1
|
Correa‐da‐Silva F, Berkhout JB, Schouten P, Sinnema M, Stumpel CTRM, Curfs LMG, Höybye C, Mahfouz A, Meijer OC, Pereira AM, Fliers E, Swaab DF, Kalsbeek A, Yi C. Selective changes in vasopressin neurons and astrocytes in the suprachiasmatic nucleus of Prader-Willi syndrome subjects. J Neuroendocrinol 2025; 37:e70015. [PMID: 40055943 PMCID: PMC12045672 DOI: 10.1111/jne.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 05/03/2025]
Abstract
The hypothalamic suprachiasmatic nucleus (SCN) hosts the central circadian pacemaker and regulates daily rhythms in physiology and behavior. The SCN is composed of peptidergic neuron populations expressing arginine vasopressin (AVP) and vasoactive intestinal polypeptide (VIP), as well as glial cells. Patients with Prader-Willi Syndrome (PWS) commonly experience circadian disturbances, which are particularly evident in their sleep/wake patterns. Using publicly available single-cell RNA sequencing data, we assessed the cell-type specificity of PWS-causative genes in murine SCN, which revealed the differential presence of PWS-related genes in glial and neural subpopulations. We then investigated neurons and glial cells in the SCN using immunohistochemistry in the postmortem hypothalami of PWS subjects and matched controls. We profiled neural populations characterized by AVP and VIP, astroglia characterized by glial fibrillary acid protein (GFAP), and microglia marked by ionized calcium-binding adapter molecule 1 (Iba1) and NADPH oxidase 2 (NOX2). Our analysis revealed an increased total number, neuronal density, and relative staining intensity of AVP-containing neurons in the PWS compared to controls while VIP-containing cells were unaltered. In contrast, GFAP-expressing astroglial cells were significantly lower in PWS subjects. Moreover, we did not detect any differences in microglia between PWS subjects and controls. Collectively, our findings show that PWS selectively affects AVP-containing neurons and GFAP-expressing astrocytes in the SCN. As each of these cell populations can affect the daily rhythmicity of the SCN biological clock machinery, the disruption of these cells may contribute to the circadian disturbances in patients with PWS.
Collapse
Affiliation(s)
- Felipe Correa‐da‐Silva
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Gastroenterology Endocrinology and MetabolismAmsterdamThe Netherlands
- Department of Clinical Chemistry, Laboratory of EndocrinologyAmsterdam University Medical Center, Location AMCAmsterdamThe Netherlands
- Netherlands Institute for NeuroscienceAmsterdamThe Netherlands
| | - Jari B. Berkhout
- Dept. of Medicine Div. EndocrinologyLeiden University Medical CentreLeidenThe Netherlands
| | - Pim Schouten
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Gastroenterology Endocrinology and MetabolismAmsterdamThe Netherlands
| | - Margje Sinnema
- Department of Clinical GeneticsMaastricht University Medical CenterMaastrichtThe Netherlands
| | | | - Leopold M. G. Curfs
- Governor Kremers CentreMaastricht University Medical CentreMaastrichtThe Netherlands
| | - Charlotte Höybye
- Department of Endocrinology and Department of Molecular Medicine and SurgeryKarolinska University Hospital and Karolinska InstituteStockholmSweden
| | - Ahmed Mahfouz
- Delft Bioinformatics LabTechnical University DelftDelftThe Netherlands
- Dept. of Human GeneticsLeiden University Medical CentreLeidenThe Netherlands
| | - Onno C. Meijer
- Dept. of Medicine Div. EndocrinologyLeiden University Medical CentreLeidenThe Netherlands
| | - Alberto M. Pereira
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Gastroenterology Endocrinology and MetabolismAmsterdamThe Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Gastroenterology Endocrinology and MetabolismAmsterdamThe Netherlands
| | - Dick F. Swaab
- Netherlands Institute for NeuroscienceAmsterdamThe Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Gastroenterology Endocrinology and MetabolismAmsterdamThe Netherlands
- Department of Clinical Chemistry, Laboratory of EndocrinologyAmsterdam University Medical Center, Location AMCAmsterdamThe Netherlands
- Netherlands Institute for NeuroscienceAmsterdamThe Netherlands
| | - Chun‐Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Gastroenterology Endocrinology and MetabolismAmsterdamThe Netherlands
- Department of Clinical Chemistry, Laboratory of EndocrinologyAmsterdam University Medical Center, Location AMCAmsterdamThe Netherlands
- Netherlands Institute for NeuroscienceAmsterdamThe Netherlands
| |
Collapse
|
2
|
Lan H, Wu K, Deng C, Wang S. Morning vs. evening: the role of exercise timing in enhancing fat oxidation in young men. Front Physiol 2025; 16:1574757. [PMID: 40337243 PMCID: PMC12055498 DOI: 10.3389/fphys.2025.1574757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025] Open
Abstract
Objective This study aimed to investigate the acute effects of exercise timing (morning vs. evening) on fat oxidation and energy expenditure in young men, with a focus on interactions between exercise and meal timing. Methods Eighteen male college students (23.47 ± 2.11 years) completed a randomized crossover trial under five conditions: sedentary control (SC), exercise before breakfast (EBB), exercise after breakfast (EAB), exercise before dinner (EBD), and exercise after dinner (EAD). Indirect calorimetry (COSMED K5) measured substrate utilization during exercise, post-exercise recovery (0-4 h), and the following morning. Total exercise volume (running distance) was standardized, and energy expenditure was normalized to body weight (kcal/kg). Results During the sedentary control test, participants showed similar trends in total energy expenditure. Dring exercise, the EBB group demonstrated significantly higher fat expenditure compared to EAB (P < 0.05), EBD (P < 0.01), and EAD (P < 0.01). Morning exercise overall exhibited superior fat oxidation (P < 0.01). Post-exercise (0-4 h), EBB sustained elevated fat utilization (P < 0.01 vs. EBD/EAD), while EAD showed enhanced fat oxidation the following morning (P < 0.01 vs. EAB). Conclusion The findings suggest that exercise timing may influence temporal patterns of fat oxidation, with morning fasting potentially favoring acute lipid utilization, while evening exercise appears to correlate with delayed metabolic adjustments. Although total energy expenditure remained comparable across conditions, the observed shifts in substrate allocation imply a possible circadian-sensitive modulation of energy partitioning. These preliminary observations underscore the need for further investigation to clarify the long-term physiological relevance of such timing-dependent metabolic responses.
Collapse
Affiliation(s)
| | | | | | - Songtao Wang
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| |
Collapse
|
3
|
Sakai K, Nakazato Y, Shiimura Y, Zhang W, Nakazato M. Ghrelin-LEAP2 interactions along the stomach-liver axis. Endocr J 2025; 72:341-353. [PMID: 39756956 PMCID: PMC11997273 DOI: 10.1507/endocrj.ej24-0543] [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: 10/02/2024] [Accepted: 11/16/2024] [Indexed: 01/07/2025] Open
Abstract
Ghrelin produced in the stomach promotes food intake and GH secretion, and acts as an anabolic peptide during starvation. Ghrelin binds to the growth hormone secretagogue receptor, a G protein-coupled receptor (GPCR), whose high-resolution complex structures have been determined in the apo state and when bound to an antagonist. Anamorelin, a low-molecular-weight ghrelin agonist, has been launched in Japan for the treatment of cancer cachexia, and its therapeutic potential has attracted attention due to the various biological activities of ghrelin. In 2019, liver-expressed antimicrobial peptide (LEAP2), initially discovered as an antimicrobial peptide produced in the liver, was identified to be upregulated in the stomach of diet-induced obese mice after vertical sleeve gastrectomy. LEAP2 binds to the GHSR and antagonizes ghrelin's activities. The serum concentrations of human LEAP2 are positively correlated with body mass index, body fat accumulation, and fasting serum concentrations of glucose and triglyceride. Serum LEAP2 elevated and ghrelin reduced in obesity. Ghrelin and LEAP2 regulate body weight, food intake, and GH and blood glucose concentrations, and other physiological phenomena through their interactions with the same receptor, GHSR.
Collapse
Affiliation(s)
- Katsuya Sakai
- Division of Respirology, Rheumatology, Infectious Diseases, and Neurology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yuki Nakazato
- Division of Respirology, Rheumatology, Infectious Diseases, and Neurology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yuki Shiimura
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka 830-0011, Japan
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Weidong Zhang
- Laboratory of Veterinary Physiology, Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
- Laboratory of Biomolecular Analysis, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Masamitsu Nakazato
- Forefront Research Center, Graduate School of Science, Osaka University, Osaka 560-0043, Japan
| |
Collapse
|
4
|
Goel M, Mittal A, Jain VR, Bharadwaj A, Modi S, Ahuja G, Jain A, Kumar K. Integrative Functions of the Hypothalamus: Linking Cognition, Emotion and Physiology for Well-being and Adaptability. Ann Neurosci 2025; 32:128-142. [PMID: 39544638 PMCID: PMC11559822 DOI: 10.1177/09727531241255492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 03/04/2024] [Indexed: 11/17/2024] Open
Abstract
Background The hypothalamus, a small yet crucial neuroanatomical structure, integrates external (e.g., environmental) and internal (e.g., physiological/hormonal) stimuli. This integration governs various physiological processes and influences cognitive, emotional, and behavioral outcomes. It serves as a functional bridge between the nervous and endocrine systems, maintaining homeostasis and coordinating bodily functions. Summary Recent advancements in the neurobiology of the hypothalamus have elucidated its functional map, establishing a causal relationship between its responses-such as respiration, sleep, and stress-and various physiological processes. The hypothalamus facilitates and coordinates these complex processes by processing diverse stimuli, enabling the body to maintain internal balance and respond effectively to external demands. This review delves into the hypothalamus's intricate connections with cognition, emotion, and physiology, exploring how these interactions promote overall well-being and adaptability. Key Message Targeted external stimuli can modulate hypothalamic neuronal activities, impacting the physiological, cognitive, and emotional landscape. The review highlights non-invasive techniques, such as controlled breathing exercises, optimized sleep architecture, and stress management, as potential methods to enhance hypothalamic function. Ultimately, this comprehensive review underscores the multifaceted role of the hypothalamus in integrating signals, maintaining homeostasis, and influencing cognition, emotion, and physiology.
Collapse
Affiliation(s)
- Mansi Goel
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi (IIITD, New Delhi, India
| | - Aayushi Mittal
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi (IIITD, New Delhi, India
| | - Vijaya Raje Jain
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi (IIITD, New Delhi, India
| | | | - Shivani Modi
- Ceekr Concepts Private Limited, New Delhi, India
| | - Gaurav Ahuja
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi (IIITD, New Delhi, India
| | - Ankur Jain
- Ceekr Concepts Private Limited, New Delhi, India
| | | |
Collapse
|
5
|
Rajan PK, Udoh UAS, Finley R, Pierre SV, Sanabria J. The Biological Clock of Liver Metabolism in Metabolic Dysfunction-Associated Steatohepatitis Progression to Hepatocellular Carcinoma. Biomedicines 2024; 12:1961. [PMID: 39335475 PMCID: PMC11428469 DOI: 10.3390/biomedicines12091961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024] Open
Abstract
Circadian rhythms are endogenous behavioral or physiological cycles that are driven by a daily biological clock that persists in the absence of geophysical or environmental temporal cues. Circadian rhythm-related genes code for clock proteins that rise and fall in rhythmic patterns driving biochemical signals of biological processes from metabolism to physiology and behavior. Clock proteins have a pivotal role in liver metabolism and homeostasis, and their disturbances are implicated in various liver disease processes. Encoded genes play critical roles in the initiation and progression of metabolic dysfunction-associated steatohepatitis (MASH) to hepatocellular carcinoma (HCC) and their proteins may become diagnostic markers as well as therapeutic targets. Understanding molecular and metabolic mechanisms underlying circadian rhythms will aid in therapeutic interventions and may have broader clinical applications. The present review provides an overview of the role of the liver's circadian rhythm in metabolic processes in health and disease, emphasizing MASH progression and the oncogenic associations that lead to HCC.
Collapse
Affiliation(s)
- Pradeep Kumar Rajan
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Utibe-Abasi S Udoh
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Robert Finley
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
| | - Juan Sanabria
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
- Department of Nutrition and Metabolomic Core Facility, School of Medicine, Case Western Reserve University, Cleveland, OH 44100, USA
| |
Collapse
|
6
|
Speksnijder EM, Bisschop PH, Siegelaar SE, Stenvers DJ, Kalsbeek A. Circadian desynchrony and glucose metabolism. J Pineal Res 2024; 76:e12956. [PMID: 38695262 DOI: 10.1111/jpi.12956] [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: 12/19/2023] [Revised: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 05/09/2024]
Abstract
The circadian timing system controls glucose metabolism in a time-of-day dependent manner. In mammals, the circadian timing system consists of the main central clock in the bilateral suprachiasmatic nucleus (SCN) of the anterior hypothalamus and subordinate clocks in peripheral tissues. The oscillations produced by these different clocks with a period of approximately 24-h are generated by the transcriptional-translational feedback loops of a set of core clock genes. Glucose homeostasis is one of the daily rhythms controlled by this circadian timing system. The central pacemaker in the SCN controls glucose homeostasis through its neural projections to hypothalamic hubs that are in control of feeding behavior and energy metabolism. Using hormones such as adrenal glucocorticoids and melatonin and the autonomic nervous system, the SCN modulates critical processes such as glucose production and insulin sensitivity. Peripheral clocks in tissues, such as the liver, muscle, and adipose tissue serve to enhance and sustain these SCN signals. In the optimal situation all these clocks are synchronized and aligned with behavior and the environmental light/dark cycle. A negative impact on glucose metabolism becomes apparent when the internal timing system becomes disturbed, also known as circadian desynchrony or circadian misalignment. Circadian desynchrony may occur at several levels, as the mistiming of light exposure or sleep will especially affect the central clock, whereas mistiming of food intake or physical activity will especially involve the peripheral clocks. In this review, we will summarize the literature investigating the impact of circadian desynchrony on glucose metabolism and how it may result in the development of insulin resistance. In addition, we will discuss potential strategies aimed at reinstating circadian synchrony to improve insulin sensitivity and contribute to the prevention of type 2 diabetes.
Collapse
Affiliation(s)
- Esther M Speksnijder
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Peter H Bisschop
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Sarah E Siegelaar
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Dirk Jan Stenvers
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
7
|
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
|
8
|
Hurtado-Alvarado G, Soto-Tinoco E, Santacruz-Martínez E, Prager-Khoutorsky M, Escobar C, Buijs RM. Suprachiasmatic nucleus promotes hyperglycemia induced by sleep delay. Curr Biol 2023; 33:4343-4352.e4. [PMID: 37725978 DOI: 10.1016/j.cub.2023.08.071] [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: 06/15/2023] [Revised: 08/05/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023]
Abstract
Short sleep is linked to disturbances in glucose metabolism and may induce a prediabetic condition. The biological clock in the suprachiasmatic nucleus (SCN) regulates the glucose rhythm in the circulation and the sleep-wake cycle. SCN vasopressin neurons (SCNVP) control daily glycemia by regulating the entrance of glucose into the arcuate nucleus (ARC). Thus, we hypothesized that sleep delay may influence SCN neuronal activity. We, therefore, investigated the role of SCNVP when sleep is disrupted by forced locomotor activity. After 2 h of sleep delay, rats exhibited decreased SCNVP neuronal activity, a decrease in the glucose transporter GLUT1 expression in tanycytes lining the third ventricle, lowered glucose entrance into the ARC, and developed hyperglycemia. The association between reduced SCNVP neuronal activity and hyperglycemia in sleep-delayed rats was evidenced by injecting intracerebroventricular vasopressin; this increased GLUT1 immunoreactivity in tanycytes, thus promoting normoglycemia. Following sleep recovery, glucose levels decreased, whereas SCNVP neuronal activity increased. These results imply that sleep-delay-induced changes in SCNVP activity lead to glycemic impairment, inferring that disruption of biological clock function might represent a critical step in developing type 2 diabetes.
Collapse
Affiliation(s)
- Gabriela Hurtado-Alvarado
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, 04510 Mexico City, Mexico
| | - Eva Soto-Tinoco
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, 04510 Mexico City, Mexico
| | - Esteban Santacruz-Martínez
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, 04510 Mexico City, Mexico
| | - Masha Prager-Khoutorsky
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, 3655 Promenade Sir-William-Osler, Montréal, QC H3G 1Y6, Canada
| | - Carolina Escobar
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico
| | - Ruud M Buijs
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, 04510 Mexico City, Mexico.
| |
Collapse
|
9
|
de Leeuw M, Verhoeve SI, van der Wee NJA, van Hemert AM, Vreugdenhil E, Coomans CP. The role of the circadian system in the etiology of depression. Neurosci Biobehav Rev 2023; 153:105383. [PMID: 37678570 DOI: 10.1016/j.neubiorev.2023.105383] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/19/2023] [Accepted: 09/02/2023] [Indexed: 09/09/2023]
Abstract
Circadian rhythms have evolved in almost all organisms enabling them to anticipate alternating changes in the environment. As a consequence, the circadian clock controls a broad range of bodily functions including appetite, sleep, activity and cortisol levels. The circadian clock synchronizes itself to the external world mainly by environmental light cues and can be disturbed by a variety of factors, including shift-work, jet-lag, stress, ageing and artificial light at night. Interestingly, mood has also been shown to follow a diurnal rhythm. Moreover, circadian disruption has been associated with various mood disorders and patients suffering from depression have irregular biological rhythms in sleep, appetite, activity and cortisol levels suggesting that circadian rhythmicity is crucially involved in the etiology and pathophysiology of depression. The aim of the present review is to give an overview and discuss recent findings in both humans and rodents linking a disturbed circadian rhythm to depression. Understanding the relation between a disturbed circadian rhythm and the etiology of depression may lead to novel therapeutic and preventative strategies.
Collapse
Affiliation(s)
- Max de Leeuw
- Department of Psychiatry, Leiden University Medical Center, Postal Zone B1-P, P.O. Box 9600, Leiden 2300 RC, the Netherlands; Mental Health Care Rivierduinen, Bipolar Disorder Outpatient Clinic, PO Box 405, Leiden 2300 AK, the Netherlands.
| | - Sanne I Verhoeve
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, Leiden 2300 RC, the Netherlands
| | - Nic J A van der Wee
- Department of Psychiatry, Leiden University Medical Center, Postal Zone B1-P, P.O. Box 9600, Leiden 2300 RC, the Netherlands
| | - Albert M van Hemert
- Department of Psychiatry, Leiden University Medical Center, Postal Zone B1-P, P.O. Box 9600, Leiden 2300 RC, the Netherlands
| | - Erno Vreugdenhil
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, Leiden 2300 RC, the Netherlands
| | - Claudia P Coomans
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, Leiden 2300 RC, the Netherlands
| |
Collapse
|
10
|
Cincotta AH. Brain Dopamine-Clock Interactions Regulate Cardiometabolic Physiology: Mechanisms of the Observed Cardioprotective Effects of Circadian-Timed Bromocriptine-QR Therapy in Type 2 Diabetes Subjects. Int J Mol Sci 2023; 24:13255. [PMID: 37686060 PMCID: PMC10487918 DOI: 10.3390/ijms241713255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 09/10/2023] Open
Abstract
Despite enormous global efforts within clinical research and medical practice to reduce cardiovascular disease(s) (CVD), it still remains the leading cause of death worldwide. While genetic factors clearly contribute to CVD etiology, the preponderance of epidemiological data indicate that a major common denominator among diverse ethnic populations from around the world contributing to CVD is the composite of Western lifestyle cofactors, particularly Western diets (high saturated fat/simple sugar [particularly high fructose and sucrose and to a lesser extent glucose] diets), psychosocial stress, depression, and altered sleep/wake architecture. Such Western lifestyle cofactors are potent drivers for the increased risk of metabolic syndrome and its attendant downstream CVD. The central nervous system (CNS) evolved to respond to and anticipate changes in the external (and internal) environment to adapt survival mechanisms to perceived stresses (challenges to normal biological function), including the aforementioned Western lifestyle cofactors. Within the CNS of vertebrates in the wild, the biological clock circuitry surveils the environment and has evolved mechanisms for the induction of the obese, insulin-resistant state as a survival mechanism against an anticipated ensuing season of low/no food availability. The peripheral tissues utilize fat as an energy source under muscle insulin resistance, while increased hepatic insulin resistance more readily supplies glucose to the brain. This neural clock function also orchestrates the reversal of the obese, insulin-resistant condition when the low food availability season ends. The circadian neural network that produces these seasonal shifts in metabolism is also responsive to Western lifestyle stressors that drive the CNS clock into survival mode. A major component of this natural or Western lifestyle stressor-induced CNS clock neurophysiological shift potentiating the obese, insulin-resistant state is a diminution of the circadian peak of dopaminergic input activity to the pacemaker clock center, suprachiasmatic nucleus. Pharmacologically preventing this loss of circadian peak dopaminergic activity both prevents and reverses existing metabolic syndrome in a wide variety of animal models of the disorder, including high fat-fed animals. Clinically, across a variety of different study designs, circadian-timed bromocriptine-QR (quick release) (a unique formulation of micronized bromocriptine-a dopamine D2 receptor agonist) therapy of type 2 diabetes subjects improved hyperglycemia, hyperlipidemia, hypertension, immune sterile inflammation, and/or adverse cardiovascular event rate. The present review details the seminal circadian science investigations delineating important roles for CNS circadian peak dopaminergic activity in the regulation of peripheral fuel metabolism and cardiovascular biology and also summarizes the clinical study findings of bromocriptine-QR therapy on cardiometabolic outcomes in type 2 diabetes subjects.
Collapse
|
11
|
Jones AA, Arble DM. In light of breathing: environmental light is an important modulator of breathing with clinical implications. Front Neurosci 2023; 17:1217799. [PMID: 37521684 PMCID: PMC10373889 DOI: 10.3389/fnins.2023.1217799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
In vertebrate animals, the automatic, rhythmic pattern of breathing is a highly regulated process that can be modulated by various behavioral and physiological factors such as metabolism, sleep-wake state, activity level, and endocrine signaling. Environmental light influences many of these modulating factors both indirectly by organizing daily and seasonal rhythms of behavior and directly through acute changes in neural signaling. While several observations from rodent and human studies suggest that environmental light affects breathing, few have systematically evaluated the underlying mechanisms and clinical relevance of environmental light on the regulation of respiratory behavior. Here, we provide new evidence and discuss the potential neurobiological mechanisms by which light modulates breathing. We conclude that environmental light should be considered, from bench to bedside, as a clinically relevant modulator of respiratory health and disease.
Collapse
|
12
|
Ribble A, Hellmann J, Conklin DJ, Bhatnagar A, Haberzettl P. Fine particulate matter (PM 2.5)-induced pulmonary oxidative stress contributes to increases in glucose intolerance and insulin resistance in a mouse model of circadian dyssynchrony. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 877:162934. [PMID: 36934930 PMCID: PMC10164116 DOI: 10.1016/j.scitotenv.2023.162934] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/28/2023] [Accepted: 03/14/2023] [Indexed: 05/06/2023]
Abstract
Results of human and animal studies independently suggest that either ambient fine particulate matter (PM2.5) air pollution exposure or a disturbed circadian rhythm (circadian dyssynchrony) are important contributing factors to the rapidly evolving type-2-diabetes (T2D) epidemic. The objective of this study is to investigate whether circadian dyssynchrony increases the susceptibility to PM2.5 and how PM2.5 affects metabolic health in circadian dyssynchrony. We examined systemic and organ-specific changes in glucose homeostasis and insulin sensitivity in mice maintained on a regular (12/12 h light/dark) or disrupted (18/6 h light/dark, light-induced circadian dyssynchrony, LICD) light cycle exposed to air or concentrated PM2.5 (CAP, 6 h/day, 30 days). Exposures during Zeitgeber ZT3-9 or ZT11-17 (Zeitgeber in circadian time, ZT0 = begin of light cycle) tested for time-of-day PM2.5 sensitivity (chronotoxicity). Mice transgenic for lung-specific overexpression of extracellular superoxide dismutase (ecSOD-Tg) were used to assess the contribution of CAP-induced pulmonary oxidative stress. Both, CAP exposure from ZT3-9 or ZT11-17, decreased glucose tolerance and insulin sensitivity in male mice with LICD, but not in female mice or in mice kept on a regular light cycle. Although changes in glucose homeostasis in CAP-exposed male mice with LICD were not associated with obesity, they were accompanied by white adipose tissue (WAT) inflammation, impaired insulin signaling in skeletal muscle and liver, and systemic and pulmonary oxidative stress. Preventing CAP-induced oxidative stress in the lungs mitigated the CAP-induced decrease in glucose tolerance and insulin sensitivity in LICD. Our results demonstrate that circadian dyssynchrony is a novel susceptibility state for PM2.5 and suggest that PM2.5 by inducing pulmonary oxidative stress increases glucose intolerance and insulin resistance in circadian dyssynchrony.
Collapse
Affiliation(s)
- Amanda Ribble
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Jason Hellmann
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Daniel J Conklin
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Aruni Bhatnagar
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Petra Haberzettl
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
13
|
Mendoza-Viveros L, Marmolejo-Gutierrez C, Cid-Castro C, Escalante-Covarrubias Q, Montellier E, Carreño-Vázquez E, Noriega LG, Velázquez-Villegas LA, Tovar AR, Sassone-Corsi P, Aguilar-Arnal L, Orozco-Solis R. Astrocytic circadian clock control of energy expenditure by transcriptional stress responses in the ventromedial hypothalamus. Glia 2023; 71:1626-1647. [PMID: 36919670 DOI: 10.1002/glia.24360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/28/2023] [Accepted: 03/04/2023] [Indexed: 03/16/2023]
Abstract
Hypothalamic circuits compute systemic information to control metabolism. Astrocytes residing within the hypothalamus directly sense nutrients and hormones, integrating metabolic information, and modulating neuronal responses. Nevertheless, the role of the astrocytic circadian clock on the control of energy balance remains unclear. We used mice with a targeted ablation of the core-clock gene Bmal1 within Gfap-expressing astrocytes to gain insight on the role played by this transcription factor in astrocytes. While this mutation does not substantially affect the phenotype in mice fed normo-caloric diet, under high-fat diet we unmasked a thermogenic phenotype consisting of increased energy expenditure, and catabolism in brown adipose and overall metabolic improvement consisting of better glycemia control, and body composition. Transcriptomic analysis in the ventromedial hypothalamus revealed an enhanced response to moderate cellular stress, including ER-stress response, unfolded protein response and autophagy. We identified Xbp1 and Atf1 as two key transcription factors enhancing cellular stress responses. Therefore, we unveiled a previously unknown role of the astrocytic circadian clock modulating energy balance through the regulation of cellular stress responses within the VMH.
Collapse
Affiliation(s)
- Lucia Mendoza-Viveros
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, Mexico
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México UNAM, México City, Mexico
- Centro de Investigación sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE-CINVESTAV), México City, México
| | | | - Carolina Cid-Castro
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, Mexico
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México UNAM, México City, Mexico
- Centro de Investigación sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE-CINVESTAV), México City, México
| | | | | | | | - Lilia G Noriega
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Armando R Tovar
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Lorena Aguilar-Arnal
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México UNAM, México City, Mexico
| | - Ricardo Orozco-Solis
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, Mexico
- Centro de Investigación sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE-CINVESTAV), México City, México
| |
Collapse
|
14
|
Tiwari A, Rathor P, Trivedi PK, Ch R. Multi-Omics Reveal Interplay between Circadian Dysfunction and Type2 Diabetes. BIOLOGY 2023; 12:301. [PMID: 36829576 PMCID: PMC9953493 DOI: 10.3390/biology12020301] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023]
Abstract
Type 2 diabetes is one of the leading threats to human health in the 21st century. It is a metabolic disorder characterized by a dysregulated glucose metabolism resulting from impaired insulin secretion or insulin resistance. More recently, accumulated epidemiological and animal model studies have confirmed that circadian dysfunction caused by shift work, late meal timing, and sleep loss leads to type 2 diabetes. Circadian rhythms, 24-h endogenous biological oscillations, are a fundamental feature of nearly all organisms and control many physiological and cellular functions. In mammals, light synchronizes brain clocks and feeding is a main stimulus that synchronizes the peripheral clocks in metabolic tissues, such as liver, pancreas, muscles, and adipose tissues. Circadian arrhythmia causes the loss of synchrony of the clocks of these metabolic tissues and leads to an impaired pancreas β-cell metabolism coupled with altered insulin secretion. In addition to these, gut microbes and circadian rhythms are intertwined via metabolic regulation. Omics approaches play a significant role in unraveling how a disrupted circadian metabolism causes type 2 diabetes. In the present review, we emphasize the discoveries of several genes, proteins, and metabolites that contribute to the emergence of type 2 diabetes mellitus (T2D). The implications of these discoveries for comprehending the circadian clock network in T2D may lead to new therapeutic solutions.
Collapse
Affiliation(s)
- Ashutosh Tiwari
- Metabolomics Lab, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
| | - Priya Rathor
- Metabolomics Lab, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
| | - Prabodh Kumar Trivedi
- Department of Biotechnology, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
- Academy of Council of Scientific and Industrial Research (ACSIR), Gaziabad 201002, India
| | - Ratnasekhar Ch
- Metabolomics Lab, CSIR-Central Institute of Medicinal & Aromatic Plants (CIMAP), Lucknow 226015, India
- Academy of Council of Scientific and Industrial Research (ACSIR), Gaziabad 201002, India
- School of Biological Sciences, Queen’s University Belfast, Belfast BT9 5DL, UK
| |
Collapse
|
15
|
Meng JJ, Shen JW, Li G, Ouyang CJ, Hu JX, Li ZS, Zhao H, Shi YM, Zhang M, Liu R, Chen JT, Ma YQ, Zhao H, Xue T. Light modulates glucose metabolism by a retina-hypothalamus-brown adipose tissue axis. Cell 2023; 186:398-412.e17. [PMID: 36669474 DOI: 10.1016/j.cell.2022.12.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/22/2022] [Accepted: 12/13/2022] [Indexed: 01/20/2023]
Abstract
Public health studies indicate that artificial light is a high-risk factor for metabolic disorders. However, the neural mechanism underlying metabolic modulation by light remains elusive. Here, we found that light can acutely decrease glucose tolerance (GT) in mice by activation of intrinsically photosensitive retinal ganglion cells (ipRGCs) innervating the hypothalamic supraoptic nucleus (SON). Vasopressin neurons in the SON project to the paraventricular nucleus, then to the GABAergic neurons in the solitary tract nucleus, and eventually to brown adipose tissue (BAT). Light activation of this neural circuit directly blocks adaptive thermogenesis in BAT, thereby decreasing GT. In humans, light also modulates GT at the temperature where BAT is active. Thus, our work unveils a retina-SON-BAT axis that mediates the effect of light on glucose metabolism, which may explain the connection between artificial light and metabolic dysregulation, suggesting a potential prevention and treatment strategy for managing glucose metabolic disorders.
Collapse
Affiliation(s)
- Jian-Jun Meng
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Jia-Wei Shen
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Guang Li
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Chang-Jie Ouyang
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Jia-Xi Hu
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Zi-Shuo Li
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Hang Zhao
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yi-Ming Shi
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Mei Zhang
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Rong Liu
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Ju-Tao Chen
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yu-Qian Ma
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Huan Zhao
- College of Biology, Food and Environment, Hefei University, Hefei 230601, China
| | - Tian Xue
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
16
|
Trebucq LL, Lamberti ML, Rota R, Aiello I, Borio C, Bilen M, Golombek DA, Plano SA, Chiesa JJ. Chronic circadian desynchronization of feeding-fasting rhythm generates alterations in daily glycemia, LDL cholesterolemia and microbiota composition in mice. Front Nutr 2023; 10:1154647. [PMID: 37125029 PMCID: PMC10145162 DOI: 10.3389/fnut.2023.1154647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/15/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction The circadian system synchronizes behavior and physiology to the 24-h light- dark (LD) cycle. Timing of food intake and fasting periods provide strong signals for peripheral circadian clocks regulating nutrient assimilation, glucose, and lipid metabolism. Mice under 12 h light:12 h dark (LD) cycles exhibit behavioral activity and feeding during the dark period, while fasting occurs at rest during light. Disruption of energy metabolism, leading to an increase in body mass, was reported in experimental models of circadian desynchronization. In this work, the effects of chronic advances of the LD cycles (chronic jet-lag protocol, CJL) were studied on the daily homeostasis of energy metabolism and weight gain. Methods Male C57 mice were subjected to a CJL or LD schedule, measuring IPGTT, insulinemia, microbiome composition and lipidemia. Results Mice under CJL show behavioral desynchronization and feeding activity distributed similarly at the light and dark hours and, although feeding a similar daily amount of food as compared to controls, show an increase in weight gain. In addition, ad libitum glycemia rhythm was abolished in CJL-subjected mice, showing similar blood glucose values at light and dark. CJL also generated glucose intolerance at dark in an intraperitoneal glucose tolerance test (IPGTT), with increased insulin release at both light and dark periods. Low-density lipoprotein (LDL) cholesterolemia was increased under this condition, but no changes in HDL cholesterolemia were observed. Firmicutes/Bacteroidetes ratio was analyzed as a marker of circadian disruption of microbiota composition, showing opposite phases at the light and dark when comparing LD vs. CJL. Discussion Chronic misalignment of feeding/fasting rhythm leads to metabolic disturbances generating nocturnal hyperglycemia, glucose intolerance and hyperinsulinemia in a IPGTT, increased LDL cholesterolemia, and increased weight gain, underscoring the importance of the timing of food consumption with respect to the circadian system for metabolic health.
Collapse
Affiliation(s)
- Laura Lucía Trebucq
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes (UNQ), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bernal, Argentina
| | - Melisa Luciana Lamberti
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes (UNQ), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bernal, Argentina
| | - Rosana Rota
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes (UNQ), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bernal, Argentina
| | - Ignacio Aiello
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes (UNQ), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bernal, Argentina
| | - Cristina Borio
- Laboratorio de Ingeniería Genética, Biología Celular y Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes (UNQ), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bernal, Argentina
| | - Marcos Bilen
- Laboratorio de Ingeniería Genética, Biología Celular y Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes (UNQ), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bernal, Argentina
| | - Diego Andrés Golombek
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes (UNQ), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bernal, Argentina
- Escuela de Educacion, Universidad de San Andrés, Victoria, Argentina
| | - Santiago Andrés Plano
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes (UNQ), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bernal, Argentina
- Institute for Biomedical Research (BIOMED), Catholic University of Argentina (UCA), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- *Correspondence: Santiago Andrés Plano,
| | - Juan José Chiesa
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes (UNQ), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bernal, Argentina
- Juan José Chiesa,
| |
Collapse
|
17
|
Olejniczak I, Campbell B, Tsai YC, Tyagarajan SK, Albrecht U, Ripperger JA. Suprachiasmatic to paraventricular nuclei interaction generates normal food searching rhythms in mice. Front Physiol 2022; 13:909795. [PMID: 36277219 PMCID: PMC9582613 DOI: 10.3389/fphys.2022.909795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 09/23/2022] [Indexed: 11/29/2022] Open
Abstract
Searching for food follows a well-organized decision process in mammals to take up food only if necessary. Moreover, scavenging is preferred during their activity phase. Various time-dependent regulatory processes have been identified originating from the suprachiasmatic nuclei (SCN), which convert external light information into synchronizing output signals. However, a direct impact of the SCN on the timing of normal food searching has not yet been found. Here, we revisited the function of the SCN to affect when mice look for food. We found that this process was independent of light but modified by the palatability of the food source. Surprisingly, reducing the output from the SCN, in particular from the vasopressin releasing neurons, reduced the amount of scavenging during the early activity phase. The SCN appeared to transmit a signal to the paraventricular nuclei (PVN) via GABA receptor A1. Finally, the interaction of SCN and PVN was verified by retrograde transport-mediated complementation. None of the genetic manipulations affected the uptake of more palatable food. The data indicate that the PVN are sufficient to produce blunted food searching rhythms and are responsive to hedonistic feeding. Nevertheless, the search for normal food during the early activity phase is significantly enhanced by the SCN.
Collapse
Affiliation(s)
- Iwona Olejniczak
- Department of Biology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Benjamin Campbell
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Yuan-Chen Tsai
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Shiva K. Tyagarajan
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Urs Albrecht
- Department of Biology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Jürgen A. Ripperger
- Department of Biology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- *Correspondence: Jürgen A. Ripperger,
| |
Collapse
|
18
|
Vujović N, Piron MJ, Qian J, Chellappa SL, Nedeltcheva A, Barr D, Heng SW, Kerlin K, Srivastav S, Wang W, Shoji B, Garaulet M, Brady MJ, Scheer FAJL. Late isocaloric eating increases hunger, decreases energy expenditure, and modifies metabolic pathways in adults with overweight and obesity. Cell Metab 2022; 34:1486-1498.e7. [PMID: 36198293 PMCID: PMC10184753 DOI: 10.1016/j.cmet.2022.09.007] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 07/28/2022] [Accepted: 09/12/2022] [Indexed: 01/10/2023]
Abstract
Late eating has been linked to obesity risk. It is unclear whether this is caused by changes in hunger and appetite, energy expenditure, or both, and whether molecular pathways in adipose tissues are involved. Therefore, we conducted a randomized, controlled, crossover trial (ClinicalTrials.gov NCT02298790) to determine the effects of late versus early eating while rigorously controlling for nutrient intake, physical activity, sleep, and light exposure. Late eating increased hunger (p < 0.0001) and altered appetite-regulating hormones, increasing waketime and 24-h ghrelin:leptin ratio (p < 0.0001 and p = 0.006, respectively). Furthermore, late eating decreased waketime energy expenditure (p = 0.002) and 24-h core body temperature (p = 0.019). Adipose tissue gene expression analyses showed that late eating altered pathways involved in lipid metabolism, e.g., p38 MAPK signaling, TGF-β signaling, modulation of receptor tyrosine kinases, and autophagy, in a direction consistent with decreased lipolysis/increased adipogenesis. These findings show converging mechanisms by which late eating may result in positive energy balance and increased obesity risk.
Collapse
Affiliation(s)
- Nina Vujović
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Avenue, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA.
| | - Matthew J Piron
- Department of Medicine, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, The University of Chicago, Chicago, IL, USA
| | - Jingyi Qian
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Avenue, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah L Chellappa
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Avenue, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Arlet Nedeltcheva
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Avenue, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - David Barr
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Avenue, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Su Wei Heng
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Avenue, Boston, MA 02115, USA
| | - Kayla Kerlin
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Avenue, Boston, MA 02115, USA
| | - Suhina Srivastav
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Avenue, Boston, MA 02115, USA
| | - Wei Wang
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Avenue, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Brent Shoji
- Department of Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Marta Garaulet
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Avenue, Boston, MA 02115, USA; Department of Physiology, Regional Campus of International Excellence, University of Murcia, 30100 Murcia, Spain; Biomedical Research Institute of Murcia, IMIB-Arrixaca-UMU, University Clinical Hospital, 30120 Murcia, Spain
| | - Matthew J Brady
- Department of Medicine, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, The University of Chicago, Chicago, IL, USA
| | - Frank A J L Scheer
- Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Avenue, Boston, MA 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
19
|
LaPierre MP, Lawler K, Godbersen S, Farooqi IS, Stoffel M. MicroRNA-7 regulates melanocortin circuits involved in mammalian energy homeostasis. Nat Commun 2022; 13:5733. [PMID: 36175420 PMCID: PMC9522793 DOI: 10.1038/s41467-022-33367-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 09/14/2022] [Indexed: 11/09/2022] Open
Abstract
MicroRNAs (miRNAs) modulate physiological responses by repressing the expression of gene networks. We found that global deletion of microRNA-7 (miR-7), the most enriched miRNA in the hypothalamus, causes obesity in mice. Targeted deletion of miR-7 in Single-minded homolog 1 (Sim1) neurons, a critical component of the hypothalamic melanocortin pathway, causes hyperphagia, obesity and increased linear growth, mirroring Sim1 and Melanocortin-4 receptor (MC4R) haplo-insufficiency in mice and humans. We identified Snca (α-Synuclein) and Igsf8 (Immunoglobulin Superfamily Member 8) as miR-7 target genes that act in Sim1 neurons to regulate body weight and endocrine axes. In humans, MIR-7-1 is located in the last intron of HNRNPK, whose promoter drives the expression of both genes. Genetic variants at the HNRNPK locus that reduce its expression are associated with increased height and truncal fat mass. These findings demonstrate that miR-7 suppresses gene networks involved in the hypothalamic melanocortin pathway to regulate mammalian energy homeostasis.
Collapse
Affiliation(s)
- Mary P LaPierre
- Institute of Molecular Health Sciences, ETH Zürich, 8093, Zürich, Switzerland
| | - Katherine Lawler
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Svenja Godbersen
- Institute of Molecular Health Sciences, ETH Zürich, 8093, Zürich, Switzerland
| | - I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Markus Stoffel
- Institute of Molecular Health Sciences, ETH Zürich, 8093, Zürich, Switzerland.
- Medical Faculty, University of Zürich, 8091, Zürich, Switzerland.
| |
Collapse
|
20
|
Mu YM, Huang XD, Zhu S, Hu ZF, So KF, Ren CR, Tao Q. Alerting effects of light in healthy individuals: a systematic review and meta-analysis. Neural Regen Res 2022; 17:1929-1936. [PMID: 35142669 PMCID: PMC8848614 DOI: 10.4103/1673-5374.335141] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Light plays an essential role in psychobiological and psychophysiological processes, such as alertness. The alerting effect is influenced by light characteristics and the timing of interventions. This meta-analysis is the first to systematically review the effect of light intervention on alertness and to discuss the optimal protocol for light intervention. In this meta-analysis, registered at PROSPERO (Registration ID: CRD42020181485), we conducted a systematic search of the Web of Science, PubMed, and PsycINFO databases for studies published in English prior to August 2021. The outcomes included both subjective and objective alertness. Subgroup analyses considered a variety of factors, such as wavelength, correlated color temperature (CCT), light illuminance, and timing of interventions (daytime, night-time, or all day). Twenty-seven crossover studies and two parallel-group studies were included in this meta-analysis, with a total of 1210 healthy participants (636 (52%) male, mean age 25.62 years). The results revealed that light intervention had a positive effect on both subjective alertness (standardized mean difference (SMD) = –0.28, 95% confidence interval (CI): –0.49 to –0.06, P = 0.01) and objective alertness in healthy subjects (SMD = –0.34, 95% CI: –0.68 to –0.01, P = 0.04). The subgroup analysis revealed that cold light was better than warm light in improving subjective alertness (SMD = –0.37, 95% CI: –0.65 to –0.10, P = 0.007, I2 = 26%) and objective alertness (SMD = –0.36, 95% CI: –0.66 to –0.07, P = 0.02, I2 = 0). Both daytime (SMD = –0.22, 95% CI: –0.37 to –0.07, P = 0.005, I2 = 74%) and night-time (SMD = –0.32, 95% CI: –0.61 to –0.02, P = 0.04, I2 = 0) light exposure improved subjective alertness. The results of this meta-analysis and systematic review indicate that light exposure is associated with significant improvement in subjective and objective alertness. In addition, light exposure with a higher CCT was more effective in improving alertness than light exposure with a lower CCT. Our results also suggest that both daytime and night-time light exposure can improve subjective alertness.
Collapse
Affiliation(s)
- Yi-Man Mu
- Department of Public Health and Preventive Medicine, School of Basic Medicine; Division of Medical Psychology and Behavior Science, School of Basic Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Xiao-Dan Huang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Sui Zhu
- Department of Public Health and Preventive Medicine, School of Basic Medicine; Division of Medical Psychology and Behavior Science, School of Basic Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Zheng-Fang Hu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University; Guangzhou Regenerative Medicine and Health Guangdong Laboratory; Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, Guangdong Province; Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Science, Qingdao, Shandong Province; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Chao-Ran Ren
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University; Guangzhou Regenerative Medicine and Health Guangdong Laboratory; Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, Guangdong Province; Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Science, Qingdao, Shandong Province; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Qian Tao
- Department of Public Health and Preventive Medicine, School of Basic Medicine; Division of Medical Psychology and Behavior Science, School of Basic Medicine, Jinan University; Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, Guangdong Province; Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Science, Qingdao, Shandong Province, China
| |
Collapse
|
21
|
Circadian clock, diurnal glucose metabolic rhythm, and dawn phenomenon. Trends Neurosci 2022; 45:471-482. [PMID: 35466006 PMCID: PMC9117496 DOI: 10.1016/j.tins.2022.03.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/15/2022] [Accepted: 03/26/2022] [Indexed: 01/28/2023]
Abstract
The circadian clock provides cue-independent anticipatory signals for diurnal rhythms of baseline glucose levels and glucose tolerance. The central circadian clock is located in the hypothalamic suprachiasmatic nucleus (SCN), which comprises primarily GABAergic neurons. The SCN clock regulates physiological diurnal rhythms of endogenous glucose production (EGP) and hepatic insulin sensitivity through neurohumoral mechanisms. Disruption of the molecular circadian clock is associated with the extended dawn phenomenon (DP) in type 2 diabetes (T2D), referring to hyperglycemia in the early morning without nocturnal hypoglycemia. The DP affects nearly half of patients with diabetes, with poorly defined etiology and a lack of targeted therapy. Here we review neural and secreted factors in physiological diurnal rhythms of glucose metabolism and their pathological implications for the DP.
Collapse
|
22
|
Liu JA, Meléndez-Fernández OH, Bumgarner JR, Nelson RJ. Effects of light pollution on photoperiod-driven seasonality. Horm Behav 2022; 141:105150. [PMID: 35304351 PMCID: PMC10137835 DOI: 10.1016/j.yhbeh.2022.105150] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 12/23/2022]
Abstract
Changes to photoperiod (day length) occur in anticipation of seasonal environmental changes, altering physiology and behavior to maximize fitness. In order for photoperiod to be useful as a predictive factor of temperature or food availability, day and night must be distinct. The increasing prevalence of exposure to artificial light at night (ALAN) in both field and laboratory settings disrupts photoperiodic time measurement and may block development of appropriate seasonal adaptations. Here, we review the effects of ALAN as a disruptor of photoperiodic time measurement and season-specific adaptations, including reproduction, metabolism, immune function, and thermoregulation.
Collapse
Affiliation(s)
- Jennifer A Liu
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, West Virginia, USA.
| | | | - Jacob R Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, West Virginia, USA
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, West Virginia, USA
| |
Collapse
|
23
|
Abstract
The circadian clock is an evolutionarily highly conserved endogenous timing program that structures physiology and behavior according to the time of day. Disruption of circadian rhythms is associated with many common pathologies. The emerging field of circadian medicine aims to exploit the mechanisms of circadian physiology and clock-disease interaction for clinical diagnosis, treatment, and prevention. In this Essay, we outline the principle approaches of circadian medicine, highlight the development of the field in selected areas, and point out open questions and challenges. Circadian medicine has unambiguous health benefits over standard care but is rarely utilized. It is time for clock biology to become an integrated part of translational research.
Collapse
Affiliation(s)
- Achim Kramer
- Charité –Universitätsmedizin Berlin, Laboratory of Chronobiology, Berlin, Germany
- * E-mail: (AK); (HO)
| | - Tanja Lange
- University of Lübeck, Department of Rheumatology & Clinical Immunology, Center of Brain, Behavior and Metabolism, Lübeck, Germany
| | - Claudia Spies
- Charité –Universitätsmedizin Berlin, Department of Anesthesiology and Intensive Care Medicine, Berlin, Germany
| | - Anna-Marie Finger
- Charité –Universitätsmedizin Berlin, Laboratory of Chronobiology, Berlin, Germany
| | - Daniela Berg
- Christian-Albrechts-University Kiel, Department of Neurology, Kiel, Germany
| | - Henrik Oster
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behavior and Metabolism, Lübeck, Germany
- * E-mail: (AK); (HO)
| |
Collapse
|
24
|
Benazzi S, Gorini S, Feraco A, Caprio M. Ritmi circadiani e variabili metaboliche. L'ENDOCRINOLOGO 2021. [PMCID: PMC8569496 DOI: 10.1007/s40619-021-00983-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
I ritmi circadiani sono influenzati da numerose variabili correlate allo stile di vita, soprattutto in relazione ai ritmi imposti dalla società moderna, e vengono profondamente alterati da diverse condizioni patologiche. La fisiologia circadiana è organizzata in modo complesso e integrato; molti dei fattori che sincronizzano il sistema sono a loro volta influenzati e regolati da diversi assi ormonali. Parallelamente, i disturbi del ritmo circadiano derivano da input non ottimali dei fattori sincronizzanti o da condizioni patologiche, e le conseguenze determinano un impatto significativo in diverse condizioni, quali l’obesità e i disturbi del sonno. Durante l’attuale emergenza COVID-19 sono stati registrati crescenti tassi di alterazioni del sonno, complici la preoccupazione diffusa, un comportamento alimentare alterato e la difficoltà per molti, durante il lockdown, nel mantenere ritmi di vita regolari (Barrea et al. in J Transl Med 18:1–11, 2020). Le misure di intervento che si sono mostrate più promettenti contro la desincronizzazione circadiana sono quelle che agiscono sullo stile di vita, basate sul recupero di un corretto ritmo del sonno, la corretta esposizione alla luce solare, l’idonea distribuzione dei pasti e del timing alimentare e lo svolgimento di un’adeguata attività fisica.
Collapse
|
25
|
Time-Restricted Feeding in Mice Prevents the Disruption of the Peripheral Circadian Clocks and Its Metabolic Impact during Chronic Jetlag. Nutrients 2021; 13:nu13113846. [PMID: 34836101 PMCID: PMC8622682 DOI: 10.3390/nu13113846] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 01/17/2023] Open
Abstract
We used time-restricted feeding (TRF) to investigate whether microbial metabolites and the hunger hormone ghrelin can become the dominant entraining factor during chronic jetlag to prevent disruption of the master and peripheral clocks, in order to promote health. Therefore, hypothalamic clock gene and Agrp/Npy mRNA expression were measured in mice that were either chronically jetlagged and fed ad libitum, jetlagged and fed a TRF diet, or not jetlagged and fed a TRF diet. Fecal short-chain fatty acid (SCFA) concentrations, plasma ghrelin and corticosterone levels, and colonic clock gene mRNA expression were measured. Preventing the disruption of the food intake pattern during chronic jetlag using TRF restored the rhythmicity in hypothalamic clock gene mRNA expression of Reverbα but not of Arntl. TRF countered the changes in plasma ghrelin levels and in hypothalamic Npy mRNA expression induced by chronic jetlag, thereby reestablishing the food intake pattern. Increase in body mass induced by chronic jetlag was prevented. Alterations in diurnal fluctuations in fecal SCFAs during chronic jetlag were prevented thereby re-entraining the rhythmic expression of peripheral clock genes. In conclusion, TRF during chronodisruption re-entrains the rhythms in clock gene expression and signals from the gut that regulate food intake to normalize body homeostasis.
Collapse
|
26
|
Wilcox AG, Bains RS, Williams D, Joynson E, Vizor L, Oliver PL, Maywood ES, Hastings MH, Banks G, Nolan PM. Zfhx3-mediated genetic ablation of the SCN abolishes light entrainable circadian activity while sparing food anticipatory activity. iScience 2021; 24:103142. [PMID: 34632336 PMCID: PMC8487057 DOI: 10.1016/j.isci.2021.103142] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/16/2021] [Accepted: 09/14/2021] [Indexed: 01/20/2023] Open
Abstract
Circadian rhythms persist in almost all organisms and are crucial for maintaining appropriate timing in physiology and behaviour. Here, we describe a mouse mutant where the central mammalian pacemaker, the suprachiasmatic nucleus (SCN), has been genetically ablated by conditional deletion of the transcription factor Zfhx3 in the developing hypothalamus. Mutants were arrhythmic over the light-dark cycle and in constant darkness. Moreover, rhythms of metabolic parameters were ablated in vivo although molecular oscillations in the liver maintained some rhythmicity. Despite disruptions to SCN cell identity and circuitry, mutants could still anticipate food availability, yet other zeitgebers - including social cues from cage-mates - were ineffective in restoring rhythmicity although activity levels in mutants were altered. This work highlights a critical role for Zfhx3 in the development of a functional SCN, while its genetic ablation further defines the contribution of SCN circuitry in orchestrating physiological and behavioral responses to environmental signals. Deletion of Zfhx3 in developing hypothalamus leads to behavioral arrhythmicity SCN cell identity is absent while other retinal targets and visual functions remain Rhythms in metabolic functions are lost while some molecular rhythms in liver persist Conditional mutants can respond to food availability and other environmental cues
Collapse
Affiliation(s)
- Ashleigh G Wilcox
- MRC Harwell Institute, Harwell Science Campus, Oxfordshire OX11 0RD, UK
| | - R Sonia Bains
- MRC Harwell Institute, Harwell Science Campus, Oxfordshire OX11 0RD, UK
| | - Debbie Williams
- MRC Harwell Institute, Harwell Science Campus, Oxfordshire OX11 0RD, UK
| | - Elizabeth Joynson
- MRC Harwell Institute, Harwell Science Campus, Oxfordshire OX11 0RD, UK
| | - Lucie Vizor
- MRC Harwell Institute, Harwell Science Campus, Oxfordshire OX11 0RD, UK
| | - Peter L Oliver
- MRC Harwell Institute, Harwell Science Campus, Oxfordshire OX11 0RD, UK
| | - Elizabeth S Maywood
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Michael H Hastings
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Gareth Banks
- MRC Harwell Institute, Harwell Science Campus, Oxfordshire OX11 0RD, UK
| | - Patrick M Nolan
- MRC Harwell Institute, Harwell Science Campus, Oxfordshire OX11 0RD, UK
| |
Collapse
|
27
|
Guo X, Zheng J, Zhang S, Jiang X, Chen T, Yu J, Wang S, Ma X, Wu C. Advances in Unhealthy Nutrition and Circadian Dysregulation in Pathophysiology of NAFLD. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2021; 2:691828. [PMID: 36994336 PMCID: PMC10012147 DOI: 10.3389/fcdhc.2021.691828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022]
Abstract
Unhealthy diets and lifestyle result in various metabolic conditions including metabolic syndrome and non-alcoholic fatty liver disease (NAFLD). Much evidence indicates that disruption of circadian rhythms contributes to the development and progression of excessive hepatic fat deposition and inflammation, as well as liver fibrosis, a key characteristic of non-steatohepatitis (NASH) or the advanced form of NAFLD. In this review, we emphasize the importance of nutrition as a critical factor in the regulation of circadian clock in the liver. We also focus on the roles of the rhythms of nutrient intake and the composition of diets in the regulation of circadian clocks in the context of controlling hepatic glucose and fat metabolism. We then summarize the effects of unhealthy nutrition and circadian dysregulation on the development of hepatic steatosis and inflammation. A better understanding of how the interplay among nutrition, circadian rhythms, and dysregulated metabolism result in hepatic steatosis and inflammation can help develop improved preventive and/or therapeutic strategies for managing NAFLD.
Collapse
Affiliation(s)
- Xin Guo
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Xin Guo, ; Chaodong Wu,
| | - Juan Zheng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Shixiu Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaofan Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ting Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Jiayu Yu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Shu'e Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaomin Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, TX, United States
- *Correspondence: Xin Guo, ; Chaodong Wu,
| |
Collapse
|
28
|
Luo B, Zhou X, Tang Q, Yin Y, Feng G, Li S, Chen L. Circadian rhythms affect bone reconstruction by regulating bone energy metabolism. J Transl Med 2021; 19:410. [PMID: 34579752 PMCID: PMC8477514 DOI: 10.1186/s12967-021-03068-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/02/2021] [Indexed: 01/02/2023] Open
Abstract
Metabolism is one of the most complex cellular biochemical reactions, providing energy and substances for basic activities such as cell growth and proliferation. Early studies have shown that glucose is an important nutrient in osteoblasts. In addition, amino acid metabolism and fat metabolism also play important roles in bone reconstruction. Mammalian circadian clocks regulate the circadian cycles of various physiological functions. In vertebrates, circadian rhythms are mediated by a set of central clock genes: muscle and brain ARNT like-1 (Bmal1), muscle and brain ARNT like-2 (Bmal2), circadian rhythmic motion output cycle stagnates (Clock), cryptochrome 1 (Cry1), cryptochrome2 (Cry2), period 1 (Per1), period 2 (Per2), period 3 (Per3) and neuronal PAS domain protein 2 (Npas2). Negative feedback loops, controlled at both the transcriptional and posttranslational levels, adjust these clock genes in a diurnal manner. According to the results of studies on circadian transcriptomic studies in several tissues, most rhythmic genes are expressed in a tissue-specific manner and are affected by tissue-specific circadian rhythms. The circadian rhythm regulates several activities, including energy metabolism, feeding time, sleeping, and endocrine and immune functions. It has been reported that the circadian rhythms of mammals are closely related to bone metabolism. In this review, we discuss the regulation of the circadian rhythm/circadian clock gene in osteoblasts/osteoclasts and the energy metabolism of bone, and the relationship between circadian rhythm, bone remodeling, and energy metabolism. We also discuss the therapeutic potential of regulating circadian rhythms or changing energy metabolism on bone development/bone regeneration.
Collapse
Affiliation(s)
- Beibei Luo
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Xin Zhou
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Qingming Tang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Ying Yin
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Guangxia Feng
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Shue Li
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|
29
|
Ding G, Li X, Hou X, Zhou W, Gong Y, Liu F, He Y, Song J, Wang J, Basil P, Li W, Qian S, Saha P, Wang J, Cui C, Yang T, Zou K, Han Y, Amos CI, Xu Y, Chen L, Sun Z. REV-ERB in GABAergic neurons controls diurnal hepatic insulin sensitivity. Nature 2021; 592:763-767. [PMID: 33762728 PMCID: PMC8085086 DOI: 10.1038/s41586-021-03358-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 02/12/2021] [Indexed: 02/08/2023]
Abstract
Systemic insulin sensitivity shows a diurnal rhythm with a peak upon waking1,2. The molecular mechanism that underlies this temporal pattern is unclear. Here we show that the nuclear receptors REV-ERB-α and REV-ERB-β (referred to here as 'REV-ERB') in the GABAergic (γ-aminobutyric acid-producing) neurons in the suprachiasmatic nucleus (SCN) (SCNGABA neurons) control the diurnal rhythm of insulin-mediated suppression of hepatic glucose production in mice, without affecting diurnal eating or locomotor behaviours during regular light-dark cycles. REV-ERB regulates the rhythmic expression of genes that are involved in neurotransmission in the SCN, and modulates the oscillatory firing activity of SCNGABA neurons. Chemogenetic stimulation of SCNGABA neurons at waking leads to glucose intolerance, whereas restoration of the temporal pattern of either SCNGABA neuron firing or REV-ERB expression rescues the time-dependent glucose metabolic phenotype caused by REV-ERB depletion. In individuals with diabetes, an increased level of blood glucose after waking is a defining feature of the 'extended dawn phenomenon'3,4. Patients with type 2 diabetes with the extended dawn phenomenon exhibit a differential temporal pattern of expression of REV-ERB genes compared to patients with type 2 diabetes who do not have the extended dawn phenomenon. These findings provide mechanistic insights into how the central circadian clock regulates the diurnal rhythm of hepatic insulin sensitivity, with implications for our understanding of the extended dawn phenomenon in type 2 diabetes.
Collapse
Affiliation(s)
- Guolian Ding
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Xin Li
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Xinguo Hou
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenjun Zhou
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Yingyun Gong
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fuqiang Liu
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Yanlin He
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Laboratory of Brain Glycemia and Metabolism Control, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Jia Song
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Wang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Paul Basil
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Wenbo Li
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Sichong Qian
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
| | - Pradip Saha
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jinbang Wang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Chen Cui
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Tingting Yang
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Kexin Zou
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Younghun Han
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Christopher I Amos
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Li Chen
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China.
| | - Zheng Sun
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
30
|
"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
|
31
|
Luo S, Ezrokhi M, Cominos N, Tsai TH, Stoelzel CR, Trubitsyna Y, Cincotta AH. Experimental dopaminergic neuron lesion at the area of the biological clock pacemaker, suprachiasmatic nuclei (SCN) induces metabolic syndrome in rats. Diabetol Metab Syndr 2021; 13:11. [PMID: 33485386 PMCID: PMC7825247 DOI: 10.1186/s13098-021-00630-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The daily peak in dopaminergic neuronal activity at the area of the biological clock (hypothalamic suprachiasmatic nuclei [SCN]) is diminished in obese/insulin resistant vs lean/insulin sensitive animals. The impact of targeted lesioning of dopamine (DA) neurons specifically at the area surrounding (and that communicate with) the SCN (but not within the SCN itself) upon glucose metabolism, adipose and liver lipid gene expression, and cardiovascular biology in normal laboratory animals has not been investigated and was the focus of this study. METHODS Female Sprague-Dawley rats received either DA neuron neurotoxic lesion by bilateral intra-cannula injection of 6-hydroxydopamine (2-4 μg/side) or vehicle treatment at the area surrounding the SCN at 20 min post protriptyline ip injection (20 mg/kg) to protect against damage to noradrenergic and serotonergic neurons. RESULTS At 16 weeks post-lesion relative to vehicle treatment, peri-SCN area DA neuron lesioning increased weight gain (34.8%, P < 0.005), parametrial and retroperitoneal fat weight (45% and 90% respectively, P < 0.05), fasting plasma insulin, leptin and norepinephrine levels (180%, 71%, and 40% respectively, P < 0.05), glucose tolerance test area under the curve (AUC) insulin (112.5%, P < 0.05), and insulin resistance (44%-Matsuda Index, P < 0.05) without altering food consumption during the test period. Such lesion also induced the expression of several lipid synthesis genes in adipose and liver and the adipose lipolytic gene, hormone sensitive lipase in adipose (P < 0.05 for all). Liver monocyte chemoattractant protein 1 (a proinflammatory protein associated with metabolic syndrome) gene expression was also significantly elevated in peri-SCN area dopaminergic lesioned rats. Peri-SCN area dopaminergic neuron lesioned rats were also hypertensive (systolic BP rose from 157 ± 5 to 175 ± 5 mmHg, P < 0.01; diastolic BP rose from 109 ± 4 to 120 ± 3 mmHg, P < 0.05 and heart rate increase from 368 ± 12 to 406 ± 12 BPM, P < 0.05) and had elevated plasma norepinephrine levels (40% increased, P < 0.05) relative to controls. CONCLUSIONS These findings indicate that reduced dopaminergic neuronal activity in neurons at the area of and communicating with the SCN contributes significantly to increased sympathetic tone and the development of metabolic syndrome, without effect on feeding.
Collapse
Affiliation(s)
- Shuqin Luo
- VeroScience LLC, 1334 Main Road, Tiverton, RI, 02878, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Wood M, Whirledge S. Mechanism of glucocorticoid action in immunology—Basic concepts. REPRODUCTIVE IMMUNOLOGY 2021:147-170. [DOI: 10.1016/b978-0-12-818508-7.00020-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
33
|
McFarlane G, Guatelli-Steinberg D, Loch C, White S, Bayle P, Floyd B, Pitfield R, Mahoney P. An inconstant biorhythm: The changing pace of Retzius periodicity in human permanent teeth. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2020; 175:172-186. [PMID: 33368148 DOI: 10.1002/ajpa.24206] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/06/2020] [Accepted: 12/06/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Human tooth enamel retains evidence of growth in the form of Retzius lines. The number of daily growth increments between the regularly occurring lines defines their repeat interval, or periodicity. Retzius periodicity is often incorporated into enamel formation times, age-at-death reconstructions, or used to provide a basis from which to explore an underlying biorhythm. Biological anthropologists typically assume that RP remains constant within an individual and does not vary along the tooth-row. Here, we test that assumption. MATERIALS AND METHODS RP was calculated from n = 223 thin sections of human permanent teeth from individuals of British and southern African origin. Forty individuals provided multiple teeth (n = 102 teeth) and a further 121 individuals each provided a single tooth. RESULTS We report first evidence that RP of permanent teeth does not always remain constant within an individual. Of those individuals that provided multiple teeth, 42% (n = 17/40) demonstrated a decrease in RP along the tooth row, with most shifting by two or more days (n = 11). Across the entire sample, mean RP of anterior teeth was significantly higher than molars. Mean premolar RP tended to be intermediate between anterior teeth and molars. DISCUSSION Our data do not support the assumption that RP invariably remains constant within the permanent teeth of an individual. Transferring RP from molars to incisors within an individual can result in a miscalculation of formation time and age-at-death by up to 1 year. Implications for biological anthropologists and the source of the underlying long period biorhythm are discussed.
Collapse
Affiliation(s)
- Gina McFarlane
- Human Osteology Lab, School of Anthropology and Conservation, University of Kent, Canterbury, UK
| | - Debbie Guatelli-Steinberg
- Human Osteology Lab, School of Anthropology and Conservation, University of Kent, Canterbury, UK.,Department of Anthropology, The Ohio State University, Columbus, Ohio, USA
| | - Carolina Loch
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, New Zealand
| | - Sophie White
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, New Zealand
| | | | - Bruce Floyd
- School of Social Sciences, University of Auckland, New Zealand
| | - Rosie Pitfield
- Human Osteology Lab, School of Anthropology and Conservation, University of Kent, Canterbury, UK
| | - Patrick Mahoney
- Human Osteology Lab, School of Anthropology and Conservation, University of Kent, Canterbury, UK
| |
Collapse
|
34
|
The importance of 24-h metabolism in obesity-related metabolic disorders: opportunities for timed interventions. Int J Obes (Lond) 2020; 45:479-490. [PMID: 33235354 DOI: 10.1038/s41366-020-00719-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/18/2020] [Accepted: 11/03/2020] [Indexed: 11/08/2022]
Abstract
Various metabolic processes in the body oscillate throughout the natural day, driven by a biological clock. Circadian rhythms are also influenced by time cues from the environment (light exposure) and behaviour (eating and exercise). Recent evidence from diurnal- and circadian-rhythm studies indicates rhythmicity in various circulating metabolites, insulin secretion and -sensitivity and energy expenditure in metabolically healthy adults. These rhythms have been shown to be disturbed in adults with obesity-related metabolic disturbances. Moreover, eating and being (in)active at a time that the body is not prepared for it, as in night-shift work, is related to poor metabolic outcomes. These findings indicate the relevance of 24-h metabolism in obesity-related metabolic alterations and have also led to novel strategies, such as timing of food intake and exercise, to reinforce the circadian rhythm and thereby improving metabolic health. This review aims to deepen the understanding of the influence of the circadian system on metabolic processes and obesity-related metabolic disturbances and to discuss novel time-based strategies that may be helpful in combating metabolic disease.
Collapse
|
35
|
Zhang Z, Yu B, Wang X, Luo C, Zhou T, Zheng X, Ding J. Circadian rhythm and atherosclerosis (Review). Exp Ther Med 2020; 20:96. [PMID: 32973945 PMCID: PMC7506962 DOI: 10.3892/etm.2020.9224] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 08/11/2020] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis is the leading cause of morbidity and mortality worldwide. The underlying pathogenesis involves multiple metabolic disorders, endothelial dysfunction and a maladaptive immune response, and leads to chronic arterial wall inflammation. Numerous normal physiological activities exhibit daily rhythmicity, including energy metabolism, vascular function and inflammatory immunoreactions, and disrupted or misaligned circadian rhythms may promote the progression of atherosclerosis. However, the association between the circadian rhythm and atherosclerosis remains to be fully elucidated. In the present review, the effects of the circadian rhythm on atherosclerosis progression are discussed.
Collapse
Affiliation(s)
- Zaiqiang Zhang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Bin Yu
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Xinan Wang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Caiyun Luo
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Tian Zhou
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Xiaxia Zheng
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Jiawang Ding
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| |
Collapse
|
36
|
Liu D, Li J, Wu J, Dai J, Chen X, Huang Y, Zhang S, Tian B, Mei W. Monochromatic Blue Light Activates Suprachiasmatic Nucleus Neuronal Activity and Promotes Arousal in Mice Under Sevoflurane Anesthesia. Front Neural Circuits 2020; 14:55. [PMID: 32973462 PMCID: PMC7461971 DOI: 10.3389/fncir.2020.00055] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/27/2020] [Indexed: 01/17/2023] Open
Abstract
Background: Monochromatic blue light (MBL), with a wavelength between 400-490 nm, can regulate non-image-forming (NIF) functions of light in the central nervous system. The suprachiasmatic nucleus (SCN) in the brain is involved in the arousal-promoting response to blue light in mice. Animal and human studies showed that the responsiveness of the brain to visual stimuli is partly preserved under general anesthesia. Therefore, this study aimed to investigate whether MBL promotes arousal from sevoflurane anesthesia via activation of the SCN in mice. Methods: The induction and emergence time of sevoflurane anesthesia under MBL (460 nm and 800 lux) exposure was measured. Cortical electroencephalograms (EEGs) were recorded and the burst-suppression ratio (BSR) was calculated under MBL during sevoflurane anesthesia. The EEGs and local field potential (LFP) recordings with or without locally electrolytic ablated bilateral SCN were used to further explore the role of SCN in the arousal-promoting effect of MBL under sevoflurane anesthesia. Immunofluorescent staining of c-Fos was conducted to reveal the possible downstream mechanism of SCN activation. Results: Unlike the lack of effect on the induction time, MBL shortened the emergence time and the EEG recordings showed cortical arousal during the recovery period. MBL resulted in a significant decrease in BSR and a marked increase in EEG power at all frequency bands except for the spindle band during 2.5% sevoflurane anesthesia. MBL exposure under sevoflurane anesthesia enhances the neuronal activity of the SCN. These responses to MBL were abolished in SCN lesioned (SCNx) mice. MBL evoked a high level of c-Fos expression in the prefrontal cortex (PFC) and lateral hypothalamus (LH) compared to polychromatic white light (PWL) under sevoflurane anesthesia, while it exerted no effect on c-Fos expression in the ventrolateral preoptic area (VLPO) and locus coeruleus (LC) c-Fos expression. Conclusions: MBL promotes behavioral and electroencephalographic arousal from sevoflurane anesthesia via the activation of the SCN and its associated downstream wake-related nuclei. The clinical implications of this study warrant further study.
Collapse
Affiliation(s)
- Daiqiang Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiayan Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiayi Wu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaqi Dai
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
| | - Xinfeng Chen
- Chinese Institute for Brain Research (CIBR), ZGC Life Science Park, Beijing, China
| | - Yujie Huang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Tian
- Department of Neurobiology, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Neurological Diseases, Ministry of Education, Wuhan, China
| | - Wei Mei
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Shivshankar P, Fekry B, Eckel-Mahan K, Wetsel RA. Circadian Clock and Complement Immune System-Complementary Control of Physiology and Pathology? Front Cell Infect Microbiol 2020; 10:418. [PMID: 32923410 PMCID: PMC7456827 DOI: 10.3389/fcimb.2020.00418] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Mammalian species contain an internal circadian (i.e., 24-h) clock that is synchronized to the day and night cycles. Large epidemiological studies, which are supported by carefully controlled studies in numerous species, support the idea that chronic disruption of our circadian cycles results in a number of health issues, including obesity and diabetes, defective immune response, and cancer. Here we focus specifically on the role of the complement immune system and its relationship to the internal circadian clock system. While still an incompletely understood area, there is evidence that dysregulated proinflammatory cytokines, complement factors, and oxidative stress can be induced by circadian disruption and that these may feed back into the oscillator at the level of circadian gene regulation. Such a feedback cycle may contribute to impaired host immune response against pathogenic insults. The complement immune system including its activated anaphylatoxins, C3a and C5a, not only facilitate innate and adaptive immune response in chemotaxis and phagocytosis, but they can also amplify chronic inflammation in the host organism. Consequent development of autoimmune disorders, and metabolic diseases associated with additional environmental insults that activate complement can in severe cases, lead to accelerated tissue dysfunction, fibrosis, and ultimately organ failure. Because several promising complement-targeted therapeutics to block uncontrolled complement activation and treat autoimmune diseases are in various phases of clinical trials, understanding fully the circadian properties of the complement system, and the reciprocal regulation by these two systems could greatly improve patient treatment in the long term.
Collapse
Affiliation(s)
- Pooja Shivshankar
- Research Center for Immunology and Autoimmune Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Baharan Fekry
- Center for Metabolic and Degenerative Diseases, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Kristin Eckel-Mahan
- Center for Metabolic and Degenerative Diseases, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Rick A. Wetsel
- Research Center for Immunology and Autoimmune Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
38
|
van der Vinne V, Martin Burgos B, Harrington ME, Weaver DR. Deconstructing circadian disruption: Assessing the contribution of reduced peripheral oscillator amplitude on obesity and glucose intolerance in mice. J Pineal Res 2020; 69:e12654. [PMID: 32243642 DOI: 10.1111/jpi.12654] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/27/2022]
Abstract
Disturbing the circadian regulation of physiology by disruption of the rhythmic environment is associated with adverse health outcomes but the underlying mechanisms are unknown. Here, the response of central and peripheral circadian clocks to an advance or delay of the light-dark cycle was determined in mice. This identified transient damping of peripheral clocks as a consequence of an advanced light-dark cycle. Similar depression of peripheral rhythm amplitude was observed in mice exposed to repeated phase shifts. To assess the metabolic consequences of such peripheral amplitude depression in isolation, temporally chimeric mice lacking a functional central clock (Vgat-Cre+ Bmal1fl/fl ) were housed in the absence of environmental rhythmicity. In vivo PER2::LUC bioluminescence imaging of anesthetized and freely moving mice revealed that this resulted in a state of peripheral amplitude depression, similar in severity to that observed transiently following an advance of the light-dark cycle. Surprisingly, our mice did not show alterations in body mass or glucose tolerance in males or females on regular or high-fat diets. Overall, our results identify transient damping of peripheral rhythm amplitude as a consequence of exposure to an advanced light-dark cycle but chronic damping of peripheral clocks in isolation is insufficient to induce adverse metabolic outcomes in mice.
Collapse
Affiliation(s)
- Vincent van der Vinne
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| | | | | | - David R Weaver
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
39
|
Heyde I, Oster H. Network-Like Organization of the Circadian System Regulates Metabolic Homeostasis. Obesity (Silver Spring) 2020; 28 Suppl 1:S8-S9. [PMID: 32419315 DOI: 10.1002/oby.22773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 02/04/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Isabel Heyde
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
40
|
Moore MC, Smith MS, Swift LL, Cincotta AH, Ezrokhi M, Cominos N, Zhang Y, Farmer B, Cherrington AD. Bromocriptine mesylate improves glucose tolerance and disposal in a high-fat-fed canine model. Am J Physiol Endocrinol Metab 2020; 319:E133-E145. [PMID: 32459527 PMCID: PMC7468784 DOI: 10.1152/ajpendo.00479.2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Bromocriptine mesylate treatment was examined in dogs fed a high fat diet (HFD) for 8 wk. After 4 wk on HFD, daily bromocriptine (Bromo; n = 6) or vehicle (CTR; n = 5) injections were administered. Oral glucose tolerance tests were performed before beginning HFD (OGTT1), 4 wk after HFD began (Bromo only), and after 7.5 wk on HFD (OGTT3). After 8 wk on HFD, clamp studies were performed, with infusion of somatostatin and intraportal replacement of insulin (4× basal) and glucagon (basal). From 0 to 90 min (P1), glucose was infused via peripheral vein to double the hepatic glucose load; and from 90 to 180 min (P2), glucose was infused via the hepatic portal vein at 4 mg·kg-1·min-1, with the HGL maintained at 2× basal. Bromo decreased the OGTT glucose ΔAUC0-30 and ΔAUC0-120 by 62 and 27%, respectively, P < 0.05 for both) without significantly altering the insulin response. Bromo dogs exhibited enhanced net hepatic glucose uptake (NHGU) compared with CTR (~33 and 21% greater, P1 and P2, respectively, P < 0.05). Nonhepatic glucose uptake (non-HGU) was increased ~38% in Bromo in P2 (P < 0.05). Bromo vs. CTR had higher (P < 0.05) rates of glucose infusion (36 and 30%) and non-HGU (~40 and 27%) than CTR during P1 and P2, respectively. In Bromo vs. CTR, hepatic 18:0/16:0 and 16:1/16:0 ratios tended to be elevated in triglycerides and were higher (P < 0.05) in phospholipids, consistent with a beneficial effect of bromocriptine on liver fat accumulation. Thus, bromocriptine treatment improved glucose disposal in a glucose-intolerant model, enhancing both NHGU and non-HGU.
Collapse
Affiliation(s)
- Mary Courtney Moore
- Department of Metabolic Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Marta S Smith
- Department of Metabolic Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Larry L Swift
- Vanderbilt Diabetes Research and Training Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | | | | | - Ben Farmer
- Department of Metabolic Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
- Vanderbilt Diabetes Research and Training Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Alan D Cherrington
- Department of Metabolic Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
41
|
Purnell BS, Buchanan GF. Free-running circadian breathing rhythms are eliminated by suprachiasmatic nucleus lesion. J Appl Physiol (1985) 2020; 129:49-57. [PMID: 32501775 PMCID: PMC7469233 DOI: 10.1152/japplphysiol.00211.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/06/2020] [Accepted: 06/02/2020] [Indexed: 11/22/2022] Open
Abstract
It is widely agreed that breathing is subject to circadian regulation. Circadian differences in respiratory physiology significantly impact a number of diseases including sleep apnea, asthma, and seizure-induced death. The effect of time of day on breathing has been previously characterized; however, an endogenous free-running respiratory rhythm in mammals has not previously been described. Furthermore, it is assumed that circadian rhythms in breathing are dependent on the hypothalamic suprachiasmatic nucleus (SCN), the home of the mammalian central circadian oscillator, but this has not been shown experimentally. The breathing of mice was monitored during wakefulness using whole body plethysmography at six times of day while housed under light-dark conditions and at six circadian phases while housed under constant darkness. Respiratory frequency and minute ventilation, but not tidal volume, were significantly higher during the active phase in both entrained and free-running conditions. To determine whether circadian regulation of breathing requires the SCN, in separate sets of animals this structure was electrolytically lesioned bilaterally or a sham surgery was performed, and breathing was measured at six different time points. Time-dependent oscillations in breathing were lost in SCN-lesioned animals, but not those subjected to sham surgery. These results suggest that breathing is subject to circadian regulation via the SCN. Mechanistic insights into the circadian regulation of breathing may lead to targeted interventions to reduce the morbidity and mortality associated with diseases with respiratory pathophysiology.NEW & NOTEWORTHY It has long been appreciated that breathing is altered by time of day. This study demonstrates that rhythmicity in breathing persists in constant darkness but is dependent on the suprachiasmatic nucleus in the hypothalamus. Understanding circadian rhythms in breathing may be important for the treatment and prevention of diseases such as sleep apnea and sudden unexpected death in epilepsy.
Collapse
Affiliation(s)
- Benton S Purnell
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Gordon F Buchanan
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
42
|
Maroni MJ, Capri KM, Arruda NL, Gelineau RR, Deane HV, Concepcion HA, DeCourcey H, Monteiro De Pina IK, Cushman AV, Chasse MH, Logan RW, Seggio JA. Substrain specific behavioral responses in male C57BL/6N and C57BL/6J mice to a shortened 21-hour day and high-fat diet. Chronobiol Int 2020; 37:809-823. [PMID: 32400203 DOI: 10.1080/07420528.2020.1756840] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Altered circadian rhythms have negative consequences on health and behavior. Emerging evidence suggests genetics influences the physiological and behavioral responses to circadian disruption. We investigated the effects of a 21 h day (T = 21 cycle), with high-fat diet consumption, on locomotor activity, explorative behaviors, and health in male C57BL/6J and C57BL/6N mice. Mice were exposed to either a T = 24 or T = 21 cycle and given standard rodent chow (RC) or a 60% high-fat diet (HFD) followed by behavioral assays and physiological measures. We uncovered numerous strain differences within the behavioral and physiological assays, mainly that C57BL/6J mice exhibit reduced susceptibility to the obesogenic effects of (HFD) and anxiety-like behavior as well as increased circadian and novelty-induced locomotor activity compared to C57BL/6N mice. There were also substrain-specific differences in behavioral responses to the T = 21 cycle, including exploratory behaviors and circadian locomotor activity. Under the 21-h day, mice consuming RC displayed entrainment, while mice exposed to HFD exhibited a lengthening of activity rhythms. In the open-field and light-dark box, mice exposed to the T = 21 cycle had increased novelty-induced locomotor activity with no further effects of diet, suggesting daylength may affect mood-related behaviors. These results indicate that different circadian cycles impact metabolic and behavioral responses depending on genetic background, and despite circadian entrainment.
Collapse
Affiliation(s)
- Marissa J Maroni
- Department of Biological Sciences, Bridgewater State University , Bridgewater, Massachusetts, USA.,Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania, USA
| | - Kimberly M Capri
- Department of Biological Sciences, Bridgewater State University , Bridgewater, Massachusetts, USA.,Department of Mathematics and Statistics, Boston University , Boston, Massachusetts, USA
| | - Nicole L Arruda
- Department of Biological Sciences, Bridgewater State University , Bridgewater, Massachusetts, USA.,Chapel Hill, Biological and Biomedical Sciences Program, University of North Carolina , Chapel Hill, North Carolina, USA
| | - Rachel R Gelineau
- Department of Biological Sciences, Bridgewater State University , Bridgewater, Massachusetts, USA
| | - Hannah V Deane
- Department of Biological Sciences, Bridgewater State University , Bridgewater, Massachusetts, USA
| | - Holly A Concepcion
- Department of Biological Sciences, Bridgewater State University , Bridgewater, Massachusetts, USA
| | - Holly DeCourcey
- Department of Biological Sciences, Bridgewater State University , Bridgewater, Massachusetts, USA
| | | | - Alexis V Cushman
- Department of Biological Sciences, Bridgewater State University , Bridgewater, Massachusetts, USA
| | - Madison H Chasse
- Department of Biological Sciences, Bridgewater State University , Bridgewater, Massachusetts, USA
| | - Ryan W Logan
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania, USA.,Center for Systems Neurogenetics of Addiction, The Jackson Laboratory , Bar Harbor, Maine, USA
| | - Joseph A Seggio
- Department of Biological Sciences, Bridgewater State University , Bridgewater, Massachusetts, USA
| |
Collapse
|
43
|
Pickel L, Sung HK. Feeding Rhythms and the Circadian Regulation of Metabolism. Front Nutr 2020; 7:39. [PMID: 32363197 PMCID: PMC7182033 DOI: 10.3389/fnut.2020.00039] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/20/2020] [Indexed: 12/16/2022] Open
Abstract
The molecular circadian clock regulates metabolic processes within the cell, and the alignment of these clocks between tissues is essential for the maintenance of metabolic homeostasis. The possibility of misalignment arises from the differential responsiveness of tissues to the environmental cues that synchronize the clock (zeitgebers). Although light is the dominant environmental cue for the master clock of the suprachiasmatic nucleus, many other tissues are sensitive to feeding and fasting. When rhythms of feeding behavior are altered, for example by shift work or the constant availability of highly palatable foods, strong feedback is sent to the peripheral molecular clocks. Varying degrees of phase shift can cause the systemic misalignment of metabolic processes. Moreover, when there is a misalignment between the endogenous rhythms in physiology and environmental inputs, such as feeding during the inactive phase, the body's ability to maintain homeostasis is impaired. The loss of phase coordination between the organism and environment, as well as internal misalignment between tissues, can produce cardiometabolic disease as a consequence. The aim of this review is to synthesize the work on the mechanisms and metabolic effects of circadian misalignment. The timing of food intake is highlighted as a powerful environmental cue with the potential to destroy or restore the synchrony of circadian rhythms in metabolism.
Collapse
Affiliation(s)
- Lauren Pickel
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Hoon-Ki Sung
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
44
|
de Luis DA, Izaola O, Primo D, Aller R. A circadian rhythm-related MTNR1B genetic variant (rs10830963) modulate body weight change and insulin resistance after 9 months of a high protein/low carbohydrate vs a standard hypocaloric diet. J Diabetes Complications 2020; 34:107534. [PMID: 32057567 DOI: 10.1016/j.jdiacomp.2020.107534] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS The risk allele (G) of rs10830963 in the melatonin receptor 1 B (MTNR1B) gene presents an association with biochemical parameters and obesity. We study the effect of this SNP on insulin resistance and weight loss secondary to two hypocaloric diets. METHODS 270 obese subjects were randomly allocated during 9 months (Diet HP: a high protein/low carbohydrate vs. Diet S: a standard severe hypocaloric diets). Anthropometric parameters, fasting blood glucose, C-reactive protein (CRP), insulin concentration, insulin resistance (HOMA-IR), lipid profile and adipocytokines levels were measured. Genotype of MTNR1B gene polymorphism (rs10830963) was evaluated. RESULTS All adiposity parameters, systolic blood pressure and leptin levels decreased in all subjects after both diets. This improvement of adiposity parameters was higher in non-G allele carriers than G allele carriers. After weight loss with Diet HP, (CC vs. CG + GG at 9 months); total cholesterol (delta: -9.9 ± 2.4 mg/dl vs. -4.8 ± 2.2 mg/dl:p < 0.05), LDL-cholesterol (delta: -8.3 ± 1.9 mg/dl vs. -5.1 ± 2.2 mg/dl: p < 0.05), insulin (delta: -4.7 ± 0.8 UI/L vs. -0.9 ± 1.0 UI/L: p < 0.05), triglycerides (delta: -17.7 ± 3.9 mg/dl vs. -6.1 ± 2.8 mg/dl: p < 0.05) and HOMA IR (delta: -0.8 ± 0.2 units vs. -0.2 ± 0.1 units: p < 0.05) improved only in no G allele carriers. After weight loss with Diet S in non G allele carriers, insulin levels (delta (CC vs. CG + GG): -3.4 ± 0.6 UI/L vs. -1.2 ± 0.4 UI/L: p < 0.05), triglycerides (delta: -29.2 ± 3.4 mg/dl vs. -8.2 ± 3.8 mg/dl: p < 0.05), HOMA-IR (delta (CC vs. CG + GG): -1.1 ± 0.2 units vs. -0.1 ± 0.1 units: p < 0.05), total cholesterol (delta: -15.9 ± 7.4 mg/dl vs. -5.8 ± 2.9 mg/dl:ns) and LDL-cholesterol (delta: -13.7 ± 5.9 mg/dl vs. -6.0 ± 2.9 mg/dl: ns) decreased, too. CONCLUSIONS our study detected a relationship of rs10830963 variant of MTNR1B gene with adiposity changes, cholesterol changes and insulin resistance modification induced by two different hypocaloric during 9 months.
Collapse
Affiliation(s)
- Daniel Antonio de Luis
- Endocrinology and Nutrition Research Center, School of Medicine, Department of Endocrinology and Nutrition, Hospital Clinico Universitario, University of Valladolid, Valladolid, Spain.
| | - Olatz Izaola
- Endocrinology and Nutrition Research Center, School of Medicine, Department of Endocrinology and Nutrition, Hospital Clinico Universitario, University of Valladolid, Valladolid, Spain
| | - David Primo
- Endocrinology and Nutrition Research Center, School of Medicine, Department of Endocrinology and Nutrition, Hospital Clinico Universitario, University of Valladolid, Valladolid, Spain
| | - Rocio Aller
- Endocrinology and Nutrition Research Center, School of Medicine, Department of Endocrinology and Nutrition, Hospital Clinico Universitario, University of Valladolid, Valladolid, Spain
| |
Collapse
|
45
|
Nishimura Y, Mabuchi K, Omura N, Igarashi A, Miura M, Mima N, Negishi H, Morimoto K, Takamata A. Fluoxetine Mimics the Anorectic Action of Estrogen and Its Regulation of Circadian Feeding in Ovariectomized Female Rats. Nutrients 2020; 12:nu12030849. [PMID: 32235766 PMCID: PMC7146435 DOI: 10.3390/nu12030849] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 01/09/2023] Open
Abstract
Our previous study demonstrated that chronic estrogen replacement in ovariectomized rats reduces food intake and augments c-Fos expression in the suprachiasmatic nucleus (SCN), specifically during the light phase. Here, we hypothesized that serotonergic neurons in the central nervous system (CNS), which have anorectic action and play a role in regulating circadian rhythm, mediate the light phase-specific anorectic action of estrogen, and that selective serotonin reuptake inhibitors (SSRIs) mimic the hypophagic action of estrogen. Female Wistar rats were ovariectomized and treated with estradiol (E2) or cholesterol by subcutaneously implanting a silicon capsule containing E2 or cholesterol. Then, half of the cholesterol-treated rats were injected with the SSRI fluoxetine (5 mg/kg) (FLX group), while the remaining rats in the cholesterol-treated group (CON group) and all those in the E2 group were injected with saline subcutaneously twice daily at the onsets of the light and dark phases. Both E2 and FLX reduced food intake during the light phase but not the dark phase, and reduced body weight gain. In addition, both E2 and FLX augmented the c-Fos expression in the SCN, specifically during the light phase. These data indicate that FLX exerts estrogen-like antiobesity and hypophagic actions by modifying circadian feeding patterns, and suggest that estrogen regulates circadian feeding rhythm via serotonergic neurons in the CNS.
Collapse
Affiliation(s)
- Yuri Nishimura
- Department of Environmental Health, Nara Women’s University, Kitauoya Nishimachi, Nara 630-8506, Japan; (Y.N.); (K.M.); (N.O.); (A.I.); (M.M.); (N.M.); (H.N.); (K.M.)
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Kaori Mabuchi
- Department of Environmental Health, Nara Women’s University, Kitauoya Nishimachi, Nara 630-8506, Japan; (Y.N.); (K.M.); (N.O.); (A.I.); (M.M.); (N.M.); (H.N.); (K.M.)
| | - Natsumi Omura
- Department of Environmental Health, Nara Women’s University, Kitauoya Nishimachi, Nara 630-8506, Japan; (Y.N.); (K.M.); (N.O.); (A.I.); (M.M.); (N.M.); (H.N.); (K.M.)
| | - Ayako Igarashi
- Department of Environmental Health, Nara Women’s University, Kitauoya Nishimachi, Nara 630-8506, Japan; (Y.N.); (K.M.); (N.O.); (A.I.); (M.M.); (N.M.); (H.N.); (K.M.)
| | - Megumi Miura
- Department of Environmental Health, Nara Women’s University, Kitauoya Nishimachi, Nara 630-8506, Japan; (Y.N.); (K.M.); (N.O.); (A.I.); (M.M.); (N.M.); (H.N.); (K.M.)
| | - Nanako Mima
- Department of Environmental Health, Nara Women’s University, Kitauoya Nishimachi, Nara 630-8506, Japan; (Y.N.); (K.M.); (N.O.); (A.I.); (M.M.); (N.M.); (H.N.); (K.M.)
| | - Hiroko Negishi
- Department of Environmental Health, Nara Women’s University, Kitauoya Nishimachi, Nara 630-8506, Japan; (Y.N.); (K.M.); (N.O.); (A.I.); (M.M.); (N.M.); (H.N.); (K.M.)
| | - Keiko Morimoto
- Department of Environmental Health, Nara Women’s University, Kitauoya Nishimachi, Nara 630-8506, Japan; (Y.N.); (K.M.); (N.O.); (A.I.); (M.M.); (N.M.); (H.N.); (K.M.)
| | - Akira Takamata
- Department of Environmental Health, Nara Women’s University, Kitauoya Nishimachi, Nara 630-8506, Japan; (Y.N.); (K.M.); (N.O.); (A.I.); (M.M.); (N.M.); (H.N.); (K.M.)
- Correspondence: ; Tel./Fax: +81-742-20-3469
| |
Collapse
|
46
|
de Luis DA, Izaola O, Primo D, Aller R. Dietary-fat effect of the rs10830963 polymorphism in MTNR1B on insulin resistance in response to 3 months weight-loss diets. ENDOCRINOLOGÍA, DIABETES Y NUTRICIÓN (ENGLISH ED.) 2020. [DOI: 10.1016/j.endien.2019.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
47
|
Postolache TT, Gulati A, Okusaga OO, Stiller JW. An Introduction to Circadian Endocrine Physiology: Implications for Exercise and Sports Performance. ENDOCRINOLOGY OF PHYSICAL ACTIVITY AND SPORT 2020. [DOI: 10.1007/978-3-030-33376-8_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
48
|
del Río-Martín A, Pérez-Taboada I, Fernández-Pérez A, Moratalla R, de la Villa P, Vallejo M. Hypomorphic Expression of Pitx3 Disrupts Circadian Clocks and Prevents Metabolic Entrainment of Energy Expenditure. Cell Rep 2019; 29:3678-3692.e4. [DOI: 10.1016/j.celrep.2019.11.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 05/13/2019] [Accepted: 11/06/2019] [Indexed: 12/11/2022] Open
|
49
|
Kolbe I, Leinweber B, Brandenburger M, Oster H. Circadian clock network desynchrony promotes weight gain and alters glucose homeostasis in mice. Mol Metab 2019; 30:140-151. [PMID: 31767165 PMCID: PMC6807374 DOI: 10.1016/j.molmet.2019.09.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 09/23/2019] [Accepted: 09/28/2019] [Indexed: 11/21/2022] Open
Abstract
Objective A network of endogenous circadian clocks adapts physiology and behavior to recurring changes in environmental demands across the 24-hour day cycle. Circadian disruption promotes weight gain and type 2 diabetes development. In this study, we aim to dissect the roles of different tissue clocks in the regulation of energy metabolism. Methods We used mice with genetically ablated clock function in the circadian pacemaker of the suprachiasmatic nucleus (SCN) under different light and feeding conditions to study peripheral clock resetting and the role of the peripheral clock network in the regulation of glucose handling and metabolic homeostasis. Results In SCN clock-deficient mice, behavioral and non-SCN tissue clock rhythms are sustained under rhythmic lighting conditions but deteriorate quickly in constant darkness. In parallel to the loss of behavioral and molecular rhythms, the animals develop adiposity and impaired glucose utilization in constant darkness. Restoring peripheral clock rhythmicity and synchrony by time-restricted feeding normalizes body weight and glucose metabolism. Conclusions These data reveal the importance of an overall synchronized circadian clockwork for the maintenance of metabolic homeostasis. In mice with a non-functional SCN clock (SCN-KO), metabolic rhythms are retained in light-dark, but not in constant darkness (DD) conditions. Normal body weight regulation and glucose utilization do not require a functional SCN clock. Restoring peripheral clock gene expression rhythms via time-restricted feeding restores metabolic homeostasis in SCN-KO mice in DD.
Collapse
Affiliation(s)
- Isa Kolbe
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | - Brinja Leinweber
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | - Matthias Brandenburger
- Fraunhofer Research Institution for Marine Biotechnology and Cell Technology, Lübeck, Germany
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
50
|
Abstract
Circadian rhythms are driven by a transcription-translation feedback loop that separates anabolic and catabolic processes across the Earth's 24-h light-dark cycle. Central pacemaker neurons that perceive light entrain a distributed clock network and are closely juxtaposed with hypothalamic neurons involved in regulation of sleep/wake and fast/feeding states. Gaps remain in identifying how pacemaker and extrapacemaker neurons communicate with energy-sensing neurons and the distinct role of circuit interactions versus transcriptionally driven cell-autonomous clocks in the timing of organismal bioenergetics. In this review, we discuss the reciprocal relationship through which the central clock drives appetitive behavior and metabolic homeostasis and the pathways through which nutrient state and sleep/wake behavior affect central clock function.
Collapse
Affiliation(s)
- Jonathan Cedernaes
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Nathan Waldeck
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Joseph Bass
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| |
Collapse
|