1
|
Mortimer T, Smith JG, Muñoz-Cánoves P, Benitah SA. Circadian clock communication during homeostasis and ageing. Nat Rev Mol Cell Biol 2025; 26:314-331. [PMID: 39753699 DOI: 10.1038/s41580-024-00802-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 03/28/2025]
Abstract
Maintaining homeostasis is essential for continued health, and the progressive decay of homeostatic processes is a hallmark of ageing. Daily environmental rhythms threaten homeostasis, and circadian clocks have evolved to execute physiological processes in a manner that anticipates, and thus mitigates, their effects on the organism. Clocks are active in almost all cell types; their rhythmicity and functional output are determined by a combination of tissue-intrinsic and systemic inputs. Numerous inputs for a specific tissue are produced by the activity of circadian clocks of other tissues or cell types, generating a form of crosstalk known as clock communication. In mammals, the central clock in the hypothalamus integrates signals from external light-dark cycles to align peripheral clocks elsewhere in the body. This regulation is complemented by a tissue-specific milieu of external, systemic and niche inputs that modulate and cooperate with the cellular circadian clock machinery of a tissue to tailor its functional output. These mechanisms of clock communication decay during ageing, and growing evidence suggests that this decline might drive ageing-related morbidities. Dietary, behavioural and pharmacological interventions may offer the possibility to overcome these changes and in turn improve healthspan.
Collapse
Affiliation(s)
- Thomas Mortimer
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| | - Jacob G Smith
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain.
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain.
| | - Pura Muñoz-Cánoves
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
- Altos Labs Inc., San Diego Institute of Science, San Diego, CA, USA.
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| |
Collapse
|
2
|
Pearl AJ, Maddern XJ, Pinares-Garcia P, Ursich LT, Anversa RG, Shesham A, Brown RM, Reed FM, Giardino WJ, Lawrence AJ, Walker LC. Midbrain ghrelin receptor signalling regulates binge drinking in a sex specific manner. Nat Commun 2025; 16:2568. [PMID: 40089486 PMCID: PMC11910522 DOI: 10.1038/s41467-025-57880-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Risky drinking rates are rising, particularly in women, yet sex as a biological variable has only recently gained traction. The centrally projecting Edinger-Westphal (EWcp) nucleus has emerged as a key regulator of alcohol consumption. Here we found that EWcppeptidergic cells reduce binge drinking specifically in female mice. We show this effect is mediated by the ghrelin receptor (GHSR), with EWcppeptidergic inhibition blocking ghrelin-induced drinking and Ghsr knockdown in EWcppeptidergic, but not EWcpglutamatergic or ventral tegmental area cells, reducing binge drinking in females, independent of circulating sex hormones. Female mice showed higher EWcp Ghsr expression, and EWcppeptidergic neurons were more sensitive to ghrelin. Moreover, intra-EWcp delivery of GHSR inverse agonist and antagonist reduced binge drinking, suggesting direct actions of ghrelin. These findings highlight the EWcp as a critical mediator of excessive alcohol consumption via GHSR in female mice, offering insights into the ghrelin system's role in alcohol consumption.
Collapse
Affiliation(s)
- Amy J Pearl
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
| | - Xavier J Maddern
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Paulo Pinares-Garcia
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
| | - Lauren T Ursich
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Roberta G Anversa
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Arnav Shesham
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Robyn M Brown
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Felicia M Reed
- Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| | - William J Giardino
- Dept. of Psychiatry and Behavioural Sciences, Stanford University School of Medicine, Stanford, CA, 94305-5453, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, 94305-5453, USA
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia.
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3052, Australia.
| |
Collapse
|
3
|
Deota S, Pendergast JS, Kolthur-Seetharam U, Esser KA, Gachon F, Asher G, Dibner C, Benitah SA, Escobar C, Muoio DM, Zhang EE, Hotamışlıgil GS, Bass J, Takahashi JS, Rabinowitz JD, Lamia KA, de Cabo R, Kajimura S, Longo VD, Xu Y, Lazar MA, Verdin E, Zierath JR, Auwerx J, Drucker DJ, Panda S. The time is now: accounting for time-of-day effects to improve reproducibility and translation of metabolism research. Nat Metab 2025; 7:454-468. [PMID: 40097742 DOI: 10.1038/s42255-025-01237-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
The constant expansion of the field of metabolic research has led to more nuanced and sophisticated understanding of the complex mechanisms that underlie metabolic functions and diseases. Collaborations with scientists of various fields such as neuroscience, immunology and drug discovery have further enhanced the ability to probe the role of metabolism in physiological processes. However, many behaviours, endocrine and biochemical processes, and the expression of genes, proteins and metabolites have daily ~24-h biological rhythms and thus peak only at specific times of the day. This daily variation can lead to incorrect interpretations, lack of reproducibility across laboratories and challenges in translating preclinical studies to humans. In this Review, we discuss the biological, environmental and experimental factors affecting circadian rhythms in rodents, which can in turn alter their metabolic pathways and the outcomes of experiments. We recommend that these variables be duly considered and suggest best practices for designing, analysing and reporting metabolic experiments in a circadian context.
Collapse
Affiliation(s)
- Shaunak Deota
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Ullas Kolthur-Seetharam
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
- Tata Institute of Fundamental Research, Hyderabad, India
| | - Karyn A Esser
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - Frédéric Gachon
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Gad Asher
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Charna Dibner
- Department of Surgery and Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute for Science and Technology, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Carolina Escobar
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Deborah M Muoio
- Departments of Medicine and Pharmacology & Cancer Biology, Duke Molecular Physiology Institute, Durham, NC, USA
| | | | - Gökhan S Hotamışlıgil
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Katja A Lamia
- Department of Molecular and Cellular Biology and Department of Molecular Medicine, the Scripps Research Institute, La Jolla, CA, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Shingo Kajimura
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, USA
| | - Valter D Longo
- Longevity Institute, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- AIRC Institute of Molecular Oncology, Italian Foundation for Cancer Research Institute of Molecular Oncology, Milan, Italy
| | - Ying Xu
- CAM-SU Genomic Resource Center, Soochow University, Suzhou, China
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity and Metabolism and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Daniel J Drucker
- The Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and the Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
4
|
Volcko KL, Taghipourbibalan H, McCutcheon JE. Intermittent protein restriction elevates food intake and plasma ghrelin in male mice. Appetite 2024; 203:107671. [PMID: 39265824 DOI: 10.1016/j.appet.2024.107671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/06/2024] [Accepted: 09/07/2024] [Indexed: 09/14/2024]
Abstract
Low-protein diets affect body weight, body composition, food intake, and food preferences in mice. Furthermore, single periods of protein restriction can have lasting effects on these parameters. We sought to examine the effect of multiple, short, bouts of protein restriction, relative to long-term maintenance on either a control (NR) or protein-restricted (PR) diet. We found that male mice experiencing intermittent protein restriction (IPR) were indistinguishable from NR mice in terms of body weight and composition, but had food intake and plasma ghrelin as high as mice on PR diet, even when they were returned to control diet. This was not found in female mice. The results of this experiment highlight the importance of diet history on food intake and ghrelin levels in male mice, and the difference in how PR diet might affect male and female mice.
Collapse
Affiliation(s)
- K L Volcko
- Department of Psychology, UiT the Arctic University of Norway, Tromsø, Norway.
| | - H Taghipourbibalan
- Department of Psychology, UiT the Arctic University of Norway, Tromsø, Norway
| | - J E McCutcheon
- Department of Psychology, UiT the Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
5
|
Page AJ. Plasticity of gastrointestinal vagal afferents in terms of feeding-related physiology and pathophysiology. J Physiol 2024; 602:4763-4776. [PMID: 37737742 DOI: 10.1113/jp284075] [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: 06/09/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023] Open
Abstract
Gastrointestinal vagal afferents play an important role in communicating food related information from the gut to the brain. This information initiates vago-vagal reflexes essential for gut functions, including gut motility and secretions. These afferents also play a role in energy homeostasis, signalling the arrival, amount and nutrient composition of a meal to the central nervous system where it is processed ultimately leading to termination of a meal. Vagal afferent responses to food related stimuli demonstrate a high degree of plasticity, responding to short term changes in nutritional demand, such as the fluctuations that occur across a 24-hr or in response to a fast, as well as long term changes in energy demand, such as occurs during pregnancy. This plasticity is disrupted in disease states, such as obesity or chronic stress where there is hypo- and hypersensitivity of these afferents, respectively. Improved understanding of the plasticity of these afferents will enable identification of new treatment options for diseases associated with vagal afferent function.
Collapse
Affiliation(s)
- Amanda J Page
- Vagal Afferent Research Group, School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institute, SAHMRI, Adelaide, South Australia, Australia
| |
Collapse
|
6
|
Osorio M J, Mitchell SE, Hambly C, Allison DB, Speakman JR. Not feeling the heat? Effects of dietary protein on satiation and satiety in mice are not due to its impact on body temperature. Appetite 2024; 200:107421. [PMID: 38759755 DOI: 10.1016/j.appet.2024.107421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/09/2024] [Accepted: 05/12/2024] [Indexed: 05/19/2024]
Abstract
Dietary protein modulates food intake (FI) via unclear mechanism(s). One possibility is that higher protein leads to greater post-ingestive heat production (Specific dynamic action: SDA) leading to earlier meal termination (increased satiation), and inhibition of further intake (increased satiety). The influence of dietary protein on feeding behaviour in C57BL/6J mice was tested using an automated FI monitoring system (BioDAQ), simultaneous to body temperature (Tb). Total FI, inter meal intervals (IMI, satiety) and meal size (MS, satiation) were related to changes in Tb after consuming low (5%, LP), moderate (15%, MP) and high (30%, HP) protein diets. Diets were tested over three conditions: 1) room temperature (RT, 21 ± 1 °C), 2) room temperature and running wheels (RTRW) and 3) low temperature (10 °C) and running wheels (LTRW). The differences between diets and conditions were also compared using mixed models. Mice housed at RT fed HP diet, reduced total FI compared with LP and MP due to earlier meal termination (satiation effect). FI was lowered in RTRW conditions with no differences between diets. FI significantly increased under LTRW conditions for all diets, with protein content leading to earlier meal termination (satiation) but not the intervals between feeding bouts (satiety). Tb fell immediately after feeding in all conditions. Despite a reduction in total FI in mice fed HP, mediated via increased satiation, this effect was not linked to increased Tb during meals. We conclude effects of dietary protein on intake are not mediated via SDA and Tb.
Collapse
Affiliation(s)
- Jazmin Osorio M
- School of Biological Sciences, University of Aberdeen, AB24 2TZ, Scotland, UK; Institute of Neurobiology, University of Lübeck, 23562, Lübeck, Germany
| | - Sharon E Mitchell
- School of Biological Sciences, University of Aberdeen, AB24 2TZ, Scotland, UK
| | - Catherine Hambly
- School of Biological Sciences, University of Aberdeen, AB24 2TZ, Scotland, UK
| | - David B Allison
- Indiana University School of Public Health - Bloomington, Indiana, 47405, USA
| | - John R Speakman
- School of Biological Sciences, University of Aberdeen, AB24 2TZ, Scotland, UK; Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China; China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
7
|
Peng F, Lu J, Su K, Liu X, Luo H, He B, Wang C, Zhang X, An F, Lv D, Luo Y, Su Q, Jiang T, Deng Z, He B, Xu L, Guo T, Xiang J, Gu C, Wang L, Xu G, Xu Y, Li M, Kelley KW, Cui B, Liu Q. Oncogenic fatty acid oxidation senses circadian disruption in sleep-deficiency-enhanced tumorigenesis. Cell Metab 2024; 36:1598-1618.e11. [PMID: 38772364 DOI: 10.1016/j.cmet.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 03/15/2024] [Accepted: 04/25/2024] [Indexed: 05/23/2024]
Abstract
Circadian disruption predicts poor cancer prognosis, yet how circadian disruption is sensed in sleep-deficiency (SD)-enhanced tumorigenesis remains obscure. Here, we show fatty acid oxidation (FAO) as a circadian sensor relaying from clock disruption to oncogenic metabolic signal in SD-enhanced lung tumorigenesis. Both unbiased transcriptomic and metabolomic analyses reveal that FAO senses SD-induced circadian disruption, as illustrated by continuously increased palmitoyl-coenzyme A (PA-CoA) catalyzed by long-chain fatty acyl-CoA synthetase 1 (ACSL1). Mechanistically, SD-dysregulated CLOCK hypertransactivates ACSL1 to produce PA-CoA, which facilitates CLOCK-Cys194 S-palmitoylation in a ZDHHC5-dependent manner. This positive transcription-palmitoylation feedback loop prevents ubiquitin-proteasomal degradation of CLOCK, causing FAO-sensed circadian disruption to maintain SD-enhanced cancer stemness. Intriguingly, timed β-endorphin resets rhythmic Clock and Acsl1 expression to alleviate SD-enhanced tumorigenesis. Sleep quality and serum β-endorphin are negatively associated with both cancer development and CLOCK/ACSL1 expression in patients with cancer, suggesting dawn-supplemented β-endorphin as a potential chronotherapeutic strategy for SD-related cancer.
Collapse
Affiliation(s)
- Fei Peng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Jinxin Lu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Keyu Su
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xinyu Liu
- Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, Liaoning, China
| | - Huandong Luo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Bin He
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Cenxin Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Xiaoyu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Fan An
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Dekang Lv
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Yuanyuan Luo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, Liaoning, China
| | - Qitong Su
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Tonghui Jiang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Ziqian Deng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Bin He
- State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Lingzhi Xu
- Department of Oncology, The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Tao Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jin Xiang
- State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Chundong Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ling Wang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Guowang Xu
- Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, Liaoning, China
| | - Ying Xu
- Cambridge-Soochow University Genomic Resource Center, Soochow University, Suzhou, Jiangsu, China
| | - Mindian Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Keith W Kelley
- Department of Pathology, College of Medicine and Department of Animal Sciences, College of ACES, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Bai Cui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Quentin Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
8
|
Kulkarni SS, Singh O, Zigman JM. The intersection between ghrelin, metabolism and circadian rhythms. Nat Rev Endocrinol 2024; 20:228-238. [PMID: 38123819 PMCID: PMC11760189 DOI: 10.1038/s41574-023-00927-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2023] [Indexed: 12/23/2023]
Abstract
Despite the growing popular interest in sleep and diet, many gaps exist in our scientific understanding of the interaction between circadian rhythms and metabolism. In this Review, we explore a promising, bidirectional role for ghrelin in mediating this interaction. Ghrelin both influences and is influenced by central and peripheral circadian systems. Specifically, we focus on how ghrelin impacts outputs of circadian rhythm, including neuronal activity, circulating growth hormone levels, locomotor activity and eating behaviour. We also consider the effects of circadian rhythms on ghrelin expression and the consequences of disrupted circadian patterns, such as shift work and jet lag, on ghrelin secretion. Our Review is aimed at both the casual reader interested in gaining more insight into the scientific context surrounding the trending topics of sleep and metabolism, as well as experienced scientists in the fields of ghrelin and circadian biology seeking inspiration and a comprehensive overview of how these fields are related.
Collapse
Affiliation(s)
- Soumya S Kulkarni
- Medical Scientist Training Program, UT Southwestern Medical Center, Dallas, TX, USA
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Omprakash Singh
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.
- Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
9
|
Eyikudamaci G, Ege H, Ensen N, Yelmen N. Role of insulin resistance and leptin in the effect of intermittent feeding with a high-protein ketogenic diet on body composition in rats. Nutrition 2024; 117:112213. [PMID: 37832180 DOI: 10.1016/j.nut.2023.112213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 10/15/2023]
Abstract
OBJECTIVES The prevalence of obesity and overweight in children has been increasing rapidly worldwide and threatens society with various chronic diseases that these children are born with. High-protein ketogenic diets and intermittent nutrition are thought to be protective against obesity and metabolic syndrome MetS. However, the exact effects and results, insulin resistance, and the role of leptin in the functioning mechanism of these diets have not been fully elucidated. The aim of this study was to investigate the roles of insulin resistance and leptin hormone on the effects of body composition with a high-protein ketogenic diet and intermittent nutrition combination. METHODS Thirty-two young non-obese rats were randomly divided into four equal groups. Both the standard diet and the high-protein ketogenic diet were given ad libitum and intermittently to the rats for 6 wk. The body weight and fat mass of the rats were measured at the end of the experiment. The fasting glucose, leptin, insulin, high- and low-density lipoprotein, and triacylglycerols were measured with the blood samples. RESULTS The lowest body weight was observed in the intermittent and high-protein ketogenic diet group, followed by the free high-protein ketogenic diet and standard intermittent diet group, respectively. Also, the lowest body fat mass was observed in the intermittent and high-protein ketogenic diet group, followed by the standard intermittent diet group. Although there was no change in leptin, insulin, high- and low-density lipoprotein, and triacylglycerol levels in any group, the lowest blood glucose rate was observed in the intermittent and high-protein ketogenic diet group. CONCLUSION The results of the present study revealed that an intermittent high-protein ketogenic diet is more effective than others in weight loss without disrupting biochemical health parameters, and the applied diets do not prevent growth and development.
Collapse
Affiliation(s)
- Gul Eyikudamaci
- Department of Physiology, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Hasan Ege
- Department of Physiology, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Nihal Ensen
- Department of Chest Diseases, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Nermin Yelmen
- Department of Physiology, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey.
| |
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
|
Harris C, Czaja K. Can Circadian Eating Pattern Adjustments Reduce Risk or Prevent Development of T2D? Nutrients 2023; 15:nu15071762. [PMID: 37049602 PMCID: PMC10096926 DOI: 10.3390/nu15071762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/30/2023] [Accepted: 04/01/2023] [Indexed: 04/07/2023] Open
Abstract
Type 2 diabetes (T2D) is a chronic condition that occurs in insulin-resistant people with reduced glucose uptake. It is contributed to and exacerbated by a poor diet that results in accumulation of adipose tissue, high blood sugar, and other metabolic issues. Because humans have undergone food scarcity throughout history, our species has adapted a fat reserve genotype. This adaptation is no longer beneficial, as eating at a higher frequency than that of our ancestors has had a significant effect on T2D development. Eating at high frequencies disrupts the circadian clock, the circadian rhythm, and the composition of the gut microbiome, as well as hormone secretion and sensitivity. The current literature suggests an improved diet requires meal consistency, avoiding late-night eating, low meal frequency, and fasting to increase metabolic health. In addition, fasting as a treatment for T2D must be used correctly for beneficial results. Early time-restricted eating (TRE) provides many benefits such as improving insulin resistance, cognitive function, and glycemic control. Alternate-day fasting (ADF), 5:2 fasting, and long-term fasting all have benefits; however, they may be less advantageous than early TRE. Therefore, eating pattern adjustments can be used to reduce T2D if used correctly.
Collapse
Affiliation(s)
- Carlee Harris
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Krzysztof Czaja
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
12
|
Villanueva-Carmona T, Cedó L, Madeira A, Ceperuelo-Mallafré V, Rodríguez-Peña MM, Núñez-Roa C, Maymó-Masip E, Repollés-de-Dalmau M, Badia J, Keiran N, Mirasierra M, Pimenta-Lopes C, Sabadell-Basallote J, Bosch R, Caubet L, Escolà-Gil JC, Fernández-Real JM, Vilarrasa N, Ventura F, Vallejo M, Vendrell J, Fernández-Veledo S. SUCNR1 signaling in adipocytes controls energy metabolism by modulating circadian clock and leptin expression. Cell Metab 2023; 35:601-619.e10. [PMID: 36977414 DOI: 10.1016/j.cmet.2023.03.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/21/2022] [Accepted: 03/03/2023] [Indexed: 03/30/2023]
Abstract
Adipose tissue modulates energy homeostasis by secreting leptin, but little is known about the factors governing leptin production. We show that succinate, long perceived as a mediator of immune response and lipolysis, controls leptin expression via its receptor SUCNR1. Adipocyte-specific deletion of Sucnr1 influences metabolic health according to nutritional status. Adipocyte Sucnr1 deficiency impairs leptin response to feeding, whereas oral succinate mimics nutrient-related leptin dynamics via SUCNR1. SUCNR1 activation controls leptin expression via the circadian clock in an AMPK/JNK-C/EBPα-dependent manner. Although the anti-lipolytic role of SUCNR1 prevails in obesity, its function as a regulator of leptin signaling contributes to the metabolically favorable phenotype in adipocyte-specific Sucnr1 knockout mice under standard dietary conditions. Obesity-associated hyperleptinemia in humans is linked to SUCNR1 overexpression in adipocytes, which emerges as the major predictor of adipose tissue leptin expression. Our study establishes the succinate/SUCNR1 axis as a metabolite-sensing pathway mediating nutrient-related leptin dynamics to control whole-body homeostasis.
Collapse
Affiliation(s)
- Teresa Villanueva-Carmona
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Lídia Cedó
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Ana Madeira
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Victòria Ceperuelo-Mallafré
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain; Department of Medicine and Surgery, Universitat Rovira i Virgili (URV), Reus 43201, Spain
| | - M-Mar Rodríguez-Peña
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Catalina Núñez-Roa
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Elsa Maymó-Masip
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Maria Repollés-de-Dalmau
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain; Department of Medicine and Surgery, Universitat Rovira i Virgili (URV), Reus 43201, Spain
| | - Joan Badia
- Institut d'Oncologia de la Catalunya Sud, Hospital Universitari Sant Joan de Reus, IISPV, Reus 43204, Spain
| | - Noelia Keiran
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Mercedes Mirasierra
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain; Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid (CSIC/UAM), Madrid 28029, Spain
| | - Carolina Pimenta-Lopes
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Joan Sabadell-Basallote
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Ramón Bosch
- Department of Pathology, Oncological Pathology and Bioinformatics Research Group, Hospital de Tortosa Verge de la Cinta, IISPV, Tortosa 43500, Spain
| | - Laura Caubet
- General and Digestive Surgery Service, Hospital Sant Pau i Santa Tecla, Institut d'Investigació Sanitària Pere Virgili, Tarragona 43003, Spain
| | - Joan Carles Escolà-Gil
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain; Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona 08041, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona 08193, Spain
| | - José-Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), Salt 17190, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CB06/03/010), Instituto de Salud Carlos III, Madrid 28029, Spain; Department of Medical Sciences, School of Medicine, University of Girona, Girona 17004, Spain
| | - Nuria Vilarrasa
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain; Department of Endocrinology and Nutrition, Hospital Universitari Bellvitge - IDIBELL, Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Francesc Ventura
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Mario Vallejo
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain; Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid (CSIC/UAM), Madrid 28029, Spain
| | - Joan Vendrell
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain; Department of Medicine and Surgery, Universitat Rovira i Virgili (URV), Reus 43201, Spain
| | - Sonia Fernández-Veledo
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain.
| |
Collapse
|
13
|
Fritz EM, Pierre A, De Bundel D, Singewald N. Ghrelin receptor agonist MK0677 and overnight fasting do not rescue deficient fear extinction in 129S1/SvImJ mice. Front Psychiatry 2023; 14:1094948. [PMID: 36846243 PMCID: PMC9947350 DOI: 10.3389/fpsyt.2023.1094948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/03/2023] [Indexed: 02/11/2023] Open
Abstract
The hunger hormone ghrelin has been implicated in the modulation of anxiety- and fear-related behaviors in rodents and humans, while its dysregulation may be associated with psychiatric illness. Along these lines, the ghrelin system has been suggested as a potential target to facilitate fear extinction, which is the main mechanism underlying cognitive behavioral therapy. So far, this hypothesis has not been tested in individuals that have difficulties to extinguish fear. Thus, we investigated pharmacological (ghrelin receptor agonist MK0677) and non-pharmacological (overnight fasting) strategies to target the ghrelin system in the 129S1/SvImJ (S1) mouse strain, which models the endophenotype of impaired fear extinction that has been associated with treatment resistance in anxiety and PTSD patients. MK0677 induced food intake and overnight fasting increased plasma ghrelin levels in S1 mice, suggesting that the ghrelin system is responsive in the S1 strain. However, neither systemic administration of MK0677 nor overnight fasting had an effect on fear extinction in S1 mice. Similarly, our groups previously reported that both interventions did not attenuate fear in extinction-competent C57BL/6J mice. In summary, our findings are in contrast to several studies reporting beneficial effects of GHSR agonism and overnight fasting on fear- and anxiety-related behaviors in rodents. Rather, our data agree with accumulating evidence of divergent behavioral effects of ghrelin system activation and underscore the hypothesis that potential benefits of targeting the ghrelin system in fear extinction may be dependent on factors (e.g., previous stress exposure) that are not yet fully understood.
Collapse
Affiliation(s)
- Eva Maria Fritz
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Innsbruck, Austria
| | - Anouk Pierre
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Dimitri De Bundel
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
14
|
Marasco V, Kaiya H, Pola G, Fusani L. Ghrelin, not corticosterone, is associated with transitioning of phenotypic states in a migratory Galliform. Front Endocrinol (Lausanne) 2023; 13:1058298. [PMID: 36699038 PMCID: PMC9869107 DOI: 10.3389/fendo.2022.1058298] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/28/2022] [Indexed: 01/12/2023] Open
Abstract
In both captive and free-living birds, the emergence of the migratory phenotype is signalled by rapid and marked increases in food intake and fuelling, as well as changes in amount of nocturnality or migratory restlessness. The metabolic hormone corticosterone and, as more recently suggested, the gut-derived hormone ghrelin have been suggested to play a role in mediating such phenomenal phenotypic flexibility given that they both regulate fuel metabolism and locomotion across vertebrate taxa. Here, using the Common quail (Coturnix coturnix) as our study species, we induced autumn migration followed by a non-migratory wintering phase through controlled changes in daylight. We thus compared plasma corticosterone and ghrelin concentrations between the two sampling phases and assessed whether these hormones might reflect the migratory state. While we found no differences in plasma corticosterone between the two sampling phases and no link of this hormone with changes in body mass, levels of food intake or migratory restlessness, the migratory birds had substantially higher levels of plasma ghrelin relative to the non-migratory birds. Furthermore, while ghrelin did not correlate with the gain in body mass over the entire pre-migratory fuelling phase (over an average of nine weeks preceding blood sampling), plasma ghrelin did positively correlate with the gain in body mass observed during the final fattening stages (over an average of three weeks preceding blood sampling). Again, variation in plasma ghrelin also reflected the amount of body mass depleted over both the long- and short-time frame as birds returned to their non-migratory baseline - lower levels of plasma ghrelin consistently correlated with larger losses in body mass. Thus, while our data do not highlight a role of the hormone corticosterone in sustaining pre-migratory fattening as shown in other bird species, they do add evidence for a potential role of ghrelin in mediating migratory behaviour and further suggest that this hormone might be important in regulating the transitioning of migratory states, possibly by promoting fuel mobilisation and usage.
Collapse
Affiliation(s)
- Valeria Marasco
- Konrad Lorenz Institute of Ethology, University of Veterinary Medicine, Vienna, Vienna, Austria
| | - Hiroyuki Kaiya
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
- Research Division of Drug Discovery, Grandsoul Research Institute for Immunology, Inc., Nara, Japan
| | - Gianni Pola
- Istituto Sperimentale Zootecnico per la Sicilia, Palermo, Italy
| | - Leonida Fusani
- Konrad Lorenz Institute of Ethology, University of Veterinary Medicine, Vienna, Vienna, Austria
- Department of Behavioural and Cognitive Biology, University Biology Building, University of Vienna, Vienna, Austria
| |
Collapse
|
15
|
Chrononutrition-When We Eat Is of the Essence in Tackling Obesity. Nutrients 2022; 14:nu14235080. [PMID: 36501110 PMCID: PMC9739590 DOI: 10.3390/nu14235080] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Obesity is a chronic and relapsing public health problem with an extensive list of associated comorbidities. The worldwide prevalence of obesity has nearly tripled over the last five decades and continues to pose a serious threat to wider society and the wellbeing of future generations. The pathogenesis of obesity is complex but diet plays a key role in the onset and progression of the disease. The human diet has changed drastically across the globe, with an estimate that approximately 72% of the calories consumed today come from foods that were not part of our ancestral diets and are not compatible with our metabolism. Additionally, multiple nutrient-independent factors, e.g., cost, accessibility, behaviours, culture, education, work commitments, knowledge and societal set-up, influence our food choices and eating patterns. Much research has been focused on 'what to eat' or 'how much to eat' to reduce the obesity burden, but increasingly evidence indicates that 'when to eat' is fundamental to human metabolism. Aligning feeding patterns to the 24-h circadian clock that regulates a wide range of physiological and behavioural processes has multiple health-promoting effects with anti-obesity being a major part. This article explores the current understanding of the interactions between the body clocks, bioactive dietary components and the less appreciated role of meal timings in energy homeostasis and obesity.
Collapse
|
16
|
Ramirez-Plascencia OD, Saderi N, Cárdenas Romero S, Flores Sandoval O, Báez-Ruiz A, Martínez Barajas H, Salgado-Delgado R. Temporal dysregulation of hypothalamic integrative and metabolic nuclei in rats fed during the rest phase. Chronobiol Int 2022; 39:374-385. [PMID: 34906015 DOI: 10.1080/07420528.2021.2002352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Temporal coordination of organisms according to the daytime allows a better performance of physiological processes. However, modern lifestyle habits, such as food intake during the rest phase, promote internal desynchronization and compromise homeostasis and health. The hypothalamic suprachiasmatic nucleus (SCN) synchronizes body physiology and behavior with the environmental light-dark cycle by transmitting time information to several integrative hypothalamic nuclei, such as the paraventricular nucleus (PVN), dorsomedial hypothalamic nucleus (DMH) and median preoptic area (MnPO). The SCN receives metabolic information mainly via Neuropeptide Y (NPY) inputs from the intergeniculate nucleus of the thalamus (IGL). Nowadays, there is no evidence of the response of the PVN, DMH and MnPO when the animals are subjected to internal desynchronization by restricting food access to the rest phase of the day. To explore this issue, we compared the circadian activity of the SCN, PVN, DMH and MnPO. In addition, we analyzed the daily activity of the satiety centers of the brainstem, the nucleus of the tractus solitarius (NTS) and area postrema (AP), which send metabolic information to the SCN, directly or via the thalamic intergeniculate leaflet (IGL). For that, male Wistar rats were assigned to three meal protocols: fed during the rest phase (Day Fed); fed during the active phase (Night Fed); free access to food (ad libitum). After 21 d, the daily activity patterns of these nuclei were analyzed by c-Fos immunohistochemistry, as well as NPY immunohistochemistry, in the SCN. The results show that eating during the rest period produces a phase advance in the activity of the SCN, changes the daily activity pattern in the MnPO, NTS and AP and flattens the c-Fos rhythm in the PVN and DMH. Altogether, these results validate previous observations of circadian dysregulation that occurs within the central nervous system when meals are consumed during the rest phase, a behavior that is involved in the metabolic alterations described in the literature.
Collapse
Affiliation(s)
- Oscar D Ramirez-Plascencia
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.,Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Nadia Saderi
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | | | - Omar Flores Sandoval
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Adrián Báez-Ruiz
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | | | | |
Collapse
|
17
|
Tacad DKM, Tovar AP, Richardson CE, Horn WF, Krishnan GP, Keim NL, Krishnan S. Satiety Associated with Calorie Restriction and Time-Restricted Feeding: Peripheral Hormones. Adv Nutr 2022; 13:792-820. [PMID: 35191467 PMCID: PMC9156388 DOI: 10.1093/advances/nmac014] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/08/2021] [Accepted: 02/11/2022] [Indexed: 12/14/2022] Open
Abstract
Calorie restriction (CR) is a common approach to inducing negative energy balance. Recently, time-restricted feeding (TRF), which involves consuming food within specific time windows during a 24-h day, has become popular owing to its relative ease of practice and potential to aid in achieving and maintaining a negative energy balance. TRF can be implemented intentionally with CR, or TRF might induce CR simply because of the time restriction. This review focuses on summarizing our current knowledge on how TRF and continuous CR affect gut peptides that influence satiety. Based on peer-reviewed studies, in response to CR there is an increase in the orexigenic hormone ghrelin and a reduction in fasting leptin and insulin. There is likely a reduction in glucagon-like peptide-1 (GLP-1), peptide YY (PYY), and cholecystokinin (CCK), albeit the evidence for this is weak. After TRF, unlike CR, fasting ghrelin decreased in some TRF studies, whereas it showed no change in several others. Further, a reduction in fasting leptin, insulin, and GLP-1 has been observed. In conclusion, when other determinants of food intake are held equal, the peripheral satiety systems appear to be somewhat similarly affected by CR and TRF with regard to leptin, insulin, and GLP-1. But unlike CR, TRF did not appear to robustly increase ghrelin, suggesting different influences on appetite with a potential decrease of hunger after TRF when compared with CR. However, there are several established and novel gut peptides that have not been measured within the context of CR and TRF, and studies that have evaluated effects of TRF are often short-term, with nonuniform study designs and highly varying temporal eating patterns. More evidence and studies addressing these aspects are needed to draw definitive conclusions.
Collapse
Affiliation(s)
- Debra K M Tacad
- Obesity and Metabolism Research Unit, USDA-Agricultural Research Service Western Human Nutrition Research Center, Davis, CA, USA,Department of Nutrition, University of California Davis, Davis, CA, USA
| | - Ashley P Tovar
- Department of Nutrition, University of California Davis, Davis, CA, USA
| | | | - William F Horn
- Obesity and Metabolism Research Unit, USDA-Agricultural Research Service Western Human Nutrition Research Center, Davis, CA, USA
| | - Giri P Krishnan
- Department of Medicine, School of Medicine, University of California San Diego, San Diego, CA, USA
| | | | | |
Collapse
|
18
|
Đukanović N, La Spada F, Emmenegger Y, Niederhäuser G, Preitner F, Franken P. Depriving Mice of Sleep also Deprives of Food. Clocks Sleep 2022; 4:37-51. [PMID: 35225952 PMCID: PMC8884003 DOI: 10.3390/clockssleep4010006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/31/2022] [Accepted: 02/05/2022] [Indexed: 02/06/2023] Open
Abstract
Both sleep-wake behavior and circadian rhythms are tightly coupled to energy metabolism and food intake. Altered feeding times in mice are known to entrain clock gene rhythms in the brain and liver, and sleep-deprived humans tend to eat more and gain weight. Previous observations in mice showing that sleep deprivation (SD) changes clock gene expression might thus relate to altered food intake, and not to the loss of sleep per se. Whether SD affects food intake in the mouse and how this might affect clock gene expression is, however, unknown. We therefore quantified (i) the cortical expression of the clock genes Per1, Per2, Dbp, and Cry1 in mice that had access to food or not during a 6 h SD, and (ii) food intake during baseline, SD, and recovery sleep. We found that food deprivation did not modify the SD-incurred clock gene changes in the cortex. Moreover, we discovered that although food intake during SD did not differ from the baseline, mice lost weight and increased food intake during subsequent recovery. We conclude that SD is associated with food deprivation and that the resulting energy deficit might contribute to the effects of SD that are commonly interpreted as a response to sleep loss.
Collapse
Affiliation(s)
- Nina Đukanović
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; (N.Đ.); (F.L.S.); (Y.E.)
| | - Francesco La Spada
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; (N.Đ.); (F.L.S.); (Y.E.)
| | - Yann Emmenegger
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; (N.Đ.); (F.L.S.); (Y.E.)
| | - Guy Niederhäuser
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; (N.Đ.); (F.L.S.); (Y.E.)
- Mouse Metabolic Evaluation Facility, Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; (G.N.); (F.P.)
| | - Frédéric Preitner
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; (N.Đ.); (F.L.S.); (Y.E.)
- Mouse Metabolic Evaluation Facility, Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; (G.N.); (F.P.)
| | - Paul Franken
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; (N.Đ.); (F.L.S.); (Y.E.)
- Correspondence:
| |
Collapse
|
19
|
Adlan LG, Csordás-Nagy M, Bodosi B, Kalmár G, Nyúl LG, Nagy A, Kekesi G, Büki A, Horvath G. Sleep-Wake Rhythm and Oscillatory Pattern Analysis in a Multiple Hit Schizophrenia Rat Model (Wisket). Front Behav Neurosci 2022; 15:799271. [PMID: 35153694 PMCID: PMC8831724 DOI: 10.3389/fnbeh.2021.799271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Electroencephalography studies in schizophrenia reported impairments in circadian rhythm and oscillatory activity, which may reflect the deficits in cognitive and sensory processing. The current study evaluated the circadian rhythm and the state-dependent oscillatory pattern in control Wistar and a multiple hit schizophrenia rat model (Wisket) using custom-made software for identification of the artifacts and the classification of sleep-wake stages and the active and quiet awake substages. The Wisket animals have a clear light-dark cycle similar to controls, and their sleep-wake rhythm showed only a tendency to spend more time in non-rapid eye movement (NREM) and less in rapid eye movement (REM) stages. In spite of the weak diurnal variation in oscillation in both groups, the Wisket rats had higher power in the low-frequency delta, alpha, and beta bands and lower power in the high-frequency theta and gamma bands in most stages. Furthermore, the significant differences between the two groups were pronounced in the active waking substage. These data suggest that the special changes in the oscillatory pattern of this schizophrenia rat model may have a significant role in the impaired cognitive functions observed in previous studies.
Collapse
Affiliation(s)
- Leatitia Gabriella Adlan
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Mátyás Csordás-Nagy
- Department of Technical Informatics, Faculty of Science and Informatics, Institute of Informatics, University of Szeged, Szeged, Hungary
| | - Balázs Bodosi
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - György Kalmár
- Department of Technical Informatics, Faculty of Science and Informatics, Institute of Informatics, University of Szeged, Szeged, Hungary
| | - László G. Nyúl
- Department of Image Processing and Computer Graphics, Faculty of Science and Informatics, Institute of Informatics, University of Szeged, Szeged, Hungary
| | - Attila Nagy
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Gabriella Kekesi
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Alexandra Büki
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Gyongyi Horvath
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- *Correspondence: Gyongyi Horvath,
| |
Collapse
|
20
|
Abstract
The modern way of life has dramatically affected our biological rhythms. Circadian rhythms, which are generated by an endogenous circadian clock, are observed in a large number of physiological functions including metabolism. Proper peripheral clock synchronization by different signals including appropriate feeding/fasting cycles is essential to coordinate and temporally gate metabolic processes. In this chapter, we emphasize the importance of nutrient sensing by peripheral clocks and highlight the major role of peripheral and central clock communication to locally regulate metabolic processes and ensure optimal energy storage and expenditure. As a consequence, changes in eating behavior and/or bedtime, as occurs upon shift work and jet lag, have direct consequences on metabolism and participate in the increasing prevalence of obesity and associated metabolic diseases such as type 2 diabetes and non-alcoholic fatty liver disease. In this setting, time-restricted feeding has been suggested as an efficient approach to ameliorate metabolic parameters and control body weight.
Collapse
Affiliation(s)
- Yasmine Sebti
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Aurore Hebras
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Benoit Pourcet
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| | - Hélène Duez
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| |
Collapse
|
21
|
Sleep disruption in nonalcoholic fatty liver disease: What is the role of lifestyle and diet? Eur J Gastroenterol Hepatol 2021; 33:e308-e312. [PMID: 33470710 DOI: 10.1097/meg.0000000000002049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Patients with nonalcoholic fatty liver disease (NAFLD) have sleep disruption. The aim of this study is to understand how underlying factors such as diet, degree of liver disease and morningness-eveningness tendencies contribute to this sleep disruption. METHODS Patients with NAFLD were recruited from liver clinics at a University and Veterans Affairs practice. Patients with decompensated cirrhosis were excluded. Patients completed self-reported surveys to evaluate sleep disturbance using the Epworth Sleepiness Scale (ESS) and chronotype (circadian preference) using the morningness-eveningness questionnaire (MEQ). Information on occupation, physical activity and dietary intake were collected at clinic intake. Dietary intake was evaluated via food-frequency questionnaire and analyzed as individual categories or grouped on the basis of dietary composition. RESULTS A 54 patients completed the survey; 37% were female. Median ESS was 8 ± 4.2 and 37% of NAFLD patients were found to have sleep disturbance as defined by ESS >10. Sleep disturbance was common in NAFLD regardless of the liver disease stage. Dietary factors, including higher added sugar (P = 0.01), candy intake (P = 0.01), elevated Ferritin level (P = 0.04) and elevated platelet count (P = 0.05), were significantly associated with sleep disturbance. Chronotype, time to sleep, and duration of sleep were not associated with sleep disruption. CONCLUSIONS Sleep disruption is present in NAFLD regardless of underlying cirrhosis. Interventions aimed at improving dietary and lifestyle practices such as reduced sugar intake may help mitigate the risk for sleep disruption in NAFLD. Further longitudinal studies are needed to further delineate these links.
Collapse
|
22
|
Peris-Sampedro F, Le May MV, Stoltenborg I, Schéle E, Dickson SL. A skeleton in the cupboard in ghrelin research: Where are the skinny dwarfs? J Neuroendocrinol 2021; 33:e13025. [PMID: 34427011 DOI: 10.1111/jne.13025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/09/2021] [Accepted: 08/05/2021] [Indexed: 12/15/2022]
Abstract
Based on studies delivering ghrelin or ghrelin receptor agonists, we have learned a great deal about the importance of the brain ghrelin signalling system for a wide range of physiological processes that include feeding behaviours, growth hormone secretion and glucose homeostasis. Because these processes can be considered as essential to life, the question arises as to why mouse models of depleted ghrelin signalling are not all skinny dwarfs with a host of behavioural and metabolic problems. Here, we provide a systematic detailed review of the phenotype of mice with deficient ghrelin signalling to help better understand the relevance and importance of the brain ghrelin signalling system, with a particular emphasis on those questions that remain unanswered.
Collapse
Affiliation(s)
- Fiona Peris-Sampedro
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marie V Le May
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Iris Stoltenborg
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Erik Schéle
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Suzanne L Dickson
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
23
|
Segev Y, Nujedat H, Arazi E, Assadi MH, Tarasiuk A. Changes in energy metabolism and respiration in different tracheal narrowing in rats. Sci Rep 2021; 11:19166. [PMID: 34580405 PMCID: PMC8476542 DOI: 10.1038/s41598-021-98799-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Why obstructive sleep apnea (OSA) treatment does not completely restore healthy metabolic physiology is unclear. In rats, the need for respiratory homeostasis maintenance following airway obstruction (AO) is associated with a loss of thermoregulation and abnormal metabolic physiology that persists following successful obstruction removal. Here, we explored the effect of two different types of tracheal narrowing, i.e., AO and mild airway obstruction (mAO), and its removal on respiratory homeostasis and metabolic physiology. We show that after ten weeks, mAO vs. AO consumes sufficient energy that is required to maintain respiratory homeostasis and thermoregulation. Obstruction removal was associated with largely irreversible increased feeding associated with elevated serum ghrelin, hypothalamic growth hormone secretagogue receptor 1a, and a phosphorylated Akt/Akt ratio, despite normalization of breathing and energy requirements. Our study supports the need for lifestyle eating behavior management, in addition to endocrine support, in order to attain healthy metabolic physiology in OSA patients.
Collapse
Affiliation(s)
- Yael Segev
- Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, P.O Box 105, Beer-Sheva, 84105, Israel
| | - Haiat Nujedat
- Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, P.O Box 105, Beer-Sheva, 84105, Israel
| | - Eden Arazi
- Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, P.O Box 105, Beer-Sheva, 84105, Israel
| | - Mohammad H Assadi
- Sleep-Wake Disorders Unit, Soroka University Medical Center, P.O. Box 151, Beer-Sheva, 84105, Israel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O Box 105, Beer-Sheva, 84105, Israel
| | - Ariel Tarasiuk
- Sleep-Wake Disorders Unit, Soroka University Medical Center, P.O. Box 151, Beer-Sheva, 84105, Israel.
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O Box 105, Beer-Sheva, 84105, Israel.
| |
Collapse
|
24
|
Koop S, Oster H. Eat, sleep, repeat - endocrine regulation of behavioural circadian rhythms. FEBS J 2021; 289:6543-6558. [PMID: 34228879 DOI: 10.1111/febs.16109] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/23/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
The adaptation of organisms to a rhythmic environment is mediated by an internal timing system termed the circadian clock. In mammals, molecular clocks are found in all tissues and organs. This circadian clock network regulates the release of many hormones, which in turn influence some of the most vital behavioural functions. Sleep-wake cycles are under strict circadian control with strong influence of rhythmic hormones such as melatonin, cortisol and others. Food intake, in contrast, receives circadian modulation through hormones such as leptin, ghrelin, insulin and orexin. A third behavioural output covered in this review is mating and bonding behaviours, regulated through circadian rhythms in steroid hormones and oxytocin. Together, these data emphasize the pervasive influence of the circadian clock system on behavioural outputs and its mediation through endocrine networks.
Collapse
Affiliation(s)
- Sarah Koop
- Centre of Brain, Behavior and Metabolism, Institute of Neurobiology, University of Lübeck, Germany
| | - Henrik Oster
- Centre of Brain, Behavior and Metabolism, Institute of Neurobiology, University of Lübeck, Germany
| |
Collapse
|
25
|
Jacquelot N, Belz GT, Seillet C. Neuroimmune Interactions and Rhythmic Regulation of Innate Lymphoid Cells. Front Neurosci 2021; 15:657081. [PMID: 33994930 PMCID: PMC8116950 DOI: 10.3389/fnins.2021.657081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/29/2021] [Indexed: 01/21/2023] Open
Abstract
The Earth’s rotation around its axis, is one of the parameters that never changed since life emerged. Therefore, most of the organisms from the cyanobacteria to humans have conserved natural oscillations to regulate their physiology. These daily oscillations define the circadian rhythms that set the biological clock for almost all physiological processes of an organism. They allow the organisms to anticipate and respond behaviorally and physiologically to changes imposed by the day/night cycle. As other physiological systems, the immune system is also regulated by circadian rhythms and while diurnal variation in host immune responses to lethal infection have been observed for many decades, the underlying mechanisms that affect immune function and health have only just started to emerge. These oscillations are generated by the central clock in our brain, but neuroendocrine signals allow the synchronization of the clocks in peripheral tissues. In this review, we discuss how the neuroimmune interactions create a rhythmic activity of the innate lymphoid cells. We highlight how the disruption of these rhythmic regulations of immune cells can disturb homeostasis and lead to the development of chronic inflammation in murine models.
Collapse
Affiliation(s)
- Nicolas Jacquelot
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Gabrielle T Belz
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Cyril Seillet
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
26
|
Page AJ. Gastrointestinal Vagal Afferents and Food Intake: Relevance of Circadian Rhythms. Nutrients 2021; 13:nu13030844. [PMID: 33807524 PMCID: PMC7998414 DOI: 10.3390/nu13030844] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 01/20/2023] Open
Abstract
Gastrointestinal vagal afferents (VAs) play an important role in food intake regulation, providing the brain with information on the amount and nutrient composition of a meal. This is processed, eventually leading to meal termination. The response of gastric VAs, to food-related stimuli, is under circadian control and fluctuates depending on the time of day. These rhythms are highly correlated with meal size, with a nadir in VA sensitivity and increase in meal size during the dark phase and a peak in sensitivity and decrease in meal size during the light phase in mice. These rhythms are disrupted in diet-induced obesity and simulated shift work conditions and associated with disrupted food intake patterns. In diet-induced obesity the dampened responses during the light phase are not simply reversed by reverting back to a normal diet. However, time restricted feeding prevents loss of diurnal rhythms in VA signalling in high fat diet-fed mice and, therefore, provides a potential strategy to reset diurnal rhythms in VA signalling to a pre-obese phenotype. This review discusses the role of the circadian system in the regulation of gastrointestinal VA signals and the impact of factors, such as diet-induced obesity and shift work, on these rhythms.
Collapse
Affiliation(s)
- Amanda J. Page
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; ; Tel.: +61-8-8128-4840
- Nutrition, Diabetes and Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institution (SAHMRI), Adelaide, SA 5000, Australia
| |
Collapse
|
27
|
"Shedding Light on Light": A Review on the Effects on Mental Health of Exposure to Optical Radiation. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18041670. [PMID: 33572423 PMCID: PMC7916252 DOI: 10.3390/ijerph18041670] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/11/2021] [Accepted: 02/03/2021] [Indexed: 01/10/2023]
Abstract
In relation to human health and functioning, light, or more specifically optical radiation, plays many roles, beyond allowing vision. These may be summarized as: regulation of circadian rhythms; consequences of direct exposure to the skin; and more indirect effects on well-being and functioning, also related to lifestyle and contact with natural and urban environments. Impact on mental health is relevant for any of these specifications and supports a clinical use of this knowledge for the treatment of psychiatric conditions, such as depression or anxiety, somatic symptom disorder, and others, with reference to light therapy in particular. The scope of this narrative review is to provide a summary of recent findings and evidence on the regulating functions of light on human beings’ biology, with a specific focus on mental health, its prevention and care.
Collapse
|
28
|
Lago-Sampedro A, Ho-Plagaro A, Garcia-Serrano S, Santiago-Fernandez C, Rodríguez-Díaz C, Lopez-Gómez C, Martín-Reyes F, Ruiz-Aldea G, Alcaín-Martínez G, Gonzalo M, Montiel-Casado C, Fernández JR, García-Fuentes E, Rodríguez-Pacheco F. Oleic acid restores the rhythmicity of the disrupted circadian rhythm found in gastrointestinal explants from patients with morbid obesity. Clin Nutr 2021; 40:4324-4333. [PMID: 33531179 DOI: 10.1016/j.clnu.2021.01.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND & AIMS We investigated whether oleic acid (OA), one of the main components of the Mediterranean diet, participates in the regulation of the intestinal circadian rhythm in patients with morbid obesity. METHODS Stomach and jejunum explants from patients with morbid obesity were incubated with oleic acid to analyze the regulation of clock genes. RESULTS Stomach explants showed an altered circadian rhythm in CLOCK, BMAL1, REVERBα, CRY1, and CRY2, and an absence in PER1, PER2, PER3 and ghrelin (p > 0.05). OA led to the emergence of rhythmicity in PER1, PER2, PER3 and ghrelin (p < 0.05). Jejunum explants showed an altered circadian rhythm in CLOCK, BMAL1, PER1 and PER3, and an absence in PER2, REVERBα, CRY1, CRY2 and GLP1 (p > 0.05). OA led to the emergence of rhythmicity in PER2, REVERBα, CRY1 and GLP1 (p < 0.05), but not in CRY2 (p > 0.05). OA restored the rhythmicity of acrophase and increased the amplitude for most of the genes studied in stomach and jejunum explants. OA placed PER1, PER2, PER3, REVERBα, CRY1 and CRY2 in antiphase with regard to CLOCK and BMAL1. CONCLUSIONS There is an alteration in circadian rhythm in stomach and jejunum explants in morbid obesity. OA restored the rhythmicity of the genes related with circadian rhythm, ghrelin and GLP1, although with slight differences between tissues, which could determine a different behaviour of the explants from jejunum and stomach in obesity.
Collapse
Affiliation(s)
- Ana Lago-Sampedro
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Regional Universitario, Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Ailec Ho-Plagaro
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Sara Garcia-Serrano
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Regional Universitario, Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas-CIBERDEM, Málaga, Spain
| | - Concepción Santiago-Fernandez
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Cristina Rodríguez-Díaz
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Carlos Lopez-Gómez
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Flores Martín-Reyes
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Gonzalo Ruiz-Aldea
- Departamento Biología Celular, Genética y Fisiología, Universidad de Málaga, Málaga, Spain
| | - Guillermo Alcaín-Martínez
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Montserrat Gonzalo
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Regional Universitario, Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Custodia Montiel-Casado
- Unidad de Gestión Clínica de Cirugía General, Digestiva y Trasplantes, Hospital Regional Universitario, Málaga, Spain
| | - José R Fernández
- Bioengineering & Chronobiology Labs, atlanTTic Research Center, University of Vigo, Spain
| | - Eduardo García-Fuentes
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain.
| | - Francisca Rodríguez-Pacheco
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas-CIBERDEM, Málaga, Spain
| |
Collapse
|
29
|
Espitia-Bautista E, Escobar C. Addiction-like response in brain and behavior in a rat experimental model of night-eating syndrome. Appetite 2021; 161:105112. [PMID: 33453338 DOI: 10.1016/j.appet.2021.105112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 11/04/2020] [Accepted: 01/07/2021] [Indexed: 11/28/2022]
Abstract
STUDY OBJECTIVES Individuals ailing from night eating syndrome (NES) consume more than 25% of their daily food intake during the normal sleep time, delaying their sleep or waking up in the middle of the night to eat. This study explored two experimental conditions resembling NES in Wistar rats by offering palatable food during the sleep phase, alone or combined with sleep delay. Also we explored their impact on addiction-like changes in the brain and behavior. METHODS Experiment 1 explored the brain response after a first NES-like event; experiment 2 and 3 explored addiction-like behaviors c-Fos and FosB/ΔFosB in corticolimbic regions after 4 weeks exposition to NES-like conditions and after one week of withdrawal, respectively. For all 3 experiments 6 experimental groups were used: 1. Control; 2. Restricted access (1 h) to high-sugar diet (HSD) or to 3. high-fat diet (HFD); 4., Sleep delay for 4 h (SD) (from ZT0-ZT4, rats using slow rotating wheels); 5. SD + HSD; 6. SD + HFD. RESULTS A first event of eating a palatable diet with or without SD was sufficient to stimulate c-Fos and ΔFosB. Along 4 weeks of exposure to the palatable diets rats exhibited escalation and binge eating, which was highest for the HFD. At this stage, SD did not influence behavioral changes nor the neuronal response. After one-week in withdrawal, rats exhibited craving and effort to obtain their palatable diet. The brains of rats previously exposed to sleep delay maintained high levels of FosB/ΔFosB in the accumbens shell and high c-Fos activation in the insular cortex. CONCLUSIONS In our experimental models of NES-like a HFD in the sleep phase and SD are risk factors to develop binge eating and addiction-like behaviors.
Collapse
Affiliation(s)
- Estefania Espitia-Bautista
- Facultad de Medicina, Departamento de Anatomía, Universidad Nacional Autónoma de México, 04510, México City, Mexico
| | - Carolina Escobar
- Facultad de Medicina, Departamento de Anatomía, Universidad Nacional Autónoma de México, 04510, México City, Mexico.
| |
Collapse
|
30
|
Tchio C, Baba K, Piccione G, Tosini G. Removal of melatonin receptor type 1 signalling induces dyslipidaemia and hormonal changes in mice subjected to environmental circadian disruption. Endocrinol Diabetes Metab 2021; 4:e00171. [PMID: 33532613 PMCID: PMC7831213 DOI: 10.1002/edm2.171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/02/2020] [Accepted: 07/05/2020] [Indexed: 11/26/2022] Open
Abstract
Background Melatonin is a hormone secreted by the pineal gland in a circadian rhythmic manner with peak synthesis at night. Melatonin signalling was suggested to play a critical role in metabolism during the circadian disruption. Methods Melatonin-proficient (C3H-f+/+ or WT) and melatonin receptor type 1 knockout (MT1 KO) male and female mice were phase-advanced (6 hours) once a week for 6 weeks. Every week, we measured weight, food intake and basal glucose levels. At the end of the experiment, we sacrificed the animals and measured the blood's plasma for lipids profile (total lipids, phospholipids, triglycerides and total cholesterol), metabolic hormones profiles (ghrelin, leptin, insulin, glucagon, glucagon-like-peptide and resistin) and the body composition. Results Environmental circadian disruption (ECD) did not produce any significant effects in C3H-f+/+, while it increased lipids profile in MT1 KO with the significant increase observed in total lipids and triglycerides. For metabolic hormones profile, ECD decreased plasma ghrelin and increased plasma insulin in MT1 KO females. Under control condition, MT1 KO females have significantly different body weight, fat mass, total lipids and total cholesterol than the control C3H-f+/+ females. Conclusion Our data show that melatonin-proficient mice are not affected by ECD. When the MT1 receptors are removed, ECD induced dyslipidaemia in males and females with females experiencing the most adverse effect. Overall, our data demonstrate that MT1 signalling is an essential modulator of lipid and metabolic homeostasis during ECD.
Collapse
Affiliation(s)
- Cynthia Tchio
- Circadian Rhythms and Sleep Disorders ProgramNeuroscience InstituteAtlantaGAUSA
- Department of Pharmacology and ToxicologyMorehouse School of MedicineNeuroscience InstituteAtlantaGAUSA
| | - Kenkichi Baba
- Circadian Rhythms and Sleep Disorders ProgramNeuroscience InstituteAtlantaGAUSA
- Department of Pharmacology and ToxicologyMorehouse School of MedicineNeuroscience InstituteAtlantaGAUSA
| | - Giuseppe Piccione
- Dipartimento di Medicine VeterinariaUniversita di MessinaMessinaItaly
| | - Gianluca Tosini
- Circadian Rhythms and Sleep Disorders ProgramNeuroscience InstituteAtlantaGAUSA
- Department of Pharmacology and ToxicologyMorehouse School of MedicineNeuroscience InstituteAtlantaGAUSA
| |
Collapse
|
31
|
Fritz EM, Singewald N, De Bundel D. The Good, the Bad and the Unknown Aspects of Ghrelin in Stress Coping and Stress-Related Psychiatric Disorders. Front Synaptic Neurosci 2020; 12:594484. [PMID: 33192444 PMCID: PMC7652849 DOI: 10.3389/fnsyn.2020.594484] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022] Open
Abstract
Ghrelin is a peptide hormone released by specialized X/A cells in the stomach and activated by acylation. Following its secretion, it binds to ghrelin receptors in the periphery to regulate energy balance, but it also acts on the central nervous system where it induces a potent orexigenic effect. Several types of stressors have been shown to stimulate ghrelin release in rodents, including nutritional stressors like food deprivation, but also physical and psychological stressors such as foot shocks, social defeat, forced immobilization or chronic unpredictable mild stress. The mechanism through which these stressors drive ghrelin release from the stomach lining remains unknown and, to date, the resulting consequences of ghrelin release for stress coping remain poorly understood. Indeed, ghrelin has been proposed to act as a stress hormone that reduces fear, anxiety- and depression-like behaviors in rodents but some studies suggest that ghrelin may - in contrast - promote such behaviors. In this review, we aim to provide a comprehensive overview of the literature on the role of the ghrelin system in stress coping. We discuss whether ghrelin release is more than a byproduct of disrupted energy homeostasis following stress exposure. Furthermore, we explore the notion that ghrelin receptor signaling in the brain may have effects independent of circulating ghrelin and in what way this might influence stress coping in rodents. Finally, we examine how the ghrelin system could be utilized as a therapeutic avenue in stress-related psychiatric disorders (with a focus on anxiety- and trauma-related disorders), for example to develop novel biomarkers for a better diagnosis or new interventions to tackle relapse or treatment resistance in patients.
Collapse
Affiliation(s)
- Eva Maria Fritz
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Innsbruck, Austria
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Innsbruck, Austria
| | - Dimitri De Bundel
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
32
|
da Silva FR, Junior AHL, Brant VM, Lôbo ILB, Lancha LOP, Silva A, de Mello MT. The effects of COVID-19 quarantine on eating and sleeping behaviors. NUTRIRE : REVISTA DE SOCIEDADE BRASILEIRA DE ALIMENTACAO E NUTRICAO = JOURNAL OF THE BRAZILIAN SOCIETY OF FOOD AND NUTRITION 2020; 45:25. [PMID: 38624453 PMCID: PMC7523687 DOI: 10.1186/s41110-020-00128-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/17/2020] [Indexed: 12/19/2022]
Abstract
Abstract Since the beginning of the pandemic, the population has been exposed to a substantial period of social isolation, which leads to anxiety, fear, and metabolic and immune impairments. Purpose Considering that sleep restriction influences eating behavior, we highlight that changes in it may occur during the COVID-19 quarantine. Alterations in feeding time can uncouple the body clocks, leading to circadian misalignment and consequently to a disruption in homeostasis and disturbances in many metabolic functions. Method Narrative review. Results Do not apply. Conclusion The increase of body weight is related to increased food intake in response to mental stress and more time spent at home, increased opportunity to feed, and increased visual and olfactory stimulation to eat, which represents a potential risk of overfeeding nowadays. In this article, we postulate that the unusual lifestyle imposed by the COVID-19 quarantine may induce a circadian misalignment, which is capable to induce alterations on eating and sleep behaviors.
Collapse
Affiliation(s)
- Flavia Rodrigues da Silva
- Departamento de Esportes, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, Belo Horizonte, MG 31270-901 Brazil
| | | | - Valdênio Martins Brant
- Departamento de Esportes, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, Belo Horizonte, MG 31270-901 Brazil
| | - Ingrid Ludimila Bastos Lôbo
- Departamento de Esportes, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, Belo Horizonte, MG 31270-901 Brazil
| | - Luciana Oquendo Pereira Lancha
- Escola de Educação Física e Esporte (EEFE), Universidade de São Paulo, São Paulo, Brazil
- Instituto de Bem Estar e Saúde, IBES, São Paulo, Brazil
| | - Andressa Silva
- Departamento de Esportes, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, Belo Horizonte, MG 31270-901 Brazil
| | - Marco Túlio de Mello
- Departamento de Esportes, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, Belo Horizonte, MG 31270-901 Brazil
| |
Collapse
|
33
|
Ertekin D, Kirszenblat L, Faville R, van Swinderen B. Down-regulation of a cytokine secreted from peripheral fat bodies improves visual attention while reducing sleep in Drosophila. PLoS Biol 2020; 18:e3000548. [PMID: 32745077 PMCID: PMC7426065 DOI: 10.1371/journal.pbio.3000548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 08/13/2020] [Accepted: 07/13/2020] [Indexed: 11/29/2022] Open
Abstract
Sleep is vital for survival. Yet under environmentally challenging conditions, such as starvation, animals suppress their need for sleep. Interestingly, starvation-induced sleep loss does not evoke a subsequent sleep rebound. Little is known about how starvation-induced sleep deprivation differs from other types of sleep loss, or why some sleep functions become dispensable during starvation. Here, we demonstrate that down-regulation of the secreted cytokine unpaired 2 (upd2) in Drosophila flies may mimic a starved-like state. We used a genetic knockdown strategy to investigate the consequences of upd2 on visual attention and sleep in otherwise well-fed flies, thereby sidestepping the negative side effects of undernourishment. We find that knockdown of upd2 in the fat body (FB) is sufficient to suppress sleep and promote feeding-related behaviors while also improving selective visual attention. Furthermore, we show that this peripheral signal is integrated in the fly brain via insulin-expressing cells. Together, these findings identify a role for peripheral tissue-to-brain interactions in the simultaneous regulation of sleep quality and attention, to potentially promote adaptive behaviors necessary for survival in hungry animals.
Collapse
Affiliation(s)
- Deniz Ertekin
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Leonie Kirszenblat
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Richard Faville
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
34
|
Ruddick-Collins LC, Morgan PJ, Johnstone AM. Mealtime: A circadian disruptor and determinant of energy balance? J Neuroendocrinol 2020; 32:e12886. [PMID: 32662577 DOI: 10.1111/jne.12886] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/24/2020] [Accepted: 06/14/2020] [Indexed: 12/21/2022]
Abstract
Circadian rhythms play a critical role in the physiological processes involved in energy metabolism and energy balance (EB). A large array of metabolic processes, including the expression of many energy-regulating endocrine hormones, display temporal rhythms that are driven by both the circadian clock and food intake. Mealtime has been shown to be a compelling zeitgeber in peripheral tissue rhythms. Inconsistent signalling to the periphery, because of mismatched input from the central clock vs time of eating, results in circadian disruption in which central and/or peripheral rhythms are asynchronously time shifted or their amplitudes reduced. A growing body of evidence supports the negative health effects of circadian disruption, with strong evidence in murine models that mealtime-induced circadian disruption results in various metabolic consequences, including energy imbalance and weight gain. Increased weight gain has been reported to occur even without differences in energy intake, indicating an effect of circadian disruption on energy expenditure. However, the translation of these findings to humans is not well established because the ability to undertake rigorously controlled dietary studies that explore the chronic effects on energy regulation is challenging. Establishing the neuroendocrine changes in response to both acute and chronic variations in mealtime, along with observations in populations with routinely abnormal mealtimes, may provide greater insight into underlying mechanisms that influence long-term weight management under different meal patterns. Human studies should explore mechanisms through relevant biomarkers; for example, cortisol, leptin, ghrelin and other energy-regulating neuroendocrine factors. Mistiming between aggregate hormonal signals, or between hormones with their receptors, may cause reduced signalling intensity and hormonal resistance. Understanding how mealtimes may impact on the coordination of endocrine factors is essential for untangling the complex regulation of EB. Here a review is provided on current evidence of the impacts of mealtime on energy metabolism and the underlying neuroendocrine mechanisms, with a specific focus on human research.
Collapse
Affiliation(s)
| | - Peter J Morgan
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
| | | |
Collapse
|
35
|
Circadian regulation of appetite and time restricted feeding. Physiol Behav 2020; 220:112873. [PMID: 32194073 DOI: 10.1016/j.physbeh.2020.112873] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/01/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022]
Abstract
The circadian system plays an important role in the temporal regulation of metabolic processes as well as food intake to ensure energy efficiency. The 'master' clock is located within the superchiasmatic nucleus and receives input from the retina so that it can be entrained by the light:dark cycle. In turn, the master clock entrains other clocks in the central nervous system, including areas involved in energy homeostasis such as the arcuate nucleus, and the periphery (e.g. adipose tissue and the gastrointestinal tract). This master clock is reinforced by other zeitgebers such as the timing of food intake and activity. If these zeitgebers desynchronise, such as occurs in high fat diet-induced obesity or shift work conditions, it can lead to a misalignment of circadian clocks, disruption of metabolic processes and the development of metabolic disorders. The timing of food intake is a strong zeitgeber, particularly in the gastrointestinal tract, and therefore time restricted feeding offers potential for the treatment of diet and shift work induced metabolic disorders. This review will focus on the role of the circadian system in food intake regulation and the effect of environment factors, such as high fat diet feeding or shift work, on the temporal regulation of food intake along with the benefits of time restricted feeding.
Collapse
|
36
|
Moreno JP, Crowley SJ, Alfano CA, Thompson D. Physiological mechanisms underlying children's circannual growth patterns and their contributions to the obesity epidemic in elementary school age children. Obes Rev 2020; 21:e12973. [PMID: 31737994 PMCID: PMC7002188 DOI: 10.1111/obr.12973] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/18/2019] [Accepted: 10/18/2019] [Indexed: 12/20/2022]
Abstract
Several studies since the 1990s have demonstrated that children increase their body mass index at a faster rate during summer months compared with the school year, leading some to conclude that the out-of-school summer environment is responsible. Other studies, however, have suggested that seasonality may play a role in children's height and weight changes across the year. This article reviews evidence for seasonal differences in the rate of children's height and weight gain and proposes potential physiological mechanisms that may explain these seasonal variations.
Collapse
Affiliation(s)
- Jennette P Moreno
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Stephanie J Crowley
- Biological Rhythm Research Laboratory, Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois
| | - Candice A Alfano
- Sleep and Anxiety Center of Houston (SACH), Department of Psychology, University of Houston, Houston, Texas
| | - Debbe Thompson
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
37
|
Assadi MH, Segev Y, Tarasiuk A. Upper Airway Obstruction Elicited Energy Imbalance Leads to Growth Retardation that Persists after the Obstruction Removal. Sci Rep 2020; 10:3206. [PMID: 32081973 PMCID: PMC7035324 DOI: 10.1038/s41598-020-60226-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 01/08/2020] [Indexed: 12/28/2022] Open
Abstract
Upper airway obstruction can lead to growth retardation by unclear mechanisms. We explored the effect of upper airway obstruction in juvenile rats on whole-body energy balance, growth plate metabolism, and growth. We show that after seven weeks, obstructed animals’ ventilation during room air breathing increased, and animals grew less due to abnormal growth plate metabolism. Increased caloric intake in upper airway-obstructed animals did not meet increased energy expenditure associated with increased work of breathing. Decreased whole-body energy balance induced hindrance of bone elongation following obstruction removal, and array pathways regulating growth plate development and marrow adiposity. This is the first study to show that rapidly growing animals cannot consume enough calories to maintain their energy homeostasis, leading to an impediment in growth in the effort to save energy.
Collapse
Affiliation(s)
- Mohammad H Assadi
- Sleep-Wake Disorders Unit, Soroka University Medical Center, P.O. Box 151, Beer-Sheva, 84105, Israel.,Shraga Segal Department of Microbiology and Immunology, Ben-Gurion University of the Negev, P.O. Box 105, Beer-Sheva, 84105, Israel
| | - Yael Segev
- Shraga Segal Department of Microbiology and Immunology, Ben-Gurion University of the Negev, P.O. Box 105, Beer-Sheva, 84105, Israel
| | - Ariel Tarasiuk
- Sleep-Wake Disorders Unit, Soroka University Medical Center, P.O. Box 151, Beer-Sheva, 84105, Israel. .,Department of Physiology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 105, Beer-Sheva, 84105, Israel.
| |
Collapse
|
38
|
Dissi M, Ibrahim S, Tanko Y, Mohammed A. Models of modern-day circadian rhythm disruption and their diabetogenic potentials in adult male Wistar rats. SAUDI JOURNAL FOR HEALTH SCIENCES 2020. [DOI: 10.4103/sjhs.sjhs_69_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
39
|
Qin C, Sun J, Wang J, Han Y, Yang H, Shi Q, Lv Y, Hu P. Discovery of differentially expressed genes in the intestines of Pelteobagrus vachellii within a light/dark cycle. Chronobiol Int 2019; 37:339-352. [PMID: 31809585 DOI: 10.1080/07420528.2019.1690498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In aquaculture, it is necessary to determine of the diurnal biological variations in the intestines to determine an appropriate feeding schedule. The present study aimed to examine the transcriptomes of the Pelteobagrus vachellii intestines at four time points (0 h, 6 h, 12 h, and 18 h) within a light/dark cycle. In comparison with the zeitgeber time 0 (ZT0) transcriptomes, we identified 37,842 unigenes with significant differential expression, including 6,638; 9,626; and 7,938 that genes upregulated, and 3,507; 4,703; and 5,412 genes that were down regulated at 4, 12, and 24 h respectively. The differentially expressed unigenes were subjected to enrichment analysis, which indicated the involvement of the major digestive pathways, including digestion of protein, lipid and carbohydrate, catabolic process (protein, carbohydrate and lipid), and circadian rhythm. We selected 73 key differentially expressed genes (DEGs) from among these pathways and identified DEGs that showed increased expression at night, including those encoding trypsin-3, chymotrypsinogen 2, amino acid transporter, maltase-glucoamylase, facilitated glucose transporter, lipase, phospholipase, fatty acid-binding protein, fatty acid synthase, long-chain fatty acid transport protein, and apolipoprotein. Moreover, DEGs involved of circadian rhythm were identified, including brain-muscle-Arnt-like 1 (BMAL1), cryptochrome-1, circadian locomoter output cycles protein kaput (CLOCK) and period circadian protein homolog 1-3. Finally, the expression levels of 12 unigenes were analyzed using quantitative real-time PCR, which were in accordance with RNA-sequencing analysis. In general, the expression of genes related to the digestion of proteins, lipids, and carbohydrates showed upregulated expression at night; however, the peak time of expression of transporters for different nutrition molecules showed more diversification within the light/dark cycle.
Collapse
Affiliation(s)
- Chuanjie Qin
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, PR China.,College of Life Science, Neijiang Normal University, Neijiang, PR China
| | | | - Jun Wang
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, PR China.,College of Life Science, Neijiang Normal University, Neijiang, PR China
| | | | - He Yang
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, PR China.,College of Life Science, Neijiang Normal University, Neijiang, PR China
| | - Qingchao Shi
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, PR China.,College of Life Science, Neijiang Normal University, Neijiang, PR China
| | - Yunyun Lv
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, PR China.,College of Life Science, Neijiang Normal University, Neijiang, PR China
| | - Peng Hu
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, PR China.,College of Life Science, Neijiang Normal University, Neijiang, PR China
| |
Collapse
|
40
|
Ji Y, Elkin K, Yip J, Guan L, Han W, Ding Y. From circadian clocks to non-alcoholic fatty liver disease. Expert Rev Gastroenterol Hepatol 2019; 13:1107-1112. [PMID: 31645151 DOI: 10.1080/17474124.2019.1684899] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/22/2019] [Indexed: 02/08/2023]
Abstract
Introduction: The circadian rhythm is an integral regulator of various endocrine processes in the body, including sleep-wake cycles, hormonal regulation, and metabolism. In addition to metabolic, genetic, and environmental factors, a dysregulated circadian rhythm resulting from lifestyle changes has been implicated in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). An accumulating body of evidence also supports strong association between NAFLD and metabolic disorder, the pathogenesis of which is related to periodic fluctuations in hormonal homeostasis. It is clear that endocrine and circadian rhythms are tightly interconnected. Generally, the circadian rhythm regulates flux patterns of physiological functions. The present review will discuss the modulation of bodily processes by the circadian rhythm with specific attention to the regulation of NAFLD by leptin and related hormones.Areas covered: PubMed/MEDLINE was searched for articles related to concomitant occurrence of NAFLD and T2DM between January 1995 and September 2019. Areas covered included epidemiological, physiology and pathophysiology aspects.Expert opinion: NAFLD and NASH are increasingly prevalent and may be largely mitigated with effective lifestyle modification and, potentially, circadian rhythm stabilization. Improved knowledge of the specific pathogenesis of NAFLD in addition to enhanced diagnostic screening tools and prediction of future disease burden is imperative.
Collapse
Affiliation(s)
- Yu Ji
- Department of General Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, School of Medicine, Wayne State University, Detroit, MI, USA
- Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI, USA
| | - Kenneth Elkin
- Department of Neurosurgery, School of Medicine, Wayne State University, Detroit, MI, USA
| | - James Yip
- Department of Neurosurgery, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Longfei Guan
- Department of Neurosurgery, School of Medicine, Wayne State University, Detroit, MI, USA
- Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI, USA
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Wei Han
- Department of General Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, School of Medicine, Wayne State University, Detroit, MI, USA
- Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI, USA
| |
Collapse
|
41
|
Balsevich G, Abizaid A, Chen A, Karatsoreos I, Schmidt M. Stress and glucocorticoid modulation of feeding and metabolism. Neurobiol Stress 2019; 11:100171. [PMID: 31193462 PMCID: PMC6529856 DOI: 10.1016/j.ynstr.2019.100171] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 05/07/2019] [Accepted: 05/14/2019] [Indexed: 12/04/2022] Open
Abstract
This perspective highlights research presented as part of the symposium entitled, "Stress and Glucocorticoid Modulation of Feeding and Metabolism" at the 2018 Neurobiology of Stress Workshop held in Banff, AB, Canada. The symposium comprised five researchers at different career stages who each study different aspects of the interaction between the stress response and metabolic control. Their collective results reveal the complexity of this relationship in terms of behavioural and physiological outcomes. Their work emphasizes the need to consider the level of interaction (cellular, tissue, systems) as well as the timing and context in which the interaction is studied. Rather than a comprehensive review on the work presented at the Symposium, here we discuss recurring themes that emerged at the biennial workshop, which address new avenues of research that will drive the field forward.
Collapse
Affiliation(s)
- G. Balsevich
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | - A. Abizaid
- Institute of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - A. Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstrasse 2 – 10, Munich, 80804, Germany
| | - I.N. Karatsoreos
- Department of Integrative Physiology and Neuroscience, Washington State University, 1815 Ferdinand's Lane, Pullman, WA, 99164, United States
| | - M.V. Schmidt
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstrasse 2 – 10, Munich, 80804, Germany
| |
Collapse
|
42
|
Likitnukul S, Kalandakanond-Thongsong S, Thammacharoen S. Evidence of growth hormone effect on plasma leptin in diet-induced obesity and diet-resistant rats. ASIAN BIOMED 2019. [DOI: 10.1515/abm-2019-0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Background
Plasma leptin is regulated by several factors, including growth hormone (GH), which influences the pathophysiology of obesity.
Objective
To demonstrate the short-term effect of GH on plasma leptin levels in 3 conditions in vivo with the different amount of body fat mass.
Methods
Adult male Wistar rats were fed with standard chow or hypercaloric diet (HC). The HC rats were demonstrated as HC-feeding obese (HC-O) and HC-feeding resistant (HC-R) rats. Then, they were treated with GH or saline for 3 days. Basal plasma leptin levels were measured at 24 and 32 h. For meal-induced condition, all rats were fed for 2 hand plasma leptin was measured. Further 16-h fasting period, plasma leptin, insulin, and insulin sensitivity indexes were determined.
Results
The short-term GH treatment decreased basal plasma leptin at 32 h after the first GH injection in HC-O rats. However, GH treatment had no effect on meal-induced plasma leptin in all rats. Furthermore, GH treatment attenuated fasting effect on plasma leptin in control and HC-R rats. The insulin resistance (IR) induced by the short-term GH treatment was demonstrated by higher fasting plasma insulin and the increased homeostasis model of IR in HC-R rats.
Conclusions
The study demonstrates the important role of greater fat mass in HC-O rats, which results in decreased basal plasma leptin after short-term GH treatment. For meal-induced condition, GH had no effect on plasma leptin in all rats. Interestingly, GH could attenuate fasting effect on plasma leptin in rats that have lower fat mass.
Collapse
Affiliation(s)
- Sutharinee Likitnukul
- Department of Veterinary Physiology, Faculty of Veterinary Science, Chulalongkorn University , Bangkok 10330 , Thailand
| | | | - Sumpun Thammacharoen
- Department of Veterinary Physiology, Faculty of Veterinary Science, Chulalongkorn University , Bangkok 10330 , Thailand
| |
Collapse
|
43
|
Tilston TW, Brown RD, Wateridge MJ, Arms-Williams B, Walker JJ, Sun Y, Wells T. A Novel Automated System Yields Reproducible Temporal Feeding Patterns in Laboratory Rodents. J Nutr 2019; 149:1674-1684. [PMID: 31287142 PMCID: PMC6736427 DOI: 10.1093/jn/nxz116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/23/2019] [Accepted: 05/07/2019] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND The impact of temporal feeding patterns remains a major unanswered question in nutritional science. Progress has been hampered by the absence of a reliable method to impose temporal feeding in laboratory rodents, without the confounding influence of food-hoarding behavior. OBJECTIVE The aim of this study was to develop and validate a reliable method for supplying crushed diets to laboratory rodents in consistent, relevant feeding patterns for prolonged periods. METHODS We programmed our experimental feeding station to deliver a standard diet [StD; Atwater Fuel Energy (AFE) 13.9% fat] or high-fat diet (HFD; AFE 45% fat) during nocturnal grazing [providing 1/24th of the total daily food intake (tdF/I) of ad libitum-fed controls every 30 min] and meal-fed (3 × 1-h periods of ad libitum feeding) patterns in male rats (Sprague-Dawley: 4 wk old, 72-119 g) and mice [C57/Bl6J wild-type (WT): 6 mo old, 29-37 g], and ghrelin-null littermates (Ghr-/-; 27-34 g). RESULTS Grazing yielded accurate, consistent feeding events in rats, with an approximately linear rise in nocturnal cumulative food intake [tdF/I (StD): 97.4 ± 1.5% accurate compared with manual measurement; R2 = 0.86; tdF/I (HFD): 99.0 ± 1.4% accurate; R2 = 0.86]. Meal-feeding produced 3 nocturnal meals of equal size and duration in StD-fed rats (tdF/I: 97.4 ± 0.9% accurate; R2 = 0.90), whereas the second meal size increased progressively in HFD-fed rats (44% higher on day 35 than on day 14; P < 0.01). Importantly, cumulative food intake in grazing and meal-fed rats was identical. Similar results were obtained in WT mice except that less restricted grazing induced hyperphagia (compared with meal-fed WT mice; P < 0.05 from day 1). This difference was abolished in Ghr-/- mice, with meal initiation delayed and meal duration enhanced. Neither pattern elevated corticosterone secretion in rats, but meal-feeding aligned ultradian pulses. CONCLUSIONS We have established a consistent, measurable, researcher-defined, stress-free method for imposing temporal feeding patterns in rats and mice. This approach will facilitate progress in understanding the physiologic impact of feeding patterns.
Collapse
Affiliation(s)
- Thomas W Tilston
- Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Richard D Brown
- Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Matthew J Wateridge
- Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Bradley Arms-Williams
- Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Jamie J Walker
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, United Kingdom
- Wellcome Trust Centre for Biomedical Modelling and Analysis, University of Exeter, Exeter, United Kingdom
- EPSRC Centre for Predictive Modelling in Healthcare, University of Exeter, Exeter, United Kingdom
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, United Kingdom
| | - Yuxiang Sun
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - Timothy Wells
- Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
44
|
Löfgren M, Holmberg E, Bäckström T, Egecioglu E, Dickson SL. The additive effect of allopregnanolone on ghrelin's orexigenic effect in rats. Neuropeptides 2019; 76:101937. [PMID: 31253440 DOI: 10.1016/j.npep.2019.101937] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 06/08/2019] [Accepted: 06/12/2019] [Indexed: 11/20/2022]
Abstract
The progesterone metabolite, allopregnanolone (AlloP), is a GABAA receptor modulating steroid and is known to have orexigenic and pro-obesity effects. The neurobiological mechanisms underpinning these effects are most likely due to enhanced GABAergic signaling in the lateral arcuate nucleus (ARC) and medial paraventricular nucleus (PVN) of the hypothalamus. Inspired by the finding that GABAergic signaling is also important for the orexigenic effects of the circulating hormone, ghrelin, we sought to determine the extent to which AlloP (one of the most potent endogenous GABAA-receptor modulators) operates alongside ghrelin to enhance food intake. Male rats with ad libitum access to standard chow were injected intravenously with AlloP and/or ghrelin, alone or in combination. The intake of the standard chow was greater after AlloP 1 mg/kg together with ghrelin 30 μg/kg than with 30 μg/kg ghrelin alone. Food intake was also increased for the combined treatment of AlloP 0.5 mg/kg + ghrelin 10 μg/kg, AlloP 1 mg/kg + ghrelin 10 μg/kg, and AlloP 0.5 mg/kg + ghrelin 30 μg/kg. There was no significant difference in food intake between the two ghrelin doses or between the two doses of AlloP and the vehicle. In electrophysiological studies, physiologically relevant concentrations of AlloP prolonged the current decay time of spontaneous inhibitory post-synaptic current of dissociated cells of the ARC and PVN. We conclude that AlloP enhances the hyperphagic effect of ghrelin, findings of potential relevance for the hyperphagia associated with the luteal phase of the reproductive cycle.
Collapse
Affiliation(s)
- Magnus Löfgren
- Department of Clinical Sciences, Obstetrics and Gynaecology, Umeå University Hospital, SE-Building QA, 3rd floor, 901 85 Umeå, Sweden.
| | - Ellinor Holmberg
- Department of Clinical Sciences, Obstetrics and Gynaecology, Umeå University Hospital, SE-Building QA, 3rd floor, 901 85 Umeå, Sweden
| | - Torbjörn Bäckström
- Department of Clinical Sciences, Obstetrics and Gynaecology, Umeå University Hospital, SE-Building QA, 3rd floor, 901 85 Umeå, Sweden
| | - Emil Egecioglu
- Department of Experimental Medical Science, Appetite Regulation Unit, Faculty of Medicine, Lund University, 221 84 Lund, Sweden
| | - Suzanne L Dickson
- Institute for Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 11, P.O. Box 434, SE-405 30 Gothenburg, Sweden
| |
Collapse
|
45
|
The circadian clock control of adipose tissue physiology and metabolism. Auton Neurosci 2019; 219:66-70. [DOI: 10.1016/j.autneu.2019.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/08/2019] [Accepted: 05/07/2019] [Indexed: 12/16/2022]
|
46
|
Abstract
Feeding, which is essential for all animals, is regulated by homeostatic mechanisms. In addition, food consumption is temporally coordinated by the brain over the circadian (~24 h) cycle. A network of circadian clocks set daily windows during which food consumption can occur. These daily windows mostly overlap with the active phase. Brain clocks that ensure the circadian control of food intake include a master light-entrainable clock in the suprachiasmatic nuclei of the hypothalamus and secondary clocks in hypothalamic and brainstem regions. Metabolic hormones, circulating nutrients and visceral neural inputs transmit rhythmic cues that permit (via close and reciprocal molecular interactions that link metabolic processes and circadian clockwork) brain and peripheral organs to be synchronized to feeding time. As a consequence of these complex interactions, growing evidence shows that chronodisruption and mistimed eating have deleterious effects on metabolic health. Conversely, eating, even eating an unbalanced diet, during the normal active phase reduces metabolic disturbances. Therefore, in addition to energy intake and dietary composition, appropriately timed meal patterns are critical to prevent circadian desynchronization and limit metabolic risks. This Review provides insight into the dual modulation of food intake by homeostatic and circadian processes, describes the mechanisms regulating feeding time and highlights the beneficial effects of correctly timed eating, as opposed to the negative metabolic consequences of mistimed eating.
Collapse
Affiliation(s)
- Etienne Challet
- Circadian clocks and metabolism team, Institute of Cellular and Integrative Neurosciences, Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, Strasbourg, France.
| |
Collapse
|
47
|
Perello M, Cabral A, Cornejo MP, De Francesco PN, Fernandez G, Uriarte M. Brain accessibility delineates the central effects of circulating ghrelin. J Neuroendocrinol 2019; 31:e12677. [PMID: 30582239 DOI: 10.1111/jne.12677] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/04/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022]
Abstract
Ghrelin is a hormone produced in the gastrointestinal tract that acts via the growth hormone secretagogue receptor. In the central nervous system, ghrelin signalling is able to recruit different neuronal targets that regulate the behavioural, neuroendocrine, metabolic and autonomic effects of the hormone. Notably, several studies using radioactive or fluorescent variants of ghrelin have found that the accessibility of circulating ghrelin into the mouse brain is both strikingly low and restricted to some specific brain areas. A variety of studies addressing central effects of systemically injected ghrelin in mice have also provided indirect evidence that the accessibility of plasma ghrelin into the brain is limited. Here, we review these previous observations and discuss the putative pathways that would allow plasma ghrelin to gain access into the brain together with their physiological implications. Additionally, we discuss some potential features regarding the accessibility of plasma ghrelin into the human brain based on the observations reported by studies that investigate the consequences of ghrelin administration to humans.
Collapse
Affiliation(s)
- Mario Perello
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Agustina Cabral
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - María P Cornejo
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Pablo N De Francesco
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Gimena Fernandez
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Maia Uriarte
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
48
|
Godos J, Ferri R, Caraci F, Cosentino FII, Castellano S, Galvano F, Grosso G. Adherence to the Mediterranean Diet is Associated with Better Sleep Quality in Italian Adults. Nutrients 2019; 11:E976. [PMID: 31035395 PMCID: PMC6566275 DOI: 10.3390/nu11050976] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Sleep quality has been associated with human health and diseases, including cognitive decline and dementia; however major determinants of sleep disorders are largely unknown. The aim of this study was to evaluate the association between sleep quality and adherence to the Mediterranean dietary pattern in a sample of Italian adults. METHODS A total of 1936 individuals were recruited in the urban area of Catania during 2014-2015 through random sampling. A food frequency questionnaire and validated instruments were used to assess the adherence to the Mediterranean diet and sleep quality (Pittsburg sleep quality index). Multivariate logistic regressions were performed to determine the association between exposure and outcome. RESULTS A total of 1314 individuals (67.9% of the cohort) reported adequate sleep quality: for each point increase of the Mediterranean diet score, individuals were 10% more likely to have adequate sleep quality. In an additional analysis stratifying the sample by weight status, the association between sleep quality and high adherence to the Mediterranean diet was observed only among normal/overweight individuals but not in obese participants. CONCLUSIONS high adherence to a Mediterranean diet is associated with better sleep quality either toward direct effect on health or indirect effects through improvement of weight status.
Collapse
Affiliation(s)
- Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy.
| | | | - Filippo Caraci
- Oasi Research Institute - IRCCS, 94018 Troina, Italy.
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy.
| | | | - Sabrina Castellano
- Department of Educational Sciences, University of Catania, 95124 Catania, Italy.
| | - Fabio Galvano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy.
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy.
| |
Collapse
|
49
|
Qiu J, Zhang J, Zhou Y, Li X, Li H, Liu J, Gou K, Zhao J, Cui S. MicroRNA-7 inhibits melatonin synthesis by acting as a linking molecule between leptin and norepinephrine signaling pathways in pig pineal gland. J Pineal Res 2019; 66:e12552. [PMID: 30618087 DOI: 10.1111/jpi.12552] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 12/16/2018] [Accepted: 12/28/2018] [Indexed: 12/21/2022]
Abstract
MicroRNAs, including microRNA-7 (miR-7), are important modulators of numerous gene expressions and the related biological processes. Melatonin is a key hormone regulating daily and seasonal rhythms, in which a variety of positive and negative regulatory factors, such as norepinephrine (NE) and leptin, are involved. However, the interactions among these factors and the mechanisms remain to be elucidated. The aims of the present study were to identify the functions and the related mechanisms of miR-7 in regulating melatonin synthesis and secretion through in vitro and in vivo experiments in pineal gland of pigs, which is an important animal model for agricultural and biomedical studies. Our results firstly show that miR-7 is specifically expressed in porcine pinealocytes and negatively regulates melatonin synthesis. The further functional studies show that the dynamic expression levels of miR-7 are contrary to the melatonin levels throughout the day, and the forced inhibition of endogenous miR-7 in porcine pinealocytes sharply increases arylalkylamine N-acetyltransferase (AANAT) expression by 80.0% (P = 0.0031) and melatonin levels by 81.0% (P = 0.0421), whereas miR-7 over-expression down-regulates AANAT expression by 38.6% (P = 0.0004) and melatonin levels by 37.6% (P = 0.0212). In addition, the miR-7 expression is up-regulated by leptin through the JAK/STAT3 signaling pathway, and the in vivo intracerebroventricular injection of leptin increases miR-7 expression by 80.0% (P = 0.0044) in porcine pineal glands and reduces melatonin levels by 57.1% (P = 0.0060) compared with the controls. This functional inhibition of melatonin synthesis by miR-7 is accomplished by its binding to the 3'-UTR of Raf1. Further, our results demonstrate that the RAF1/MEK/ERK signaling pathway mediates NE-induced AANAT expression, whereas leptin attenuates NE's function through miR-7. Taken together, the results demonstrated that leptin activates the JAK/STAT3 signaling pathway to increase the expression of miR-7, which acts as a negative regulatory molecule inhibiting NE-activated RAF1/MEK/ERK signaling pathway by targeting Raf1, resulting in decreased AANAT expression and melatonin synthesis. These findings suggest that miR-7 is a novel negative regulator of melatonin synthesis and links leptin- and NE-mediated signaling pathways in porcine pineal glands, which will contribute to our understanding in the establishment of the biological rhythms resulting from melatonin.
Collapse
Affiliation(s)
- Jingtao Qiu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jinglin Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yewen Zhou
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xin Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Hongjiao Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jiali Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Kemian Gou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jianguo Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Sheng Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
50
|
Kim P, Oster H, Lehnert H, Schmid SM, Salamat N, Barclay JL, Maronde E, Inder W, Rawashdeh O. Coupling the Circadian Clock to Homeostasis: The Role of Period in Timing Physiology. Endocr Rev 2019; 40:66-95. [PMID: 30169559 DOI: 10.1210/er.2018-00049] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 07/06/2018] [Indexed: 01/01/2023]
Abstract
A plethora of physiological processes show stable and synchronized daily oscillations that are either driven or modulated by biological clocks. A circadian pacemaker located in the suprachiasmatic nucleus of the ventral hypothalamus coordinates 24-hour oscillations of central and peripheral physiology with the environment. The circadian clockwork involved in driving rhythmic physiology is composed of various clock genes that are interlocked via a complex feedback loop to generate precise yet plastic oscillations of ∼24 hours. This review focuses on the specific role of the core clockwork gene Period1 and its paralogs on intra-oscillator and extra-oscillator functions, including, but not limited to, hippocampus-dependent processes, cardiovascular function, appetite control, as well as glucose and lipid homeostasis. Alterations in Period gene function have been implicated in a wide range of physical and mental disorders. At the same time, a variety of conditions including metabolic disorders also impact clock gene expression, resulting in circadian disruptions, which in turn often exacerbates the disease state.
Collapse
Affiliation(s)
- Pureum Kim
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | - Hendrik Lehnert
- Department of Internal Medicine 1, University of Lübeck, Lübeck, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Sebastian M Schmid
- Department of Internal Medicine 1, University of Lübeck, Lübeck, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Nicole Salamat
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Johanna L Barclay
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Erik Maronde
- Department of Anatomy, Goethe University Frankfurt, Frankfurt, Germany
| | - Warrick Inder
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Department of Diabetes and Endocrinology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Oliver Rawashdeh
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|