaviors. Psychiatry Res 2023; 327:115412. [PMID: 37607442 DOI: 10.1016/j.psychres.2023.115412] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 07/27/2023] [Accepted: 08/10/2023] [Indexed: 08/24/2023]
Abstract
Consumption of a high-fat diet (HFD) has been associated with reduced wakefulness and various behavioral deficits, including anxiety, depression, and anhedonia. The dopaminergic system, which plays a crucial role in sleep and ADHD, is known to be vulnerable to chronic HFD. However, the association between HFD-induced behavioral and molecular changes remains unclear. Therefore, we investigated the effects of a HFD on the dopaminergic system and its association with behavioral deficits in male mice. The mice were divided into normal diet and HFD groups and were analyzed for sleep patterns, behavior tests, and transcription levels of dopamine-related genes in the brain. The HFD group showed decreased wakefulness, increased REM sleep with fragmented patterns, decreased time spent in the center zone of the open field test, shorter immobile time in the tail suspension test, impaired visuospatial memory, and reduced sucrose preference. Additionally, the HFD group had decreased mRNA levels of D1R, COMT, and DAT in the nucleus accumbens, which negatively correlated with REM sleep proportion and REM sleep bout count. The results suggest that HFD-induced behavioral deficits were resemblance to ADHD-like behavioral phenotypes and disturbs REM sleep by dysregulating the dopaminergic system.
Collapse
Affiliation(s)
- Jiseung Kang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Mincheol Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| | - Tae Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| |
Collapse
|
9
|
Gangitano E, Baxter M, Voronkov M, Lenzi A, Gnessi L, Ray D. The interplay between macronutrients and sleep: focus on circadian and homeostatic processes. Front Nutr 2023; 10:1166699. [PMID: 37680898 PMCID: PMC10482045 DOI: 10.3389/fnut.2023.1166699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 08/04/2023] [Indexed: 09/09/2023] Open
Abstract
Sleep disturbances are an emerging risk factor for metabolic diseases, for which the burden is particularly worrying worldwide. The importance of sleep for metabolic health is being increasingly recognized, and not only the amount of sleep plays an important role, but also its quality. In this review, we studied the evidence in the literature on macronutrients and their influence on sleep, focusing on the mechanisms that may lay behind this interaction. In particular, we focused on the effects of macronutrients on circadian and homeostatic processes of sleep in preclinical models, and reviewed the evidence of clinical studies in humans. Given the importance of sleep for health, and the role of circadian biology in healthy sleep, it is important to understand how macronutrients regulate circadian clocks and sleep homeostasis.
Collapse
Affiliation(s)
- Elena Gangitano
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Matthew Baxter
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Maria Voronkov
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucio Gnessi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - David Ray
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
10
|
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
|
11
|
Patel K, Lawson M, Cheung J. Whole-food plant-based diet reduces daytime sleepiness in patients with OSA. Sleep Med 2023; 107:327-329. [PMID: 37285791 DOI: 10.1016/j.sleep.2023.05.007] [Citation(s) in RCA: 11] [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: 04/04/2023] [Revised: 04/27/2023] [Accepted: 05/11/2023] [Indexed: 06/09/2023]
Abstract
Consumption of a diet high in saturated fat has been associated with daytime sleepiness. A whole-food plant-based (WFPB) dietary pattern, which is low in saturated fat, has been shown to be beneficial in a variety of health conditions. We assessed the effect of a short-term (21 days) WFPB diet intervention on daytime sleepiness in 14 patients with obstructive sleep apnea (OSA). We found a mean decrease of 3.8 points (SD = 3.3, p = 0.003) on the Epworth Sleepiness Scale (ESS) after switching from a standard Western diet to a WFPB diet. Our results suggest that a WFPB diet could be a viable dietary intervention to reduce symptom of daytime sleepiness.
Collapse
Affiliation(s)
- Kripa Patel
- Division of Allergy, Pulmonary and Sleep Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Melanie Lawson
- Division of Allergy, Pulmonary and Sleep Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Joseph Cheung
- Division of Allergy, Pulmonary and Sleep Medicine, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
12
|
Kim LJ, Alexandre C, Pho H, Latremoliere A, Polotsky VY, Pham LV. Diet-induced obesity leads to sleep fragmentation independently of the severity of sleep-disordered breathing. J Appl Physiol (1985) 2022; 133:1284-1294. [PMID: 36201322 PMCID: PMC9678416 DOI: 10.1152/japplphysiol.00386.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/14/2022] [Accepted: 09/28/2022] [Indexed: 11/22/2022] Open
Abstract
Obesity is associated with sleep-disordered breathing (SDB) and unrefreshing sleep. Residual daytime sleepiness and sleep impairments often persist after SDB treatment in patients with obesity, which suggests an independent effect of obesity on breathing and sleep. However, examining the relationship between sleep architecture and SDB in patients with obesity is complex and can be confounded by multiple factors. The main goal of this study was to examine the relationship between obesity-related changes in sleep architecture and SDB. Sleep recordings were performed in 15 lean C57BL/6J and 17 diet-induced obesity (DIO) mice of the same genetic background. Arousals from sleep and apneas were manually scored. Respiratory arousals were classified as events associated with ≥30% drops in minute ventilation (VE) from baseline. We applied Poincaré analysis of VE during sleep to estimate breathing variability. Obesity augmented the frequency of arousals by 45% and this increase was independent of apneas. Respiratory arousals comprised only 15% of the arousals in both groups of mice. Breathing variability during non-rapid-eye-movment (NREM) sleep was significantly higher in DIO mice, but it was not associated with arousal frequency. Our results suggest that obesity induces sleep fragmentation independently of SDB severity.NEW & NOTEWORTHY Our diet-induced obesity (DIO) model reproduces sleep features of human obesity, including sleep fragmentation, increased apnea frequency, and larger breathing variability. DIO induces sleep fragmentation independently of apnea severity. Sleep fragmentation in DIO mice is mainly attributed to non-respiratory arousals. Increased breathing variability during sleep did not account for the higher arousal frequency in DIO. Our results provide a rationale to examine sleep in patients with obesity even when they are adequately treated for sleep-disordered breathing.
Collapse
Affiliation(s)
- Lenise J Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chloe Alexandre
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Huy Pho
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alban Latremoliere
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Luu V Pham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
13
|
Wang X, Xing K, He M, He T, Xiang X, Chen T, Zhang L, Li H. Time-restricted feeding is an intervention against excessive dark-phase sleepiness induced by obesogenic diet. Natl Sci Rev 2022; 10:nwac222. [PMID: 36825118 PMCID: PMC9942665 DOI: 10.1093/nsr/nwac222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/05/2022] [Accepted: 10/05/2022] [Indexed: 11/14/2022] Open
Abstract
High-fat diet (HFD)-induced obesity is a growing epidemic and major health concern. While excessive daytime sleepiness (EDS) is a common symptom of HFD-induced obesity, preliminary findings suggest that reduced wakefulness could be improved with time-restricted feeding (TRF). At present, however, the underlying neural mechanisms remain largely unknown. The paraventricular thalamic nucleus (PVT) plays a role in maintaining wakefulness. We found that chronic HFD impaired the activity of PVT neurons. Notably, inactivation of the PVT was sufficient to reduce and fragment wakefulness during the active phase in lean mice, similar to the sleep-wake alterations observed in obese mice with HFD-induced obesity. On the other hand, enhancing PVT neuronal activity consolidated wakefulness in mice with HFD-induced obesity. We observed that the fragmented wakefulness could be eliminated and reversed by TRF. Furthermore, TRF prevented the HFD-induced disruptions on synaptic transmission in the PVT, in a feeding duration-dependent manner. Collectively, our findings demonstrate that ad libitum access to a HFD results in inactivation of the PVT, which is critical to impaired nocturnal wakefulness and increased sleep, while TRF can prevent and reverse diet-induced PVT dysfunction and excessive sleepiness. We establish a link between TRF and neural activity, through which TRF can potentially serve as a lifestyle intervention against diet/obesity-related EDS.
Collapse
Affiliation(s)
- Xu Wang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China,MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Keke Xing
- Department of Anatomy, Histology & Embryology, Fourth Military Medical University, Xi’an 710032, China
| | - Mengge He
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China,MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ting He
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China,MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xinkuan Xiang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China,MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tao Chen
- Department of Anatomy, Histology & Embryology, Fourth Military Medical University, Xi’an 710032, China
| | | | | |
Collapse
|
14
|
Ramírez-Plascencia OD, Saderi N, Cárdenas-Romero S, García-García F, Peña-Escudero C, Flores-Sandoval O, Azuara-Álvarez L, Báez-Ruiz A, Salgado-Delgado R. Leptin and adiponectin regulate the activity of nuclei involved in sleep-wake cycle in male rats. Front Neurosci 2022; 16:907508. [PMID: 35937866 PMCID: PMC9355486 DOI: 10.3389/fnins.2022.907508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Epidemiological and experimental evidence recognize a relationship between sleep-wake cycles and adiposity levels, but the mechanisms that link both are not entirely understood. Adipose tissue secretes adiponectin and leptin hormones, mainly involved as indicators of adiposity levels and recently associated to sleep. To understand how two of the main adipose tissue hormones could influence sleep-wake regulation, we evaluated in male rats, the effect of direct administration of adiponectin or leptin in the ventrolateral preoptic nuclei (VLPO), a major area for sleep promotion. The presence of adiponectin (AdipoR1 and AdipoR2) and leptin receptors in VLPO were confirmed by immunohistochemistry. Adiponectin administration increased wakefulness during the rest phase, reduced delta power, and activated wake-promoting neurons, such as the locus coeruleus (LC), tuberomammillary nucleus (TMN) and hypocretin/orexin neurons (OX) within the lateral hypothalamus (LH) and perifornical area (PeF). Conversely, leptin promoted REM and NREM sleep, including increase of delta power during NREM sleep, and induced c-Fos expression in VLPO and melanin concentrating hormone expressing neurons (MCH). In addition, a reduction in wake-promoting neurons activity was found in the TMN, lateral hypothalamus (LH) and perifornical area (PeF), including in the OX neurons. Moreover, leptin administration reduced tyrosine hydroxylase (TH) immunoreactivity in the LC. Our data suggest that adiponectin and leptin act as hormonal mediators between the status of body energy and the regulation of the sleep-wake cycle.
Collapse
Affiliation(s)
- Oscar Daniel Ramírez-Plascencia
- Departamento de Fisiología Celular, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Nadia Saderi
- Departamento de Fisiología Celular, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Skarleth Cárdenas-Romero
- Departamento de Fisiología Celular, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Fabio García-García
- Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Veracruz, Mexico
| | - Carolina Peña-Escudero
- Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Veracruz, Mexico
| | - Omar Flores-Sandoval
- Departamento de Fisiología Celular, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Lucia Azuara-Álvarez
- Departamento de Fisiología Celular, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Adrián Báez-Ruiz
- Departamento de Fisiología Celular, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Roberto Salgado-Delgado
- Departamento de Fisiología Celular, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
- *Correspondence: Roberto Salgado-Delgado,
| |
Collapse
|
15
|
The Sleep-Promoting Ventrolateral Preoptic Nucleus: What Have We Learned over the Past 25 Years? Int J Mol Sci 2022; 23:ijms23062905. [PMID: 35328326 PMCID: PMC8954377 DOI: 10.3390/ijms23062905] [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] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 01/09/2023] Open
Abstract
For over a century, the role of the preoptic hypothalamus and adjacent basal forebrain in sleep-wake regulation has been recognized. However, for years, the identity and location of sleep- and wake-promoting neurons in this region remained largely unresolved. Twenty-five years ago, Saper and colleagues uncovered a small collection of sleep-active neurons in the ventrolateral preoptic nucleus (VLPO) of the preoptic hypothalamus, and since this seminal discovery the VLPO has been intensively investigated by labs around the world, including our own. Herein, we first review the history of the preoptic area, with an emphasis on the VLPO in sleep-wake control. We then attempt to synthesize our current understanding of the circuit, cellular and synaptic bases by which the VLPO both regulates and is itself regulated, in order to exert a powerful control over behavioral state, as well as examining data suggesting an involvement of the VLPO in other physiological processes.
Collapse
|
16
|
Kostin A, Alam MA, McGinty D, Alam MN. Adult hypothalamic neurogenesis and sleep-wake dysfunction in aging. Sleep 2021; 44:5986548. [PMID: 33202015 DOI: 10.1093/sleep/zsaa173] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/22/2020] [Indexed: 12/21/2022] Open
Abstract
In the mammalian brain, adult neurogenesis has been extensively studied in the hippocampal sub-granular zone and the sub-ventricular zone of the anterolateral ventricles. However, growing evidence suggests that new cells are not only "born" constitutively in the adult hypothalamus, but many of these cells also differentiate into neurons and glia and serve specific functions. The preoptic-hypothalamic area plays a central role in the regulation of many critical functions, including sleep-wakefulness and circadian rhythms. While a role for adult hippocampal neurogenesis in regulating hippocampus-dependent functions, including cognition, has been extensively studied, adult hypothalamic neurogenic process and its contributions to various hypothalamic functions, including sleep-wake regulation are just beginning to unravel. This review is aimed at providing the current understanding of the hypothalamic adult neurogenic processes and the extent to which it affects hypothalamic functions, including sleep-wake regulation. We propose that hypothalamic neurogenic processes are vital for maintaining the proper functioning of the hypothalamic sleep-wake and circadian systems in the face of regulatory challenges. Sleep-wake disturbance is a frequent and challenging problem of aging and age-related neurodegenerative diseases. Aging is also associated with a decline in the neurogenic process. We discuss a hypothesis that a decrease in the hypothalamic neurogenic process underlies the aging of its sleep-wake and circadian systems and associated sleep-wake disturbance. We further discuss whether neuro-regenerative approaches, including pharmacological and non-pharmacological stimulation of endogenous neural stem and progenitor cells in hypothalamic neurogenic niches, can be used for mitigating sleep-wake and other hypothalamic dysfunctions in aging.
Collapse
Affiliation(s)
- Andrey Kostin
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA
| | - Md Aftab Alam
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Psychiatry, University of California, Los Angeles, CA
| | - Dennis McGinty
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Psychology, University of California, Los Angeles, CA
| | - Md Noor Alam
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| |
Collapse
|
17
|
Pamboro ELS, Brown EB, Keene AC. Dietary fatty acids promote sleep through a taste-independent mechanism. GENES BRAIN AND BEHAVIOR 2020; 19:e12629. [PMID: 31845509 DOI: 10.1111/gbb.12629] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 01/28/2023]
Abstract
Consumption of foods that are high in fat contribute to obesity and metabolism-related disorders. Dietary lipids are comprised of triglycerides and fatty acids, and the highly palatable taste of dietary fatty acids promotes food consumption, activates reward centers in mammals and underlies hedonic feeding. Despite the central role of dietary fats in the regulation of food intake and the etiology of metabolic diseases, little is known about how fat consumption regulates sleep. The fruit fly, Drosophila melanogaster, provides a powerful model system for the study of sleep and metabolic traits, and flies potently regulate sleep in accordance with food availability. To investigate the effects of dietary fats on sleep regulation, we have supplemented fatty acids into the diet of Drosophila and measured their effects on sleep and activity. We found that flies fed a diet of hexanoic acid, a medium-chain fatty acid that is a by-product of yeast fermentation, slept more than flies starved on an agar diet. To assess whether dietary fatty acids regulate sleep through the taste system, we assessed sleep in flies with a mutation in the hexanoic acid receptor Ionotropic receptor 56D, which is required for fatty acid taste perception. We found that these flies also sleep more than agar-fed flies when fed a hexanoic acid diet, suggesting the sleep promoting effect of hexanoic acid is not dependent on sensory perception. Taken together, these findings provide a platform to investigate the molecular and neural basis for fatty acid-dependent modulation of sleep.
Collapse
Affiliation(s)
- Estelle L S Pamboro
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida
| | - Elizabeth B Brown
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida
| | - Alex C Keene
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida
| |
Collapse
|
18
|
Szentirmai É, Kapás L. Sleep and body temperature in TNFα knockout mice: The effects of sleep deprivation, β3-AR stimulation and exogenous TNFα. Brain Behav Immun 2019; 81:260-271. [PMID: 31220563 PMCID: PMC6754767 DOI: 10.1016/j.bbi.2019.06.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/06/2019] [Accepted: 06/16/2019] [Indexed: 01/09/2023] Open
Abstract
Increased production of pro-inflammatory cytokines is assumed to mediate increased sleep under inflammatory conditions, such as systemic infections or recovery from sleep loss. The role of cytokines in sleep regulation under normal conditions is less clear. In the present study, we investigated the role of endogenous tumor necrosis factor alpha (TNFα) in sleep regulation using TNFα knockout (KO) mice. Under control conditions at thermoneutral ambient temperature, total sleep time did not differ between TNFα KO and wild-type (WT) mice, but TNFα KO mice had increased rapid-eye-movement sleep (REMS), accompanied by decreased motor activity and body temperature. Exposure to 17 °C induced decreases in total sleep time similarly in both genotypes. Sleep deprivation by gentle handling elicited robust rebound increases in non-rapid-eye movement sleep (NREMS), REMS and electroencephalographic (EEG) slow-wave activity (SWA), accompanied by suppressed motor activity and decreased body temperature; there was no significant difference between the responses of WT and KO mice. Systemic injection of the beta3-adrenergic receptor (β3-AR) agonist CL-316,243 induced increases in NREMS and body temperature. The temperature response, but not the sleep effect, was attenuated in the KO animals. Systemic injection of TNFα induced increased NREMS, reduced REMS and biphasic temperature responses in both genotypes. In the KO mice, the NREMS-promoting effects of exogenously administered TNFα was decreased, while REMS suppression was enhanced, and the first, hypothermic, phase of temperature response was attenuated. Overall, TNFα KO mice did not show any deficiency in sleep regulation which suggests that the role of endogenous TNFα in sleep regulation is less pronounced than previously suggested.
Collapse
Affiliation(s)
- Éva Szentirmai
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, WA, USA; Sleep and Performance Research Center, Washington State University, Spokane, WA, USA.
| | - Levente Kapás
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, WA, USA; Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
| |
Collapse
|
19
|
Hou T, Wang C, Joshi S, O’Hara BF, Gong MC, Guo Z. Active Time-Restricted Feeding Improved Sleep-Wake Cycle in db/db Mice. Front Neurosci 2019; 13:969. [PMID: 31619950 PMCID: PMC6763589 DOI: 10.3389/fnins.2019.00969] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/29/2019] [Indexed: 11/13/2022] Open
Abstract
People with diabetes are more likely to experience sleep disturbance than those without. Sleep disturbance can cause daytime sleepiness in diabetic patients, which may impair their daytime performance or even lead to workplace injuries. Therefore, restoring the normal sleep-wake cycle is critical for diabetic patients who experience daytime sleepiness. Previous data on a diabetic mouse model, the db/db mice, have demonstrated that the total sleep time and sleep fragmentation are increased and the daily rhythm of the sleep-wake cycle is attenuated. Accumulating evidence has shown that active time-restricted feeding (ATRF), in which the timing of food availability is restricted to the active-phase, is beneficial to metabolic health. However, it is unknown whether ATRF restores the normal sleep-wake cycle in diabetes. To test that, we used a non-invasive piezoelectric system to monitor the sleep-wake profile in the db/db mice with ad libitum feeding (ALF) as a baseline and then followed with ATRF. The results showed that at baseline, db/db mice exhibited abnormal sleep-wake patterns: the sleep time percent during the light-phase was decreased, while during the dark-phase it was increased with unusual cycling compared to control mice. In addition, the sleep bout length during both the light-phase and the full 24-h period was shortened in db/db mice. Analysis of the sleep-wake circadian rhythm showed that ATRF effectively restored the circadian but suppressed the ultradian oscillations of the sleep-wake cycle in the db/db mice. In conclusion, ATRF may serve as a novel strategy for treating diabetes-induced irregularity of the sleep-wake cycle.
Collapse
Affiliation(s)
- Tianfei Hou
- Department of Physiology, University of Kentucky, Lexington, KY, United States
| | - Chanung Wang
- Department of Biology, University of Kentucky, Lexington, KY, United States
| | - Shreyas Joshi
- Department of Biology, University of Kentucky, Lexington, KY, United States
| | - Bruce F. O’Hara
- Department of Biology, University of Kentucky, Lexington, KY, United States
| | - Ming C. Gong
- Department of Physiology, University of Kentucky, Lexington, KY, United States
| | - Zhenheng Guo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
- Research and Development, Lexington Veterans Affairs Medical Center, Lexington, KY, United States
| |
Collapse
|
20
|
Kim Y, Vadodaria KC, Lenkei Z, Kato T, Gage FH, Marchetto MC, Santos R. Mitochondria, Metabolism, and Redox Mechanisms in Psychiatric Disorders. Antioxid Redox Signal 2019; 31:275-317. [PMID: 30585734 PMCID: PMC6602118 DOI: 10.1089/ars.2018.7606] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/21/2018] [Accepted: 12/23/2018] [Indexed: 12/17/2022]
Abstract
Significance: Our current knowledge of the pathophysiology and molecular mechanisms causing psychiatric disorders is modest, but genetic susceptibility and environmental factors are central to the etiology of these conditions. Autism, schizophrenia, bipolar disorder and major depressive disorder show genetic gene risk overlap and share symptoms and metabolic comorbidities. The identification of such common features may provide insights into the development of these disorders. Recent Advances: Multiple pieces of evidence suggest that brain energy metabolism, mitochondrial functions and redox balance are impaired to various degrees in psychiatric disorders. Since mitochondrial metabolism and redox signaling can integrate genetic and environmental environmental factors affecting the brain, it is possible that they are implicated in the etiology and progression of psychiatric disorders. Critical Issue: Evidence for direct links between cellular mitochondrial dysfunction and disease features are missing. Future Directions: A better understanding of the mitochondrial biology and its intracellular connections to the nuclear genome, the endoplasmic reticulum and signaling pathways, as well as its role in intercellular communication in the organism, is still needed. This review focuses on the findings that implicate mitochondrial dysfunction, the resultant metabolic changes and oxidative stress as important etiological factors in the context of psychiatric disorders. We also propose a model where specific pathophysiologies of psychiatric disorders depend on circuit-specific impairments of mitochondrial dysfunction and redox signaling at specific developmental stages.
Collapse
Affiliation(s)
- Yeni Kim
- Department of Child and Adolescent Psychiatry, National Center for Mental Health, Seoul, South Korea
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
| | - Krishna C. Vadodaria
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
| | - Zsolt Lenkei
- Laboratory of Dynamic of Neuronal Structure in Health and Disease, Institute of Psychiatry and Neuroscience of Paris (UMR_S1266 INSERM, University Paris Descartes), Paris, France
| | - Tadafumi Kato
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain Science, Wako, Japan
| | - Fred H. Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
| | - Maria C. Marchetto
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
| | - Renata Santos
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
- Laboratory of Dynamic of Neuronal Structure in Health and Disease, Institute of Psychiatry and Neuroscience of Paris (UMR_S1266 INSERM, University Paris Descartes), Paris, France
| |
Collapse
|
21
|
Fleury Curado T, Pho H, Berger S, Caballero-Eraso C, Shin MK, Sennes LU, Pham L, Schwartz AR, Polotsky VY. Sleep-disordered breathing in C57BL/6J mice with diet-induced obesity. Sleep 2019; 41:4996050. [PMID: 29767787 DOI: 10.1093/sleep/zsy089] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Indexed: 01/19/2023] Open
Abstract
Obesity leads to sleep-disordered breathing (SDB) manifested by recurrent upper airway obstructions termed obstructive sleep apnea (OSA) and carbon dioxide retention due to hypoventilation. The objective of this work was to characterize breathing during sleep in C57BL6/J mice with diet-induced obesity (DIO). Arterial blood gas was measured in nine obese and nine lean mice during wakefulness. Nine male mice with DIO and six lean male C57BL/6J mice were head mounted with electroencephalogram (EEG) and electromyogram (EMG) electrodes. Sleep recordings were performed in the whole body plethysmography chamber; upper airway obstruction was characterized by the presence of inspiratory flow limitation in which airflow plateaus with increases in inspiratory effort. Obese mice showed significantly lower pH and higher partial pressure of arterial CO2 (PaCO2) in arterial blood gas compared to lean mice, 7.35 ± 0.04 versus 7.46 ± 0.06 (p < 0.001) and 38 ± 8 mm Hg versus 30 ± 5 mm Hg (p < 0.001). Obese mice had similar levels of minute ventilation to lean mice during sleep and wakefulness, despite higher body weight and temperature, indicating an increase in the metabolic rate and hypoventilation. Obese mice also showed baseline hypoxemia with decreased mean oxyhemoglobin saturation across sleep/wake states. Obese mice had a higher prevalence of flow-limited breathing compared to lean mice during sleep. However, the oxygen desaturation index in lean and obese mice did not differ. We conclude that DIO in mice leads to hypoventilation. Obesity also increases the frequency of inspiratory limited breaths, but it does not translate into progression of OSA.
Collapse
Affiliation(s)
- Thomaz Fleury Curado
- Division of Pulmonary and Critical Care Medicine, Department of Medicine Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Otolaryngology, University of Sao Paulo, Sao Paulo, Brazil
| | - Huy Pho
- Division of Pulmonary and Critical Care Medicine, Department of Medicine Johns Hopkins University School of Medicine, Baltimore, MD
| | - Slava Berger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine Johns Hopkins University School of Medicine, Baltimore, MD
| | - Candela Caballero-Eraso
- Division of Pulmonary and Critical Care Medicine, Department of Medicine Johns Hopkins University School of Medicine, Baltimore, MD.,Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Mi-Kyung Shin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Luu Pham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alan R Schwartz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine Johns Hopkins University School of Medicine, Baltimore, MD
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
22
|
Panagiotou M, Deboer T. Chronic high-caloric diet accentuates age-induced sleep alterations in mice. Behav Brain Res 2019; 362:131-139. [PMID: 30639608 DOI: 10.1016/j.bbr.2019.01.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 11/15/2022]
Abstract
Obesity and sleep disturbances comprise major health problems which are likely interrelated. Diet-induced obesity in young mice has been demonstrated to lead towards an altered sleep homeostasis. In the current study, we investigated the effect of chronic (12 weeks) high-caloric diet (HCD, 45% fat) consumption on sleep and the sleep electroencephalogram (EEG) in young and older mice (6-month-old, n = 9; 18-month-old, n = 8 and 24-month-old, n = 4) and compared with age-matched controls on normal chow (n = 11, n = 9 and n = 9 respectively). Half of the 24-month-old mice did not cope well with HCD, therefore this group has a lower n and limited statistical power. We recorded EEG and the electromyogram for continuous 48-h and performed a 6-h sleep deprivation during the second day. In aged HCD fed mice (18 months old) compared to young, an aging effect was still evident, characterized by decreased waking and increased NREM sleep in the dark period, decreased REM sleep during the light period, as well as increased slow-wave-activity (SWA, EEG power in NREM sleep in 0.5-4.0 Hz). Additionally, aged HCD treated mice showed increased NREM sleep and decreased waking, compared to age-matched controls, denoting an enhanced aging phenotype in the sleep architecture. Notably, an overall increase was found in the slow component of SWA (0.5-2.5 Hz) in aged HCD fed mice compared to age-matched controls. Our data suggest that the effect of aging is the dominant variable irrespective of diet. However, a synergistic effect of aging and diet is noted indicating that chronic HCD consumption exacerbates age-associated sleep alterations.
Collapse
Affiliation(s)
- M Panagiotou
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, the Netherlands
| | - T Deboer
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
23
|
Shin A, Woo J, Kim JE, Kim D. Nodding behavior couples to vigilance fluctuation in a high-calorie diet model of drowsiness. Mol Brain 2018; 11:33. [PMID: 29880005 PMCID: PMC5992632 DOI: 10.1186/s13041-018-0377-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/30/2018] [Indexed: 12/18/2022] Open
Abstract
Drowsiness is an awake state with increased sleep drive, yet the neural correlates and underlying mechanisms remains unclear. Here, we established a mouse model of drowsiness, where mice are fasted for 1 day and then allowed to overeat high-fat food (to promote sleep) while positioned in an open-field box (to promote vigilance). They fall into a long-lasting drowsy state, as reflected by repeated and open-eyed nodding of the head while in a standing position. Simultaneous recording of electroencephalogram (EEG) and neck electromyogram (EMG) readouts revealed that this drowsy state including nodding state had multiple stages in terms of the relationship between the level of vigilance and head movement: delta oscillations decreased in power prior to the head-nodding period and increased during the non-nodding period. Cav3.1-knockout mice, which have reduced delta oscillations, showed frequent head nodding with reduced duration of nodding episodes compared to wild-type mice. This suggests that the balance of drive is tilted in favor of wakefulness, likely due to their previously proposed decrease in sleep-promoting functions. Our findings indicate that delta oscillations play a dominant role in controlling vigilance dynamics during sleep/wake competition and that our novel mouse model may be useful for studying drowsiness and related neurological disorders.
Collapse
Affiliation(s)
- Anna Shin
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jeonghoon Woo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.,Chungnam Techno Park, Cheonan, 31035, Republic of Korea
| | - Jung Eun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Daesoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
24
|
Panagiotou M, Meijer JH, Deboer T. Chronic high-caloric diet modifies sleep homeostasis in mice. Eur J Neurosci 2018; 47:1339-1352. [PMID: 29737605 DOI: 10.1111/ejn.13932] [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: 06/16/2017] [Revised: 03/13/2018] [Accepted: 03/15/2018] [Indexed: 11/29/2022]
Abstract
Obesity prevalence and sleep habit changes are commonplace nowadays, due to modern lifestyle. A bidirectional relationship likely exists between sleep quality and metabolic disruptions, which could impact quality of life. In our study, we investigated the effects of a chronic high-caloric diet on sleep architecture and sleep regulation in mice. We studied the effect of 3 months high-caloric diet (HCD, 45% fat) on sleep and the sleep electroencephalogram (EEG) in C57BL/6J mice during 24-hr baseline (BL) recordings, and after 6-hr sleep deprivation (SD). We examined the effect of HCD on sleep homeostasis, by performing parameter estimation analysis and simulations of the sleep homeostatic Process S, a measure of sleep pressure, which is reflected in the non-rapid-eye-movement (NREM) sleep slow-wave-activity (SWA, EEG power density between 0.5 and 4.0 Hz). Compared to controls (n = 11, 30.7 ± 0.8 g), mice fed with HCD (n = 9, 47.6 ± 0.8 g) showed an increased likelihood of consecutive NREM-REM sleep cycles, increased REM sleep and decreased NREM sleep EEG SWA. After SD, these effects were more pronounced. The simulation resulted in a close fit between the time course of SWA and Process S in both groups. HCD fed mice had a slower time constant (Ti = 15.98 hr) for the increase in homeostatic sleep pressure compared with controls (5.95 hr) indicating a reduced effect of waking on the increase in sleep pressure. Our results suggest that chronic HCD consumption impacts sleep regulation.
Collapse
Affiliation(s)
- Maria Panagiotou
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Johanna H Meijer
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Tom Deboer
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
25
|
Eban-Rothschild A, Appelbaum L, de Lecea L. Neuronal Mechanisms for Sleep/Wake Regulation and Modulatory Drive. Neuropsychopharmacology 2018; 43:937-952. [PMID: 29206811 PMCID: PMC5854814 DOI: 10.1038/npp.2017.294] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 11/17/2017] [Accepted: 11/24/2017] [Indexed: 12/17/2022]
Abstract
Humans have been fascinated by sleep for millennia. After almost a century of scientific interrogation, significant progress has been made in understanding the neuronal regulation and functions of sleep. The application of new methods in neuroscience that enable the analysis of genetically defined neuronal circuits with unprecedented specificity and precision has been paramount in this endeavor. In this review, we first discuss electrophysiological and behavioral features of sleep/wake states and the principal neuronal populations involved in their regulation. Next, we describe the main modulatory drives of sleep and wakefulness, including homeostatic, circadian, and motivational processes. Finally, we describe a revised integrative model for sleep/wake regulation.
Collapse
Affiliation(s)
| | - Lior Appelbaum
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| |
Collapse
|
26
|
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
|
27
|
Linehan V, Fang LZ, Hirasawa M. Short-term high-fat diet primes excitatory synapses for long-term depression in orexin neurons. J Physiol 2017; 596:305-316. [PMID: 29143330 DOI: 10.1113/jp275177] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/09/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS High-fat diet consumption is a major cause of obesity. Orexin neurons are known to be activated by a high-fat diet and in turn promote further consumption of a high-fat diet. Our study shows that excitatory synapses to orexin neurons become amenable to long-term depression (LTD) after 1 week of high-fat diet feeding. However, this effect reverses after 4 weeks of a high-fat diet. This LTD may be a homeostatic response to a high-fat diet to curb the activity of orexin neurons and hence caloric consumption. Adaptation seen after prolonged high-fat diet intake may contribute to the development of obesity. ABSTRACT Overconsumption of high-fat diets is one of the strongest contributing factors to the rise of obesity rates. Orexin neurons are known to be activated by a palatable high-fat diet and mediate the activation of the mesolimbic reward pathway, resulting in further food intake. While short-term exposure to a high-fat diet is known to induce synaptic plasticity within the mesolimbic pathway, it is unknown if such changes occur in orexin neurons. To investigate this, 3-week-old male rats were fed a palatable high-fat western diet (WD) or control chow for 1 week and then in vitro patch clamp recording was performed. In the WD condition, an activity-dependent long-term depression (LTD) of excitatory synapses was observed in orexin neurons, but not in chow controls. This LTD was presynaptic and depended on postsynaptic metabotropic glutamate receptor 5 (mGluR5) and retrograde endocannabinoid signalling. WD also increased extracellular glutamate levels, suggesting that glutamate spillover and subsequent activation of perisynaptic mGluR5 may occur more readily in the WD condition. In support of this, pharmacological inhibition of glutamate uptake was sufficient to prime chow control synapses to undergo a presynaptic LTD. Interestingly, these WD effects are transient, as extracellular glutamate levels were similar to controls and LTD was no longer observed in orexin neurons after 4 weeks of WD. In summary, excitatory synapses to orexin neurons become amenable to LTD under a palatable high-fat diet, which may represent a homeostatic mechanism to prevent overactivation of these neurons and to curtail high-fat diet consumption.
Collapse
Affiliation(s)
- Victoria Linehan
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada, A1B 3V6
| | - Lisa Z Fang
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada, A1B 3V6
| | - Michiru Hirasawa
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada, A1B 3V6
| |
Collapse
|
28
|
Restoring Serotonergic Homeostasis in the Lateral Hypothalamus Rescues Sleep Disturbances Induced by Early-Life Obesity. J Neurosci 2017; 38:441-451. [PMID: 29196316 DOI: 10.1523/jneurosci.1333-17.2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/13/2017] [Accepted: 10/12/2017] [Indexed: 01/09/2023] Open
Abstract
Early-life obesity predisposes to obesity in adulthood, a condition with broad medical implications including sleep disorders, which can exacerbate metabolic disturbances and disrupt cognitive and affective behaviors. In this study, we examined the long-term impact of transient peripubertal diet-induced obesity (ppDIO, induced between 4 and 10 weeks of age) on sleep-wake behavior in male mice. EEG and EMG recordings revealed that ppDIO increases sleep during the active phase but reduces resting-phase sleep quality. This impaired sleep phenotype persisted for up to 1 year, although animals were returned to a non-obesiogenic diet from postnatal week 11 onwards. To better understand the mechanisms responsible for the ppDIO-induced alterations in sleep, we focused on the lateral hypothalamus (LH). Mice exposed to ppDIO did not show altered mRNA expression levels of orexin and melanin-concentrating hormone, two peptides that are important for sleep-wake behavior and food intake. Conversely, the LH of ppDIO-exposed mice had reduced contents of serotonin (5-hydroxytryptamine, 5-HT), a neurotransmitter involved in both sleep-wake and satiety regulation. Interestingly, an acute peripheral injection of the satiety-signaling peptide YY 3-36 increased 5-HT turnover in the LH and ameliorated the ppDIO-induced sleep disturbances, suggesting the therapeutic potential of this peptide. These findings provide new insights into how sleep-wake behavior is programmed during early life and how peripheral and central signals are integrated to coordinate sleep.SIGNIFICANCE STATEMENT Adult physiology and behavior are strongly influenced by dynamic reorganization of the brain during puberty. The present work shows that obesity during puberty leads to persistently dysregulated patterns of sleep and wakefulness by blunting serotonergic signaling in the lateral hypothalamus. It also shows that pharmacological mimicry of satiety with peptide YY3-36 can reverse this neurochemical imbalance and acutely restore sleep composition. These findings add insight into how innate behaviors such as feeding and sleep are integrated and suggest a novel mechanism through which diet-induced obesity during puberty imposes its long-lasting effects on sleep-wake behavior.
Collapse
|
29
|
Sasaki T. Neural and Molecular Mechanisms Involved in Controlling the Quality of Feeding Behavior: Diet Selection and Feeding Patterns. Nutrients 2017; 9:nu9101151. [PMID: 29053636 PMCID: PMC5691767 DOI: 10.3390/nu9101151] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 12/20/2022] Open
Abstract
We are what we eat. There are three aspects of feeding: what, when, and how much. These aspects represent the quantity (how much) and quality (what and when) of feeding. The quantitative aspect of feeding has been studied extensively, because weight is primarily determined by the balance between caloric intake and expenditure. In contrast, less is known about the mechanisms that regulate the qualitative aspects of feeding, although they also significantly impact the control of weight and health. However, two aspects of feeding quality relevant to weight loss and weight regain are discussed in this review: macronutrient-based diet selection (what) and feeding pattern (when). This review covers the importance of these two factors in controlling weight and health, and the central mechanisms that regulate them. The relatively limited and fragmented knowledge on these topics indicates that we lack an integrated understanding of the qualitative aspects of feeding behavior. To promote better understanding of weight control, research efforts must focus more on the mechanisms that control the quality and quantity of feeding behavior. This understanding will contribute to improving dietary interventions for achieving weight control and for preventing weight regain following weight loss.
Collapse
Affiliation(s)
- Tsutomu Sasaki
- Laboratory for Metabolic Signaling, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma 371-8512, Japan.
| |
Collapse
|
30
|
Luppi M, Al-Jahmany AA, Del Vecchio F, Cerri M, Di Cristoforo A, Hitrec T, Martelli D, Perez E, Zamboni G, Amici R. Wake-sleep and cardiovascular regulatory changes in rats made obese by a high-fat diet. Behav Brain Res 2017; 320:347-355. [PMID: 28011172 DOI: 10.1016/j.bbr.2016.12.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 12/15/2016] [Accepted: 12/19/2016] [Indexed: 11/18/2022]
Abstract
Obesity is known to be associated with alterations in wake-sleep (WS) architecture and cardiovascular parameters. This study was aimed at assessing the possible influence of diet-induced obesity (DIO) on sleep homeostasis and on the WS state-dependent levels of arterial pressure (AP) and heart rate in the rat. Two groups of age-matched Sprague-Dawley rats were fed either a high-fat hypercaloric diet, leading to DIO, or a normocaloric standard diet (lean controls) for 8 weeks. While under general anesthesia, animals were implanted with instrumentation for the recording of electroencephalogram, electromyogram, arterial pressure, and deep brain temperature. The experimental protocol consisted of 48h of baseline, 12h of gentle handling, enhancing wake and depressing sleep, and 36-h post-handling recovery. Compared to lean controls, DIO rats showed: i) the same amount of rapid-eye movement (REM) and non-REM (NREM) sleep in the rest period, although the latter was characterized by more fragmented episodes; ii) an increase in both REM sleep and NREM sleep in the activity period; iii) a comparable post-handling sleep homeostatic response, in terms of either the degree of Delta power increase during NREM sleep or the quantitative compensation of the REM sleep loss at the end of the 36-h recovery period; iv) significantly higher levels of AP, irrespectively of the different WS states and of the changes in their intensity throughout the experimental protocol. Overall, these changes may be the reflection of a modification in the activity of the hypothalamic areas where WS, autonomic, and metabolic regulations are known to interact.
Collapse
Affiliation(s)
- Marco Luppi
- Department of Biomedical and Neuromotor Sciences-Physiology, Alma Mater Studiorum - University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy.
| | - Abed A Al-Jahmany
- Department of Biomedical and Neuromotor Sciences-Physiology, Alma Mater Studiorum - University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy.
| | - Flavia Del Vecchio
- Department of Biomedical and Neuromotor Sciences-Physiology, Alma Mater Studiorum - University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy.
| | - Matteo Cerri
- Department of Biomedical and Neuromotor Sciences-Physiology, Alma Mater Studiorum - University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy.
| | - Alessia Di Cristoforo
- Department of Biomedical and Neuromotor Sciences-Physiology, Alma Mater Studiorum - University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy.
| | - Timna Hitrec
- Department of Biomedical and Neuromotor Sciences-Physiology, Alma Mater Studiorum - University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy.
| | - Davide Martelli
- Department of Biomedical and Neuromotor Sciences-Physiology, Alma Mater Studiorum - University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, Australia.
| | - Emanuele Perez
- Department of Biomedical and Neuromotor Sciences-Physiology, Alma Mater Studiorum - University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy.
| | - Giovanni Zamboni
- Department of Biomedical and Neuromotor Sciences-Physiology, Alma Mater Studiorum - University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy.
| | - Roberto Amici
- Department of Biomedical and Neuromotor Sciences-Physiology, Alma Mater Studiorum - University of Bologna, Piazza di Porta San Donato, 2, 40126, Bologna, Italy.
| |
Collapse
|
31
|
Blancas-Velazquez A, Mendoza J, Garcia AN, la Fleur SE. Diet-Induced Obesity and Circadian Disruption of Feeding Behavior. Front Neurosci 2017; 11:23. [PMID: 28223912 PMCID: PMC5293780 DOI: 10.3389/fnins.2017.00023] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/12/2017] [Indexed: 12/13/2022] Open
Abstract
Feeding behavior shows a rhythmic daily pattern, which in nocturnal rodents is observed mainly during the dark period. This rhythmicity is under the influence of the hypothalamic suprachiasmatic nucleus (SCN), the main biological clock. Nevertheless, various studies have shown that in rodent models of obesity, using high-energy diets, the general locomotor activity and feeding rhythms can be disrupted. Here, we review the data on the effects of diet-induced obesity (DIO) on locomotor activity and feeding patterns, as well as the effect on the brain sites within the neural circuitry involved in metabolic and rewarding feeding behavior. In general, DIO may alter locomotor activity by decreasing total activity. On the other hand, DIO largely alters eating patterns, producing increased overall ingestion and number of eating bouts that can extend to the resting period. Furthermore, within the hypothalamic areas, little effect has been reported on the molecular circadian mechanism in DIO animals with ad libitum hypercaloric diets and little or no data exist so far on its effects on the reward system areas. We further discuss the possibility of an uncoupling of metabolic and reward systems in DIO and highlight a gap of circadian and metabolic research that may help to better understand the implications of obesity.
Collapse
Affiliation(s)
- Aurea Blancas-Velazquez
- Institute of Cellular and Integrative Neurosciences, Centre National de la Recherche Scientifique UPR-3212, University of StrasbourgStrasbourg, France; Department of Endocrinology and Metabolism, Academic Medical Center, University of AmsterdamAmsterdam, Netherlands; Metabolism and Reward Group, Netherlands Institute for NeuroscienceAmsterdam, Netherlands
| | - Jorge Mendoza
- Institute of Cellular and Integrative Neurosciences, Centre National de la Recherche Scientifique UPR-3212, University of Strasbourg Strasbourg, France
| | - Alexandra N Garcia
- Department of Endocrinology and Metabolism, Academic Medical Center, University of AmsterdamAmsterdam, Netherlands; Metabolism and Reward Group, Netherlands Institute for NeuroscienceAmsterdam, Netherlands
| | - Susanne E la Fleur
- Department of Endocrinology and Metabolism, Academic Medical Center, University of AmsterdamAmsterdam, Netherlands; Metabolism and Reward Group, Netherlands Institute for NeuroscienceAmsterdam, Netherlands
| |
Collapse
|
32
|
Tsuneki H, Sasaoka T, Sakurai T. Sleep Control, GPCRs, and Glucose Metabolism. Trends Endocrinol Metab 2016; 27:633-642. [PMID: 27461005 DOI: 10.1016/j.tem.2016.06.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/29/2016] [Accepted: 06/29/2016] [Indexed: 12/29/2022]
Abstract
Modern lifestyles prolong daily activities into the nighttime, disrupting circadian rhythms, which may cause sleep disturbances. Sleep disturbances have been implicated in the dysregulation of blood glucose levels and reported to increase the risk of type 2 diabetes (T2D) and diabetic complications. Sleep disorders are treated using anti-insomnia drugs that target ionotropic and G protein-coupled receptors (GPCRs), including γ-aminobutyric acid (GABA) agonists, melatonin agonists, and orexin receptor antagonists. A deeper understanding of the effects of these medications on glucose metabolism and their underlying mechanisms of action is crucial for the treatment of diabetic patients with sleep disorders. In this review we focus on the beneficial impact of sleep on glucose metabolism and suggest a possible strategy for therapeutic intervention against sleep-related metabolic disorders.
Collapse
Affiliation(s)
- Hiroshi Tsuneki
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Toshiyasu Sasaoka
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Takeshi Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
33
|
Abstract
Sleep and its disorders are known to affect the functions of essential organs and systems in the body. However, very little is known about how the blood-brain barrier (BBB) is regulated. A few years ago, we launched a project to determine the impact of sleep fragmentation and chronic sleep restriction on BBB functions, including permeability to fluorescent tracers, tight junction protein expression and distribution, glucose and other solute transporter activities, and mediation of cellular mechanisms. Recent publications and relevant literature allow us to summarize here the sleep-BBB interactions in five sections: (1) the structural basis enabling the BBB to serve as a huge regulatory interface; (2) BBB transport and permeation of substances participating in sleep-wake regulation; (3) the circadian rhythm of BBB function; (4) the effect of experimental sleep disruption maneuvers on BBB activities, including regional heterogeneity, possible threshold effect, and reversibility; and (5) implications of sleep disruption-induced BBB dysfunction in neurodegeneration and CNS autoimmune diseases. After reading the review, the general audience should be convinced that the BBB is an important mediating interface for sleep-wake regulation and a crucial relay station of mind-body crosstalk. The pharmaceutical industry should take into consideration that sleep disruption alters the pharmacokinetics of BBB permeation and CNS drug delivery, being attentive to the chrono timing and activation of co-transporters in subjects with sleep disorders.
Collapse
Affiliation(s)
- Weihong Pan
- 1 Biopotentials Sleep Center, Baton Rouge, LA 70809
| | - Abba J Kastin
- 2 Blood-Brain Barrier Group, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| |
Collapse
|
34
|
Effects of sleep disruption and high fat intake on glucose metabolism in mice. Psychoneuroendocrinology 2016; 68:47-56. [PMID: 26943344 PMCID: PMC4851877 DOI: 10.1016/j.psyneuen.2016.02.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 02/22/2016] [Accepted: 02/22/2016] [Indexed: 11/23/2022]
Abstract
Poor sleep quality or quantity impairs glycemic control and increases risk of disease under chronic conditions. Recovery sleep may offset adverse metabolic outcomes of accumulated sleep debt, but the extent to which this occurs is unclear. We examined whether recovery sleep improves glucose metabolism in mice subjected to prolonged sleep disruption, and whether high fat intake during sleep disruption exacerbates glycemic control. Adult male C57BL/6J mice were subjected to 18-h sleep fragmentation daily for 9 days, followed by 1 day of recovery. During sleep disruption, one group of mice was fed a high-fat diet (HFD) while another group was fed standard laboratory chow. Insulin sensitivity and glucose tolerance were assessed by insulin and glucose tolerance testing at baseline, after 3 and 7 days of sleep disruption, and at the end of the protocol after 24h of undisturbed sleep opportunity (recovery). To characterize changes in sleep architecture that are associated with sleep debt and recovery, we quantified electroencephalogram (EEG) recordings during sleep fragmentation and recovery periods from an additional group of mice. We now report that 9 days of 18-h daily sleep fragmentation significantly reduces rapid eye movement sleep (REMS) and non-rapid eye movement sleep (NREMS). Mice respond with increases in REMS, but not NREMS, during the daily 6-h undisturbed sleep opportunity. However, both REMS and NREMS increase significantly during the 24-h recovery period. Although sleep disruption alone has no effect in this protocol, high fat feeding in combination with sleep disruption impairs glucose tolerance, effects that are reversed by recovery sleep. Insulin sensitivity modestly improves after 3 days of sleep fragmentation and after 24h of recovery, with significantly greater improvements in mice exposed to HFD during sleep disruption. Improvements in both glucose tolerance and insulin sensitivity are associated with NREMS rebound, raising the possibility that this sleep phase contributes to restorative effects of recovery sleep on glycemic control.
Collapse
|
35
|
Associations between Macronutrient Intake and Obstructive Sleep Apnoea as Well as Self-Reported Sleep Symptoms: Results from a Cohort of Community Dwelling Australian Men. Nutrients 2016; 8:207. [PMID: 27070639 PMCID: PMC4848676 DOI: 10.3390/nu8040207] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 01/08/2023] Open
Abstract
Background: macronutrient intake has been found to affect sleep parameters including obstructive sleep apnoea (OSA) in experimental studies, but there is uncertainty at the population level in adults. Methods: cross-sectional analysis was conducted of participants in the Men Androgen Inflammation Lifestyle Environment and Stress cohort (n = 784, age 35–80 years). Dietary intake was measured by a validated food frequency questionnaire. Self-reported poor sleep quality and daytime sleepiness were measured by questionnaires. Overnight in-home polysomnography (PSG) was conducted among participants with without previously diagnosed OSA. Results: after adjusting for demographic, lifestyle factors, and chronic diseases, the highest quartile of fat intake was positively associated with excessive daytime sleepiness (relative risk ratio (RRR) = 1.78, 95% CI 1.10, 2.89) and apnoea-hypopnoea index (AHI) ≥20, (RRR = 2.98, 95% CI 1.20–7.38). Body mass index mediated the association between fat intake and AHI (30%), but not daytime sleepiness. There were no associations between other intake of macronutrient and sleep outcomes. Conclusion: high fat is associated with daytime sleepiness and AHI. Sleep outcomes are generally not assessed in studies investigating the effects of varying macronutrient diets on weight loss. The current result highlights the potential public health significance of doing so.
Collapse
|
36
|
Gomez-Peralta F, Abreu C, Castro JC, Alcarria E, Cruz-Bravo M, Garcia-Llorente MJ, Albornos C, Moreno C, Cepeda M, Almodóvar F. An association between liraglutide treatment and reduction in excessive daytime sleepiness in obese subjects with type 2 diabetes. BMC Endocr Disord 2015; 15:78. [PMID: 26637348 PMCID: PMC4669657 DOI: 10.1186/s12902-015-0074-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 11/30/2015] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The main purpose of the present study is to evaluate whether treatment with long-acting human glucagon-like peptide-1 liraglutide was associated with an improvement of excessive daytime sleepiness (EDS) in obese subjects with type-2 diabetes. METHODS This single-centre retrospective study included 158 obese (body mass index [BMI] ≥ 30 kg/m(2)) adult subjects with type-2 diabetes who were initiated with liraglutide treatment at least 3 months before study inclusion. Data of the Epworth Sleepiness Scale (ESS), anthropometric parameters, glucose-control and metabolic parameters were collected at liraglutide initiation (baseline) and at months 1 and 3 after liraglutide initiation. RESULTS Significant reductions in ESS score were achieved at months 1 (-1.3 ± 2.8, p < 0.001) and 3 (-1.5 ± 3.0, p < 0.001) after liraglutide introduction. After 3 months of treatment with liraglutide, significant changes in body weight (p < 0.001), BMI (p < 0.001), waist (p < 0.001) and neck circumferences (p < 0.005), HbA1c (p < 0.001), mean blood glucose (p < 0.001), fasting plasma glucose (p < 0.001), triglycerides (p < 0.01) and total cholesterol (p < 0.001) were achieved. CONCLUSIONS After 3 months of treatment with liraglutide a significant reduction in EDS was observed in obese subjects with type-2 diabetes. Besides this, significant changes in body weight and metabolic parameters of diabetes control were also accomplished. Further investigation is required to determine whether liraglutide could improve other abnormal sleep patterns and obstructive sleep apnoea.
Collapse
Affiliation(s)
- Fernando Gomez-Peralta
- Unidad de Endocrinología y Nutrición, Hospital General de Segovia, C/ Miguel Servet s/n, 40002, Segovia, Spain.
| | - Cristina Abreu
- Unidad de Endocrinología y Nutrición, Hospital General de Segovia, C/ Miguel Servet s/n, 40002, Segovia, Spain
| | - Jose Carlos Castro
- Unidad de Endocrinología y Nutrición, Hospital General de Segovia, C/ Miguel Servet s/n, 40002, Segovia, Spain
| | - Elvira Alcarria
- Unidad de Endocrinología y Nutrición, Hospital General de Segovia, C/ Miguel Servet s/n, 40002, Segovia, Spain
| | - Margarita Cruz-Bravo
- Unidad de Endocrinología y Nutrición, Hospital General de Segovia, C/ Miguel Servet s/n, 40002, Segovia, Spain
| | - Maria Jesús Garcia-Llorente
- Unidad de Endocrinología y Nutrición, Hospital General de Segovia, C/ Miguel Servet s/n, 40002, Segovia, Spain
| | - Cristina Albornos
- Unidad de Endocrinología y Nutrición, Hospital General de Segovia, C/ Miguel Servet s/n, 40002, Segovia, Spain
| | - Concepción Moreno
- Unidad de Endocrinología y Nutrición, Hospital General de Segovia, C/ Miguel Servet s/n, 40002, Segovia, Spain
| | - María Cepeda
- Servicio de Medicina Interna, Hospital General de Segovia, Segovia, Spain
| | | |
Collapse
|
37
|
Chronic Powder Diet After Weaning Induces Sleep, Behavioral, Neuroanatomical, and Neurophysiological Changes in Mice. PLoS One 2015; 10:e0143909. [PMID: 26630494 PMCID: PMC4668096 DOI: 10.1371/journal.pone.0143909] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/11/2015] [Indexed: 12/23/2022] Open
Abstract
The purpose of this study is to clarify the effects of chronic powder diet feeding on sleep patterns and other physiological/anatomical changes in mice. C57BL/6 male mice were divided into two groups from weaning: a group fed with solid food (SD) and a group fed with powder food (PD), and sleep and physiological and anatomical changes were compared between the groups. PD exhibited less cranial bone structure development and a significant weight gain. Furthermore, these PD mice showed reduced number of neurogenesis in the hippocampus. Sleep analysis showed that PD induced attenuated diurnal sleep/wake rhythm, characterized by increased sleep during active period and decreased sleep during rest period. With food deprivation (FD), PD showed less enhancement of wake/locomotor activity compared to SD, indicating reduced food-seeking behavior during FD. These results suggest that powder feeding in mice results in a cluster of detrimental symptoms caused by abnormal energy metabolism and anatomical/neurological changes.
Collapse
|
38
|
Perron IJ, Pack AI, Veasey S. Diet/Energy Balance Affect Sleep and Wakefulness Independent of Body Weight. Sleep 2015; 38:1893-903. [PMID: 26158893 DOI: 10.5665/sleep.5236] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/03/2015] [Indexed: 12/19/2022] Open
Abstract
STUDY OBJECTIVES Excessive daytime sleepiness commonly affects obese people, even in those without sleep apnea, yet its causes remain uncertain. We sought to determine whether acute dietary changes could induce or rescue wake impairments independent of body weight. DESIGN We implemented a novel feeding paradigm that generates two groups of mice with equal body weight but opposing energetic balance. Two subsets of mice consuming either regular chow (RC) or high-fat diet (HFD) for 8 w were switched to the opposite diet for 1 w. Sleep recordings were conducted at Week 0 (baseline), Week 8 (pre-diet switch), and Week 9 (post-diet switch) for all groups. Sleep homeostasis was measured at Week 8 and Week 9. PARTICIPANTS Young adult, male C57BL/6J mice. MEASUREMENTS AND RESULTS Differences in total wake, nonrapid eye movement (NREM), and rapid eye movement (REM) time were quantified, in addition to changes in bout fragmentation/consolidation. At Week 9, the two diet switch groups had similar body weight. However, animals switched to HFD (and thus gaining weight) had decreased wake time, increased NREM sleep time, and worsened sleep/wake fragmentation compared to mice switched to RC (which were in weight loss). These effects were driven by significant sleep/wake changes induced by acute dietary manipulations (Week 8 → Week 9). Sleep homeostasis, as measured by delta power increase following sleep deprivation, was unaffected by our feeding paradigm. CONCLUSIONS Acute dietary manipulations are sufficient to alter sleep and wakefulness independent of body weight and without effects on sleep homeostasis.
Collapse
Affiliation(s)
- Isaac J Perron
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Allan I Pack
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sigrid Veasey
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
39
|
Glucose Induces Slow-Wave Sleep by Exciting the Sleep-Promoting Neurons in the Ventrolateral Preoptic Nucleus: A New Link between Sleep and Metabolism. J Neurosci 2015; 35:9900-11. [PMID: 26156991 DOI: 10.1523/jneurosci.0609-15.2015] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Sleep-active neurons located in the ventrolateral preoptic nucleus (VLPO) play a crucial role in the induction and maintenance of slow-wave sleep (SWS). However, the cellular and molecular mechanisms responsible for their activation at sleep onset remain poorly understood. Here, we test the hypothesis that a rise in extracellular glucose concentration in the VLPO can promote sleep by increasing the activity of sleep-promoting VLPO neurons. We find that infusion of a glucose concentration into the VLPO of mice promotes SWS and increases the density of c-Fos-labeled neurons selectively in the VLPO. Moreover, we show in patch-clamp recordings from brain slices that VLPO neurons exhibiting properties of sleep-promoting neurons are selectively excited by glucose within physiological range. This glucose-induced excitation implies the catabolism of glucose, leading to a closure of ATP-sensitive potassium (KATP) channels. The extracellular glucose concentration monitors the gating of KATP channels of sleep-promoting neurons, highlighting that these neurons can adapt their excitability according to the extracellular energy status. Together, these results provide evidence that glucose may participate in the mechanisms of SWS promotion and/or consolidation. SIGNIFICANCE STATEMENT Although the brain circuitry underlying vigilance states is well described, the molecular mechanisms responsible for sleep onset remain largely unknown. Combining in vitro and in vivo experiments, we demonstrate that glucose likely contributes to sleep onset facilitation by increasing the excitability of sleep-promoting neurons in the ventrolateral preoptic nucleus (VLPO). We find here that these neurons integrate energetic signals such as ambient glucose directly to regulate vigilance states accordingly. Glucose-induced excitation of sleep-promoting VLPO neurons should therefore be involved in the drowsiness that one feels after a high-sugar meal. This novel mechanism regulating the activity of VLPO neurons reinforces the fundamental and intimate link between sleep and metabolism.
Collapse
|
40
|
Yajima K, Seya T, Iwayama K, Hibi M, Hari S, Nakashima Y, Ogata H, Omi N, Satoh M, Tokuyama K. Effects of nutrient composition of dinner on sleep architecture and energy metabolism during sleep. J Nutr Sci Vitaminol (Tokyo) 2015; 60:114-21. [PMID: 24975221 DOI: 10.3177/jnsv.60.114] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Energy metabolism and substrate oxidation during sleep correlate with sleep stage, suggesting that energy metabolism affects sleep architecture or vice versa. The aim of the present study was to examine whether changes in energy metabolism during sleep, induced by a high-carbohydrate or high-fat meal for dinner, affect sleep architecture. Ten healthy males participated in this study, sleeping 3 nonconsecutive nights in a whole-room calorimeter. The first night was scheduled as an adaptation to the experimental environment. The other 2 nights were experimental calorimetry in a balanced cross-over design with intrasubject comparisons. In each session, subjects comsumed a high carbohydrate (HCD: PFC=10 : 10 : 80) or high fat (HFD: PFC=10 : 78 : 12) meal at 2000 h and slept with a polysomnographic recording in a metabolic chamber for indirect calorimetry (0000 h to 0800 h). Slow wave sleep was decreased during the first sleep cycle and not changed during the second or third sleep cycle under HCD conditions compared with those of HFD. Energy expenditure was not affected by dietary condition but substrate oxidation reflected differences in dietary composition of the dinner during the first and second sleep cycle. The present study suggested the possibility that substrate availability during sleep affects substrate oxidation during sleep, and affects sleep architecture during the first sleep cycle.
Collapse
Affiliation(s)
- Katsuhiko Yajima
- Department of Administrative Nutrition, Faculty of Health and Nutrition, Tokyo Seiei College
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Fujimoto E, Sato-Mito N, Okita I, Uemura S, Ishimoto T, Sato K. Daily rhythm and heat shock protein expression in obese ob/ob mice. Nutr Neurosci 2014; 18:110-7. [PMID: 24621067 DOI: 10.1179/1476830513y.0000000103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES This study examined heat shock protein (HSP) 70 expression and rhythms of drinking behavior and locomotor activity in obesity, in order to clarify the involvement of HSPs in obesity-induced disturbance of circadian rhythms. METHODS C57BL/6J ob/ob mice were used as a murine model of severe obesity. Drinking behavior and locomotor activity of male C57BL/6J (control) mice and ob/ob mice were recorded with the behavioral analyzing system. HSP70 concentration in the homogenized supernatant of each tissue, including the brain, liver, and kidney, was measured by an enzyme-linked immunosorbent assay. RESULTS We observed an attenuated locomotor activity rhythm in the ob/ob mice compared with the control mice at 13 weeks of age and especially at 27 weeks of age. The drinking rhythm was little affected by obesity. HSP70 protein expression was reduced in the brain and kidney of the ob/ob mice compared with the control mice. However, HSP70 expression in the liver was not altered. DISCUSSION This study suggests that the obesity-induced reduction of HSP70 expression in the brain and kidney can be directly or indirectly associated with disturbance of rhythms of the master clock and peripheral clocks. The study provides a link between circadian rhythm and HSP expression in obesity; the disturbance of these factors may lead to the progression of metabolic disorders.
Collapse
|
42
|
Apolzan JW, Bray GA, Hamilton MT, Zderic TW, Han H, Champagne CM, Shepard D, Martin CK. Short-term overeating results in incomplete energy intake compensation regardless of energy density or macronutrient composition. Obesity (Silver Spring) 2014; 22:119-30. [PMID: 23913807 PMCID: PMC3873377 DOI: 10.1002/oby.20587] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 07/23/2013] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To evaluate the effects of overeating (140% of energy requirements) a high-fat low-energy density diet (HF/LED, 1.05 kcal/g), high-fat high-energy density diet (HF/HED, 1.60 kcal/g), and high-carbohydrate (HC) LED (1.05 kcal/g) for 2-days on subsequent 4-day energy intake (EI), activity levels, appetite, and mood. DESIGN AND METHODS Using a randomized cross-over design, energy expenditure and EI were standardized during overeating. RESULTS In 20 adults with a mean ± SD BMI of 30.7 ± 4.6 kg/m(2) , EI was not suppressed until the second day after overeating and accounted for ∼30% of the excess EI. Reductions in EI did not differ among the three diets or across days. Overeating had no effect on subsequent energy expenditure but steps/day decreased after the HC/LED and HF/HED. Sleep time was increased after the HF/HED compared to both LEDs. After overeating a HF/HED vs. HF/LED, carbohydrate cravings, hunger, prospective food consumption, and sadness increased and satisfaction, relaxation, and tranquility decreased. CONCLUSIONS Diet type, time, or their interaction had no impact on compensation over 4 days. No adaptive thermogenesis was observed. The HF/HED vs. HF/LED had detrimental effects on food cravings, appetite, and mood. These results suggest short-term overeating is associated with incomplete compensation.
Collapse
Affiliation(s)
- John W. Apolzan
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, USA
| | - George A. Bray
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, USA
| | - Marc T. Hamilton
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, USA
| | - Theodore W. Zderic
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, USA
| | - Hongmei Han
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, USA
| | | | - Desti Shepard
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, USA
| | - Corby K. Martin
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, USA
| |
Collapse
|
43
|
Chikahisa S, Shimizu N, Shiuchi T, Séi H. Ketone body metabolism and sleep homeostasis in mice. Neuropharmacology 2013; 79:399-404. [PMID: 24361452 DOI: 10.1016/j.neuropharm.2013.12.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 12/05/2013] [Accepted: 12/06/2013] [Indexed: 02/06/2023]
Abstract
A link has been established between energy metabolism and sleep homeostasis. The ketone bodies acetoacetate and β-hydroxybutyrate, generated from the breakdown of fatty acids, are major metabolic fuels for the brain under conditions of low glucose availability. Ketogenesis is modulated by the activity of peroxisome proliferator-activated receptor alpha (PPARα), and treatment with a PPAR activator has been shown to induce a marked increase in plasma acetoacetate and decreased β-hydroxybutyrate in mice, accompanied by increased slow-wave activity during non-rapid eye movement (NREM) sleep. The present study investigated the role of ketone bodies in sleep regulation. Six-hour sleep deprivation increased plasma ketone bodies and their ratio (acetoacetate/β-hydroxybutyrate) in 10-week-old male mice. Moreover, sleep deprivation increased mRNA expression of ketogenic genes such as PPARα and 3-hydroxy-3-methylglutarate-CoA synthase 2 in the brain and decreased ketolytic enzymes such as succinyl-CoA: 3-oxoacid CoA transferase. In addition, central injection of acetoacetate, but not β-hydroxybutyrate, markedly increased slow-wave activity during NREM sleep and suppressed glutamate release. Central metabolism of ketone bodies, especially acetoacetate, appears to play a role in the regulation of sleep homeostasis.
Collapse
Affiliation(s)
- Sachiko Chikahisa
- Department of Integrative Physiology, Institute of Health Biosciences, The University of Tokushima Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Noriyuki Shimizu
- Department of Integrative Physiology, Institute of Health Biosciences, The University of Tokushima Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Tetsuya Shiuchi
- Department of Integrative Physiology, Institute of Health Biosciences, The University of Tokushima Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hiroyoshi Séi
- Department of Integrative Physiology, Institute of Health Biosciences, The University of Tokushima Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan.
| |
Collapse
|
44
|
Waking and sleeping in the rat made obese through a high-fat hypercaloric diet. Behav Brain Res 2013; 258:145-52. [PMID: 24149066 DOI: 10.1016/j.bbr.2013.10.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/09/2013] [Accepted: 10/10/2013] [Indexed: 11/20/2022]
Abstract
Sleep restriction leads to metabolism dysregulation and to weight gain, which is apparently the consequence of an excessive caloric intake. On the other hand, obesity is associated with excessive daytime sleepiness in humans and promotes sleep in different rodent models of obesity. Since no consistent data on the wake-sleep (WS) pattern in diet-induced obesity rats are available, in the present study the effects on the WS cycle of the prolonged delivery of a high-fat hypercaloric (HC) diet leading to obesity were studied in Sprague-Dawley rats. The main findings are that animals kept under a HC diet for either four or eight weeks showed an overall decrease of time spent in wakefulness (Wake) and a clear Wake fragmentation when compared to animals kept under a normocaloric diet. The development of obesity was also accompanied with the occurrence of a larger daily amount of REM sleep (REMS). However, the capacity of HC animals to respond to a "Continuous darkness" exposure condition (obtained by extending the Dark period of the Light-Dark cycle to the following Light period) with an increase of Sequential REMS was dampened. The results of the present study indicate that if, on one hand, sleep curtailment promotes an excess of energy accumulation; on the other hand an over-exceeding energy accumulation depresses Wake. Thus, processes underlying energy homeostasis possibly interact with those underlying WS behavior, in order to optimize energy storage.
Collapse
|
45
|
Pan W, Kastin AJ. Leptin: a biomarker for sleep disorders? Sleep Med Rev 2013; 18:283-90. [PMID: 24080454 DOI: 10.1016/j.smrv.2013.07.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 06/05/2013] [Accepted: 07/18/2013] [Indexed: 12/11/2022]
Abstract
Leptin, a pleiotropic protein hormone produced mainly by fat cells, regulates metabolic activity and many other physiological functions. The intrinsic circadian rhythm of blood leptin is modulated by gender, development, feeding, fasting, sleep, obesity, and endocrine disorders. Hyperleptinemia is implicated in leptin resistance. To determine the specificity and sensitivity of leptin concentrations in sleep disorders, we summarize here the alterations of leptin in four conditions in animal and human studies: short duration of sleep, sleep fragmentation, obstructive sleep apnea (OSA), and after use of continuous positive airway pressure (CPAP) to treat OSA. The presence and causes of contradictory findings are discussed. Though sustained insufficient sleep lowers fasting blood leptin and therefore probably contributes to increased appetite, obesity and OSA independently result in hyperleptinemia. Successful treatment of OSA by CPAP is predicted to decrease hyperleptinemia, making leptin an ancillary biomarker for treatment efficacy. Current controversies also call for translational studies to determine how sleep disorders regulate leptin homeostasis and how the information can be used to improve sleep treatment.
Collapse
Affiliation(s)
- Weihong Pan
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| | - Abba J Kastin
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| |
Collapse
|
46
|
Hypersomnolence and reduced activity in pan-leptin receptor knockout mice. J Mol Neurosci 2013; 51:1038-45. [PMID: 23955775 DOI: 10.1007/s12031-013-0093-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 08/06/2013] [Indexed: 12/26/2022]
Abstract
Excessive obesity correlates with hypersomnolence and impaired cognitive function, presumably induced by metabolic factors and cytokines. Production of the adipokine leptin correlates with the amount of adiposity, and leptin has been shown to promote sleep. To determine whether leptin plays a major role in the hypersomnolence of obesity, we measured sleep architecture in pan-leptin receptor knockout (POKO) mice that do not respond to leptin because of the production of a mutant, non-signaling receptor. The obese POKO mice had more non-rapid eye movement (NREM) sleep and less waking time than their littermate controls. This was mainly seen during the light span, although increased bouts of rapid eye movement sleep were also seen in the dark span. The increase of NREM sleep correlated with the extent of obesity. The POKO mice also had decreased locomotor activity and more immobility in the open field test, but there was no increase of forced immobility nor reduction of sucrose intake as would be seen in depression. The increased NREM sleep and reduced locomotor activity in the POKO mice suggest that it was obesity, rather than leptin signaling, that played a predominant role in altering sleep architecture and activity.
Collapse
|
47
|
Borniger JC, Weil ZM, Zhang N, Nelson RJ. Dim light at night does not disrupt timing or quality of sleep in mice. Chronobiol Int 2013; 30:1016-23. [PMID: 23837748 DOI: 10.3109/07420528.2013.803196] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Artificial nighttime illumination has recently become commonplace throughout the world; however, in common with other animals, humans have not evolved in the ecological context of chronic light at night. With prevailing evidence linking the circadian, endocrine, immune, and metabolic systems, understanding these relationships is important to understanding the etiology and progression of several diseases. To eliminate the covariate of sleep disruption in light at night studies, researchers often use nocturnal animals. However, the assumption that light at night does not affect sleep in nocturnal animals remains unspecified. To test the effects of light at night on sleep, we maintained Swiss-Webster mice in standard light/dark (LD) or dim light at night (DLAN) conditions for 8-10 wks and then measured electroencephalogram (EEG) and electromyogram (EMG) biopotentials via wireless telemetry over the course of two consecutive days to determine differences in sleep timing and homeostasis. Results show no statistical differences in total percent time, number of episodes, maximum or average episode durations in wake, slow-wave sleep (SWS), or rapid eye movement (REM) sleep. No differences were evident in SWS delta power, an index of sleep drive, between groups. Mice kept in DLAN conditions showed a relative increase in REM sleep during the first few hours after the dark/light transition. Both groups displayed normal 24-h circadian rhythms as measured by voluntary running wheel activity. Groups did not differ in body mass, but a marked negative correlation of body mass with percent time spent awake and a positive correlation of body mass with time spent in SWS was evident. Elevated body mass was also associated with shorter maximum wake episode durations, indicating heavier animals had more trouble remaining in the wake vigilance state for extended periods of time. Body mass did not correlate with activity levels, nor did activity levels correlate with time spent in different sleep states. These data indicate that heavier animals tend to sleep more, potentially contributing to further weight gain. We conclude that chronic DLAN exposure does not significantly affect sleep timing or homeostasis in mice, supporting the use of dim light with nocturnal rodents in chronobiology research to eliminate the possible covariate of sleep disruption.
Collapse
Affiliation(s)
- Jeremy C Borniger
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | | | | | | |
Collapse
|
48
|
Tanno S, Terao A, Okamatsu-Ogura Y, Kimura K. Hypothalamic prepro-orexin mRNA level is inversely correlated to the non-rapid eye movement sleep level in high-fat diet-induced obese mice. Obes Res Clin Pract 2013; 7:e251-7. [DOI: 10.1016/j.orcp.2013.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 01/09/2013] [Accepted: 01/21/2013] [Indexed: 10/27/2022]
|
49
|
Trakada G, Steiropoulos P, Zarogoulidis P, Nena E, Papanas N, Maltezos E, Bouros D. A fatty meal aggravates apnea and increases sleep in patients with obstructive sleep apnea. Sleep Breath 2013; 18:53-8. [DOI: 10.1007/s11325-013-0847-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 02/14/2013] [Accepted: 04/01/2013] [Indexed: 01/23/2023]
|
50
|
Hoch T, Kreitz S, Gaffling S, Pischetsrieder M, Hess A. Manganese-enhanced magnetic resonance imaging for mapping of whole brain activity patterns associated with the intake of snack food in ad libitum fed rats. PLoS One 2013; 8:e55354. [PMID: 23408973 PMCID: PMC3567069 DOI: 10.1371/journal.pone.0055354] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 12/30/2012] [Indexed: 11/26/2022] Open
Abstract
Non-homeostatic hyperphagia, which is a major contributor to obesity-related hyperalimentation, is associated with the diet’s molecular composition influencing, for example, the energy content. Thus, specific food items such as snack food may induce food intake independent from the state of satiety. To elucidate mechanisms how snack food may induce non-homeostatic food intake, it was tested if manganese-enhanced magnetic resonance imaging (MEMRI) was suitable for mapping the whole brain activity related to standard and snack food intake under normal behavioral situation. Application of the MnCl2 solution by osmotic pumps ensured that food intake was not significantly affected by the treatment. After z-score normalization and a non-affine three-dimensional registration to a rat brain atlas, significantly different grey values of 80 predefined brain structures were recorded in ad libitum fed rats after the intake of potato chips compared to standard chow at the group level. Ten of these areas had previously been connected to food intake, in particular to hyperphagia (e.g. dorsomedial hypothalamus or the anterior paraventricular thalamic nucleus) or to the satiety system (e.g. arcuate hypothalamic nucleus or solitary tract); 27 areas were related to reward/addiction including the core and shell of the nucleus accumbens, the ventral pallidum and the ventral striatum (caudate and putamen). Eleven areas associated to sleep displayed significantly reduced Mn2+-accumulation and six areas related to locomotor activity showed significantly increased Mn2+-accumulation after the intake of potato chips. The latter changes were associated with an observed significantly higher locomotor activity. Osmotic pump-assisted MEMRI proved to be a promising technique for functional mapping of whole brain activity patterns associated to nutritional intake under normal behavior.
Collapse
Affiliation(s)
- Tobias Hoch
- Department of Chemistry and Pharmacy, Food Chemistry Division, Emil Fischer Center, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | |
Collapse
|