1
|
Zhu C, Li S, Zhang H. Heart Failure and Arrhythmias: Circadian and Epigenetic Interplay in Myocardial Electrophysiology. Int J Mol Sci 2025; 26:2728. [PMID: 40141370 PMCID: PMC11943068 DOI: 10.3390/ijms26062728] [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: 02/12/2025] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
Emerging evidence underscores the impact of circadian rhythms on cardiovascular processes, particularly in conditions such as hypertension, myocardial infarction, and heart failure, where circadian rhythm disruptions are linked to disease progression and adverse clinical outcomes. Circadian clock proteins are intricately linked to myocardial electrophysiological remodeling and epigenetic pathways associated with arrhythmias in heart failure. In the context of heart failure, circadian clock dysregulation leads to electrophysiological remodeling in the cardiomyocytes, which can precipitate life-threatening arrhythmias such as ventricular tachycardia (VT) and ventricular fibrillation (VF). This dysregulation may be influenced by environmental factors, such as diet and exercise, as well as genetic factors. Moreover, epigenetic modifications in heart failure have been implicated in the regulation of genes involved in cardiac hypertrophy, fibrosis, and inflammation. The interplay between circadian clock proteins, myocardial electrophysiological remodeling, and epigenetic pathways in heart failure-related arrhythmias is complex and multifaceted. Further research is needed to elucidate how these processes interact and contribute to the development of arrhythmias in heart failure patients. This review aims to explore the connections between circadian rhythms, myocardial electrophysiology, and arrhythmias related to heart failure, with the goal of identifying potential therapeutic targets and interventions that may counteract the adverse effects of circadian disruptions on cardiovascular health.
Collapse
Affiliation(s)
- Chen Zhu
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646099, China; (C.Z.); (S.L.)
| | - Shuang Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646099, China; (C.Z.); (S.L.)
| | - Henggui Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646099, China; (C.Z.); (S.L.)
- Beijing Academy of Artificial Intelligence, Beijing 100084, China
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
2
|
Tofani GSS, Clarke G, Cryan JF. I "Gut" Rhythm: the microbiota as a modulator of the stress response and circadian rhythms. FEBS J 2025; 292:1454-1479. [PMID: 39841560 PMCID: PMC11927059 DOI: 10.1111/febs.17400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/20/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025]
Abstract
Modern habits are becoming more and more disruptive to health. As our days are often filled with circadian disruption and stress exposures, we need to understand how our responses to these external stimuli are shaped and how their mediators can be targeted to promote health. A growing body of research demonstrates the role of the gut microbiota in influencing brain function and behavior. The stress response and circadian rhythms, which are essential to maintaining appropriate responses to the environment, are known to be impacted by the gut microbiota. Gut microbes have been shown to alter the host's response to stress and modulate circadian rhythmicity. Although studies demonstrated strong links between the gut microbiota, circadian rhythms and the stress response, such studies were conducted in an independent manner not conducive to understanding the interface between these factors. Due to the interconnected nature of the stress response and circadian rhythms, in this review we explore how the gut microbiota may play a role in regulating the integration of stress and circadian signals in mammals and the consequences for brain health and disease.
Collapse
Affiliation(s)
- Gabriel S S Tofani
- APC Microbiome, University College Cork, Ireland
- Department of Anatomy & Neuroscience, University College Cork, Ireland
| | - Gerard Clarke
- APC Microbiome, University College Cork, Ireland
- Department of Psychiatry & Neurobehavioural Science, University College Cork, Ireland
| | - John F Cryan
- APC Microbiome, University College Cork, Ireland
- Department of Anatomy & Neuroscience, University College Cork, Ireland
| |
Collapse
|
3
|
Madbouly NA, Kamal SM, El-Amir AM. Chronic artificial light exposure in daytime and reversed light: Dark cycle inhibit anti-apoptotic cytokines and defect Bcl-2 in peripheral lymphoid tissues during acute systemic inflammatory response to lipopolysaccharide. Int Immunopharmacol 2025; 145:113768. [PMID: 39672023 DOI: 10.1016/j.intimp.2024.113768] [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: 06/17/2024] [Revised: 11/21/2024] [Accepted: 11/30/2024] [Indexed: 12/15/2024]
Abstract
AIMS The disturbed light: dark (LD) cycle has been associated with critical complications, including obesity, diabetes and cancer. In the present study, we investigated the chronic effects of artificial light at daytime (AL) and light at night (RAL) after intraperitoneal (i.p.) injection of saline and 0.5 mg/kg lipopolysaccharide (LPS) in male Wistar rats. METHODS Liver and kidney parameters, fasting blood glucose (FBG), melatonin level, immunohistochemical examinations of B-cell lymphoma-2 (Bcl-2) in spleen and mesenteric lymph and serum antiapoptotic cytokines [interleukin (IL-) 2, 7 and 1]. KEY FINDINGS After 16 weeks of a daily disturbed LD cycle, RAL increased body weight, upgraded FBG and altered liver and kidney functions with surprisingly increased daytime plasma melatonin. AL + LPS and RAL + LPS rats suffered significantly higher oxidative-nitrosative stress compared to NL + LPS. Oxidative-nitrosative stress was associated with multi-organ inflammation in hepatic, renal, pancreatic, splenic and mesenteric lymph node tissues due to LPS-induced endotoxemia. Anti-apoptotic Bcl-2 activity in peripheral lymphoid organs (spleen and mesenteric lymph node) was lowered due to AL and RAL regimens. At the same pattern, lowering of antiapoptotic serum levels of IL-2, IL-7 and IL-15 indicate alteration of cell cycle and the shifted ability of cells to undergo apoptosis due to abnormal light pollution. SIGNIFICANCE Here, the increased lymphocyte apoptosis in lymphoid tissues due to disturbed LD cycle defects the host defense, dysregulates the inflammatory immune response and dysregulates the immune tolerance during acute systemic inflammation due to LPS.
Collapse
|
4
|
Jiao H, Kalsbeek A, Yi CX. Microglia, circadian rhythm and lifestyle factors. Neuropharmacology 2024; 257:110029. [PMID: 38852838 DOI: 10.1016/j.neuropharm.2024.110029] [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: 02/19/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Microglia, a vital homeostasis-keeper of the central nervous system, perform critical functions such as synaptic pruning, clearance of cellular debris, and participation in neuroinflammatory processes. Recent research has shown that microglia exhibit strong circadian rhythms that not only actively regulate their own immune activity, but also affect neuronal function. Disruptions of the circadian clock have been linked to a higher risk of developing a variety of diseases. In this article we will provide an overview of how lifestyle factors impact microglial function, with a focus on disruptions caused by irregular sleep-wake patterns, reduced physical activity, and eating at the wrong time-of-day. We will also discuss the potential connection between these lifestyle factors, disrupted circadian rhythms, and the role of microglia in keeping brain health. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Han Jiao
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam, the Netherlands; Department of Clinical Chemistry, Laboratory of Endocrinology, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam, the Netherlands; Department of Clinical Chemistry, Laboratory of Endocrinology, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam, the Netherlands; Department of Clinical Chemistry, Laboratory of Endocrinology, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Amsterdam, the Netherlands.
| |
Collapse
|
5
|
Baek JH, Zhu Y, Jackson CL, Mark Park YM. Artificial Light at Night and Type 2 Diabetes Mellitus. Diabetes Metab J 2024; 48:847-863. [PMID: 39313230 PMCID: PMC11449813 DOI: 10.4093/dmj.2024.0237] [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: 05/08/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
The widespread and pervasive use of artificial light at night (ALAN) in our modern 24-hour society has emerged as a substantial disruptor of natural circadian rhythms, potentially leading to a rise in unhealthy lifestyle-related behaviors (e.g., poor sleep; shift work). This phenomenon has been associated with an increased risk of type 2 diabetes mellitus (T2DM), which is a pressing global public health concern. However, to date, reviews summarizing associations between ALAN and T2DM have primarily focused on the limited characteristics of exposure (e.g., intensity) to ALAN. This literature review extends beyond prior reviews by consolidating recent studies from 2000 to 2024 regarding associations between both indoor and outdoor ALAN exposure and the incidence or prevalence of T2DM. We also described potential biological mechanisms through which ALAN modulates glucose metabolism. Furthermore, we outlined knowledge gaps and investigated how various ALAN characteristics beyond only light intensity (including light type, timing, duration, wavelength, and individual sensitivity) influence T2DM risk. Recognizing the detrimental impact of ALAN on sleep health and the behavioral correlates of physical activity and dietary patterns, we additionally summarized studies investigating the potential mediating role of each component in the relationship between ALAN and glucose metabolism. Lastly, we proposed implications of chronotherapies and chrononutrition for diabetes management in the context of ALAN exposure.
Collapse
Affiliation(s)
- Jong-Ha Baek
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
- Department of Epidemiology, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yong Zhu
- Department of Epidemiology, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Chandra L. Jackson
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
- Division of Intramural Research, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Yong-Moon Mark Park
- Department of Epidemiology, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
6
|
Li B, Suzuki-Kerr H, Martis RM, Lim CJJ, Wang ZA, Nguyen TX, Donaldson PJ, Poulsen RC, Lim JC. Time of day differences in the regulation of glutathione levels in the rat lens. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1407582. [PMID: 39211001 PMCID: PMC11358124 DOI: 10.3389/fopht.2024.1407582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024]
Abstract
Introduction Evidence in non-ocular tissues indicate that the antioxidant glutathione (GSH) may be regulated in a circadian manner leading to the idea that GSH levels in the lens may also be controlled in a circadian manner to anticipate periods of oxidative stress. Methods Male rat Wistar lenses (6 weeks) were collected every 4 hours over a 24-hour period at 6am, 10am, 2pm, 6pm, 10pm and 2am and quantitative-PCR, western blotting and immunohistochemistry performed to examine the expression of core clock genes and proteins (BMAL1, CLOCK, CRY1-2, PER 1-3) and their subcellular localisation over a 24-hour period. Western blotting of lenses was also performed to examine the expression of NRF2, a transcription factor involved in regulating genes involved in GSH homeostasis and GSH related enzymes (GCLC, GS and GR) over the 24-hour period. Finally, HLPC was used to measure GSH levels in the aqueous humour and lenses every 4 hours over a 24-hour period. Results The rat lens contains the core molecular components of a circadian clock with the expression of core clock proteins, NRF2 and GSH related enzymes fluctuating over a 24-hour period. BMAL1 expression was highest during the day, with BMAL1 localised to the nuclei at 10am. NRF2 expression remained constant over the 24-hour period, although appeared to move in and out of the nuclei every 4 hours. GSH related enzyme expression tended to peak at the start of night which correlated with high levels of GSH in the lens and lower levels of GSH in the aqueous humour. Conclusion The lens contains the key components of a circadian clock, and time-of-day differences exist in the expression of GSH and GSH related enzymes involved in maintaining GSH homeostasis. GSH levels in the rat lens were highest at the start of night which represents the active phase of the rat when high GSH levels may be required to counteract oxidative stress induced by cellular metabolism. Future work to directly link the clock to regulation of GSH levels in the lens will be important in determining whether the clock can be used to help restore GSH levels in the lens.
Collapse
Affiliation(s)
- Bo Li
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
- New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Haruna Suzuki-Kerr
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
- New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Renita M. Martis
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
- New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Christopher J. J. Lim
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
- New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Zhou-ai Wang
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
- New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Tai X. Nguyen
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
- New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Paul J. Donaldson
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
- New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Raewyn C. Poulsen
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
| | - Julie C. Lim
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
- New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| |
Collapse
|
7
|
Rao F, Xue T. Circadian-independent light regulation of mammalian metabolism. Nat Metab 2024; 6:1000-1007. [PMID: 38831000 DOI: 10.1038/s42255-024-01051-6] [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] [Received: 11/15/2023] [Accepted: 04/16/2024] [Indexed: 06/05/2024]
Abstract
The daily light-dark cycle is a key zeitgeber (time cue) for entraining an organism's biological clock, whereby light sensing by retinal photoreceptors, particularly intrinsically photosensitive retinal ganglion cells, stimulates the suprachiasmatic nucleus of the hypothalamus, a central pacemaker that in turn orchestrates the rhythm of peripheral metabolic activities. Non-rhythmic effects of light on metabolism have also been long known, and their transduction mechanisms are only beginning to unfold. Here, we summarize emerging evidence that, in mammals, light exposure or deprivation profoundly affects glucose homeostasis, thermogenesis and other metabolic activities in a clock-independent manner. Such light regulation could involve melanopsin-based, intrinsically photosensitive retinal ganglion cell-initiated brain circuits via the suprachiasmatic nucleus of the hypothalamus and other nuclei, or direct stimulation of opsins expressed in the hypothalamus, adipose tissue, blood vessels and skin to regulate sympathetic tone, lipolysis, glucose uptake, mitochondrial activation, thermogenesis, food intake, blood pressure and melanogenesis. These photic signalling events may coordinate with circadian-based mechanisms to maintain metabolic homeostasis, with dysregulation of this system underlying metabolic diseases caused by aberrant light exposure, such as environmental night light and shift work.
Collapse
Affiliation(s)
- Feng Rao
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Tian Xue
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
8
|
Bohmke NJ, Dixon DL, Kirkman DL. Chrono-nutrition for hypertension. Diabetes Metab Res Rev 2024; 40:e3760. [PMID: 38287721 DOI: 10.1002/dmrr.3760] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 10/27/2023] [Accepted: 11/15/2023] [Indexed: 01/31/2024]
Abstract
Despite the advancement in blood pressure (BP) lowering medications, uncontrolled hypertension persists, underscoring a stagnation of effective clinical strategies. Novel and effective lifestyle therapies are needed to prevent and manage hypertension to mitigate future progression to cardiovascular and chronic kidney diseases. Chrono-nutrition, aligning the timing of eating with environmental cues and internal biological clocks, has emerged as a potential strategy to improve BP in high-risk populations. The aim of this review is to provide an overview of the circadian physiology of BP with an emphasis on renal and vascular circadian biology. The potential of Chrono-nutrition as a lifestyle intervention for hypertension is discussed and current evidence for the efficacy of time-restricted eating is presented.
Collapse
Affiliation(s)
- Natalie J Bohmke
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Dave L Dixon
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Danielle L Kirkman
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
9
|
Méndez-Hernández R, Rumanova VS, Guzmán-Ruiz MA, Foppen E, Moreno-Morton R, Hurtado-Alvarado G, Escobar C, Kalsbeek A, Buijs RM. Minor Changes in Daily Rhythms Induced by a Skeleton Photoperiod Are Associated with Increased Adiposity and Glucose Intolerance. Adv Biol (Weinh) 2023; 7:e2200116. [PMID: 35818679 DOI: 10.1002/adbi.202200116] [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: 04/19/2022] [Revised: 06/24/2022] [Indexed: 11/22/2023]
Abstract
Eating during the rest phase is associated with metabolic syndrome, proposed to result from a conflict between food consumption and the energy-saving state imposed by the circadian system. However, in nocturnal rodents, eating during the rest phase (day-feeding, DF) also implies food intake during light exposure. To investigate whether light exposure contributes to DF-induced metabolic impairments, animals receive food during the subjective day without light. A skeleton photoperiod (SP) is used to entrain rats to a 12:12 cycle with two short light pulses framing the subjective day. DF-induced adiposity is prevented by SP, suggesting that the conflict between light and feeding stimulates fat accumulation. However, all animals under SP conditions develop glucose intolerance regardless of their feeding schedule. Moreover, animals under SP with ad libitum or night-feeding have increased adiposity. SP animals show a delayed onset of the daily rise in body temperature and energy expenditure and shorter duration of nighttime activity, which may contribute to the metabolic disturbances. These data emphasize that metabolic homeostasis can only be achieved when all daily cycling variables are synchronized. Even small shifts in the alignment of different metabolic rhythms, such as those induced by SP, may predispose individuals to metabolic disease.
Collapse
Affiliation(s)
- Rebeca Méndez-Hernández
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, Mexico City, 04510, Mexico
| | - Valentina S Rumanova
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Ilkovičova ulica č. 6, Bratislava, 842 15, Slovakia
- Netherlands Institute for Neuroscience (NIN), Meibergdreef 47, Amsterdam, 1105 BA, The Netherlands
| | - Mara A Guzmán-Ruiz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, Mexico City, 04510, Mexico
| | - Ewout Foppen
- Netherlands Institute for Neuroscience (NIN), Meibergdreef 47, Amsterdam, 1105 BA, The Netherlands
- Laboratory of Endocrinology, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Rodrigo Moreno-Morton
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, Mexico City, 04510, Mexico
| | - Gabriela Hurtado-Alvarado
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, Mexico City, 04510, Mexico
| | - Carolina Escobar
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, Mexico City, 04510, Mexico
| | - Andries Kalsbeek
- Netherlands Institute for Neuroscience (NIN), Meibergdreef 47, Amsterdam, 1105 BA, The Netherlands
- Laboratory of Endocrinology, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Ruud M Buijs
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, Mexico City, 04510, Mexico
| |
Collapse
|
10
|
Meléndez-Fernández OH, Walton JC, DeVries AC, Nelson RJ. The role of daylight exposure on body mass in male mice. Physiol Behav 2023; 266:114186. [PMID: 37028499 PMCID: PMC10225047 DOI: 10.1016/j.physbeh.2023.114186] [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: 02/21/2023] [Revised: 03/25/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023]
Abstract
Physiology and behavior are synchronized to the external environment by endogenous circadian rhythms that are set to precisely 24 h by exposure to bright light early in the day. Exposure to artificial light outside of the typical solar day, such as during the night, may impair aspects of physiology and behavior in human and non-human animals. Both the intensity and the wavelength of light are important in mediating these effects. The present report is the result of an unplanned change in our vivarium lighting conditions, which led to the observation that dim light during the daytime affects body mass similarly to dim nighttime light exposure in male Swiss Webster mice. Mice exposed to bright days (≥125 lux) with dark nights (0 lux) gained significantly less weight than those exposed to bright days with dim light at night (5 lux) or dim days (≤60 lux) with either dark nights or dim light at night. Notably, among the mice exposed to dim daytime light, no weight gain differences were observed between dark nights and dim light at night exposure; however dim light at night exposure shifted food intake to the inactive phase as previously reported. The mechanisms mediating these effects remain unspecified, but it appears that dimly illuminated days may have similar adverse metabolic effects as exposure to artificial light at night.
Collapse
Affiliation(s)
| | - James C Walton
- Department of Neuroscience, Rockefeller Neuroscience Institute, Morgantown, WV 26505 USA
| | - A Courtney DeVries
- Department of Neuroscience, Rockefeller Neuroscience Institute, Morgantown, WV 26505 USA; Department of Medicine, Division of Oncology/Hematology, Morgantown, WV 26505 USA; West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26505, USA
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, Morgantown, WV 26505 USA
| |
Collapse
|
11
|
Young IE, Poobalan A, Steinbeck K, O'Connor HT, Parker HM. Distribution of energy intake across the day and weight loss: A systematic review and meta-analysis. Obes Rev 2023; 24:e13537. [PMID: 36530130 PMCID: PMC10078448 DOI: 10.1111/obr.13537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/27/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022]
Abstract
Consuming a greater proportion of total energy intake earlier in the day rather than in the evening is proposed to positively influence weight loss and health, potentially due to greater synchronization of human body circadian rhythms. This systematic review provides an update on existing evidence regarding earlier distributed eating patterns in weight loss interventions. Using a robust search strategy in five electronic databases, nine randomized controlled trials investigating the impact of energy intake distribution on weight loss were identified. Following critical appraisal, a random-effects meta-analyses found that, in the context of an energy-reduced diet, distributing energy intake with a focus on earlier intake resulted in significantly greater weight loss (-1.23 kg; 95% CI 2.40, -0.06, p = 0.04). Improvements in HOMA-IR, fasting glucose, and LDL cholesterol were also seen. The current study provides a timely update on the evidence linking distribution of total daily energy intake and health, showing that a focus on earlier intakes can result in greater short-term weight loss compared with later intakes. Future studies are needed to elucidate the impact that earlier intakes may have on weight management and metabolic health.
Collapse
Affiliation(s)
- Isabel E Young
- Faculty of Medicine and Health, School of Health Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Amudha Poobalan
- School of Medicine, Medical Sciences and Nutrition, The University of Aberdeen, Aberdeen, UK
| | - Katharine Steinbeck
- Faculty of Medicine and Health, Specialty of Child and Adolescent Health, Clinical School at The Children's Hospital Westmead, The University of Sydney, Sydney, New South Wales, Australia.,The Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Helen T O'Connor
- Faculty of Medicine and Health, School of Health Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Helen M Parker
- Faculty of Medicine and Health, School of Health Science, The University of Sydney, Sydney, New South Wales, Australia.,The Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
12
|
Verma AK, Singh S, Rizvi SI. Aging, circadian disruption and neurodegeneration: Interesting interplay. Exp Gerontol 2023; 172:112076. [PMID: 36574855 DOI: 10.1016/j.exger.2022.112076] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 11/26/2022] [Accepted: 12/22/2022] [Indexed: 12/26/2022]
Abstract
The circadian system is an intricate molecular network of coordinating circadian clocks that organize the internal synchrony of the organism in response to the environment. These rhythms are maintained by genetically programmed positive and negative auto-regulated transcriptional and translational feedback loops that sustain 24-hour oscillations in mRNA and protein components of the endogenous circadian clock. Since inter and intracellular activity of the central pacemaker appears to reduce with aging, the interaction between the circadian clock and aging continues to elude our understanding. In this review article, we discuss circadian clock components at the molecular level and how aging adversely affects circadian clock functioning in rodents and humans. The natural decline in melatonin levels with aging strongly contributes to circadian dysregulation resulting in the development of neurological anomalies. Additionally, inappropriate environmental conditions such as Artificial Light at Night (ALAN) can cause circadian disruption or chronodisruption (CD) which can result in a variety of pathological diseases, including premature aging. Furthermore, we summarize recent evidence suggesting that CD may also be a predisposing factor for the development of age-related neurodegenerative diseases (NDDs) such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), although more investigation is required to prove this link. Finally, certain chrono-enhancement approaches have been offered as intervention strategies to prevent, alleviate, or mitigate the impacts of CD. This review thus aims to bring together recent advancements in the chronobiology of the aging process, as well as its role in NDDs.
Collapse
Affiliation(s)
- Avnish Kumar Verma
- Department of Biochemistry, University of Allahabad, Allahabad 211002, India
| | - Sandeep Singh
- Department of Biochemistry, University of Allahabad, Allahabad 211002, India; Psychedelics Research Group, Biological Psychiatry Laboratory and Hadassah BrainLabs, Hadassah Medical Center, Hebrew University, Jerusalem, Israel
| | - Syed Ibrahim Rizvi
- Department of Biochemistry, University of Allahabad, Allahabad 211002, India.
| |
Collapse
|
13
|
Moeller JS, Bever SR, Finn SL, Phumsatitpong C, Browne MF, Kriegsfeld LJ. Circadian Regulation of Hormonal Timing and the Pathophysiology of Circadian Dysregulation. Compr Physiol 2022; 12:4185-4214. [PMID: 36073751 DOI: 10.1002/cphy.c220018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Circadian rhythms are endogenously generated, daily patterns of behavior and physiology that are essential for optimal health and disease prevention. Disruptions to circadian timing are associated with a host of maladies, including metabolic disease and obesity, diabetes, heart disease, cancer, and mental health disturbances. The circadian timing system is hierarchically organized, with a master circadian clock located in the suprachiasmatic nucleus (SCN) of the anterior hypothalamus and subordinate clocks throughout the CNS and periphery. The SCN receives light information via a direct retinal pathway, synchronizing the master clock to environmental time. At the cellular level, circadian rhythms are ubiquitous, with rhythms generated by interlocking, autoregulatory transcription-translation feedback loops. At the level of the SCN, tight cellular coupling maintains rhythms even in the absence of environmental input. The SCN, in turn, communicates timing information via the autonomic nervous system and hormonal signaling. This signaling couples individual cellular oscillators at the tissue level in extra-SCN brain loci and the periphery and synchronizes subordinate clocks to external time. In the modern world, circadian disruption is widespread due to limited exposure to sunlight during the day, exposure to artificial light at night, and widespread use of light-emitting electronic devices, likely contributing to an increase in the prevalence, and the progression, of a host of disease states. The present overview focuses on the circadian control of endocrine secretions, the significance of rhythms within key endocrine axes for typical, homeostatic functioning, and implications for health and disease when dysregulated. © 2022 American Physiological Society. Compr Physiol 12: 1-30, 2022.
Collapse
Affiliation(s)
- Jacob S Moeller
- Graduate Group in Endocrinology, University of California, Berkeley, California, USA
| | - Savannah R Bever
- Department of Psychology, University of California, Berkeley, California, USA
| | - Samantha L Finn
- Department of Psychology, University of California, Berkeley, California, USA
| | | | - Madison F Browne
- Department of Psychology, University of California, Berkeley, California, USA
| | - Lance J Kriegsfeld
- Graduate Group in Endocrinology, University of California, Berkeley, California, USA.,Department of Psychology, University of California, Berkeley, California, USA.,Department of Integrative Biology, University of California, Berkeley, California, USA.,The Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA
| |
Collapse
|
14
|
Goncharova N, Chigarova O, Oganyan T. Age-related and individual features of the HPA axis stress responsiveness under constant light in nonhuman primates. Front Endocrinol (Lausanne) 2022; 13:1051882. [PMID: 36699023 PMCID: PMC9870316 DOI: 10.3389/fendo.2022.1051882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis is a key adaptive neuroendocrine system, dysfunction of which plays an important role in the increasing incidence of stress-dependent age-related pathology. Among the environmental factors effecting increase age-related diseases, great importance is given to disturbances of the light-dark schedule, particularly with increased illumination at night. While disruption of the light-dark schedule has long been recognized as a powerful behavioral stressor, little is known regarding stress reactivity of the HPA under constant light (CL) conditions, especially with aging and depending on the features of stress behavior. The purpose of this investigation was to study the age-related and individual features of the HPA axis response to acute stress exposure (ASE) under chronic CL in nonhuman primates that are known to differ in behavioral responsiveness to stress. Young and old female rhesus monkeys (with control standard behavior or anxiety and depression-like behavior) were exposed to CL (24 h light/day, 330-400 lux for 4 to 8 weeks). Control young and old monkeys were exposed to standard lighting (SL) with natural light during the day and darkness at night. All animals were subjected to ASE (restriction of mobility for 2 hours), functional tests with corticotrophin-releasing hormone and arginine-vasopressin, and study of circadian rhythms of cortisol and pineal melatonin secretion. For the first time an inhibitory effect of CL on the reaction of the adrenal cortex to ASE was revealed in all individuals, regardless of age and preexisting behavior stress reactivity, the mechanisms of which were age-dependent: due to inhibition of the pituitary ACTH secretion in young animals and mainly not affecting the ACTH secretion in old individuals. There were no significant changes in melatonin secretion both in young and old animals. The observed CL inhibition of adrenal cortical reactivity to ASE may be useful to correct increased vulnerability to ASE observed in individuals with preexisting anxiety and depression-like stress behaviors. On the other hand, the CL induced decrease in adrenal stress reactivity of behaviorally normal animals suggests a potential risk of reducing the adaptive capacity of the organism under conditions of continuous light exposure.
Collapse
|
15
|
Light at night disrupts biological clocks, calendars, and immune function. Semin Immunopathol 2021; 44:165-173. [PMID: 34731290 PMCID: PMC8564795 DOI: 10.1007/s00281-021-00899-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/13/2021] [Indexed: 12/15/2022]
Abstract
Light at night is a pervasive problem in our society; over 80% of the world’s population experiences significant light pollution. Exacerbating this issue is the reality that artificially lit outdoor areas are growing by 2.2% per year and continuously lit areas brighten by 2.2% each year due to the rapid growths in population and urbanization. Furthermore, the increase in the prevalence of night shift work and smart device usage contributes to the inescapable nature of artificial light at night (ALAN). Although previously assumed to be innocuous, ALAN has deleterious effects on the circadian system and circadian-regulated physiology, particularly immune function. Due to the relevance of ALAN to the general population, it is important to understand its roles in disrupting immune function. This review presents a synopsis of the effects of ALAN on circadian clocks and immune function. We delineate the role of ALAN in altering clock gene expression and suppressing melatonin. We review the effects of light at night on inflammation and the innate and adaptive immune systems in various species to demonstrate the wide range of ALAN consequences. Finally, we propose future directions to provide further clarity and expansion of the field.
Collapse
|
16
|
Vu CHV, Kawashima M, Nakamura W, Nakamura TJ, Tsubota K. Circadian clock regulates tear secretion in the lacrimal gland. Exp Eye Res 2021; 206:108524. [PMID: 33662354 DOI: 10.1016/j.exer.2021.108524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/12/2021] [Accepted: 02/23/2021] [Indexed: 11/28/2022]
Abstract
Although diurnal variations have been observed in tear film parameters in various species, the molecular mechanisms that control circadian tear secretion remain unclear. The aim of our study was to evaluate the role of clock genes in the lacrimal gland (LG) in regulation of tear secretion. Tear volume was measured by cotton thread test in core clock genes deficient (Cry1-/-Cry2-/--) mice which are behaviorally arrhythmic. Real-time quantitative RT-PCR was used to examine expression profiles of core clock genes in the LG including Per1, Per2, Per3, Clock, Bmal1. All experiments were performed under a 12 h of light and 12 h of darkness (LD) and constant dark (DD) conditions. Under both LD and DD conditions, diurnal and circadian rhythms were observed in tear secretion of wild-type mice with tear volume increased in the objective and subjective night while disruption in diurnal and circadian variations of tear secretion were found in Cry1-/-Cry2-/--mice. In wild-type mice, the expression level of major clock genes in the LG showed oscillatory patterns under both LD and DD conditions. In contrast, expression clock genes in the lacrimal gland of Cry1-/-Cry2-/-- mice showed complete loss of oscillation regardless of environmental light conditions. These findings confirmed the presence of diurnal and circadian rhythms of tear secretion and provided evidences supporting a critical role for the clock in the control of tear secretion.
Collapse
Affiliation(s)
- Chi Hoang Viet Vu
- Department of Ophthalmology, Keio University, School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Motoko Kawashima
- Department of Ophthalmology, Keio University, School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Wataru Nakamura
- Department of Oral Chrono-Physiology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Takahiro J Nakamura
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan.
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University, School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Tsubota Laboratory, Inc., 304 Toshin-ekimae Bldg., 34 Shinanomachi, Shinjuku-ku, Tokyo, 160-0016, Japan.
| |
Collapse
|
17
|
Chen R, Weitzner AS, McKennon LA, Fonken LK. Light at night during development in mice has modest effects on adulthood behavior and neuroimmune activation. Behav Brain Res 2021; 405:113171. [PMID: 33577883 DOI: 10.1016/j.bbr.2021.113171] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/01/2021] [Accepted: 02/04/2021] [Indexed: 11/15/2022]
Abstract
Exposure to light at night (LAN) can disrupt the circadian system, thereby altering neuroimmune reactivity and related behavior. Increased exposure to LAN affects people of all ages - and could have particularly detrimental effects during early-life and adolescence. Despite this, most research on the behavioral and physiological effects of LAN has been conducted in adult animals. Here we evaluated the effects of dim LAN during critical developmental windows on adulthood neuroimmune function and affective/sickness behaviors. Male and female C57BL/6 J mice were exposed to dim LAN [12:12 light (150 lx)/dim (15 lx) cycle] during early life (PND10-24) or adolescence (PND30-44) [control: 12:12 light (150 lx)/dark (0 lx) cycle]. Behaviors were assessed during juvenile (PND 42-44) and adult (PND60) periods. Contrary to our hypothesis, juvenile mice that were exposed to dim LAN did not exhibit changes in anxiety- or depressive-like behaviors. By adulthood, adolescent LAN-exposed female mice showed a modest anxiety-like phenotype in one behavioral task but not another. Adolescent LAN exposure also induced depressive-like behavior in a forced swim task in adulthood in both male and female mice. Additionally, developmental LAN exacerbated the hippocampal cytokine response (IL-1β) following peripheral LPS in female, but not male mice. These results suggest female mice may be more susceptible to developmental LAN than male mice: LAN female mice had a modest anxiety-like phenotype in adulthood, and upon LPS challenge, higher hippocampal IL-1β expression. Taken together, developmental LAN exposure in mice promotes a modest increase in susceptibility to anxiety- and depressive-like symptoms.
Collapse
Affiliation(s)
- Ruizhuo Chen
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX 78712, USA
| | - Aidan S Weitzner
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX 78712, USA
| | - Lara A McKennon
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX 78712, USA
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
18
|
Lim ASP. Diurnal and seasonal molecular rhythms in the human brain and their relation to Alzheimer disease. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:271-284. [PMID: 34225968 DOI: 10.1016/b978-0-12-819975-6.00017-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diurnal and seasonal rhythms influence many aspects of human physiology including brain function. Moreover, altered diurnal and seasonal behavioral and physiological rhythms have been linked to Alzheimer's disease and related dementias (ADRD). Understanding the molecular basis for these links may lead to identification of novel targets to mitigate the negative impact of normal and abnormal diurnal and seasonal rhythms on ADRD or to alleviate the adverse consequences of ADRD on normal diurnal and seasonal rhythms. Diurnally and seasonally rhythmic gene expression and epigenetic modification in the human neocortex may be a key mechanism underlying these links. This chapter will first review the observed epidemiological links between normal and abnormal diurnal and seasonal rhythmicity, cognitive impairment, and ADRD. Then it will review normal diurnal and seasonal rhythms of brain epigenetic modification and gene expression in model organisms. Finally, it will review evidence for diurnal and seasonal rhythms of epigenetic modification and gene expression the human brain in aging, Alzheimer's disease, and other brain disorders.
Collapse
Affiliation(s)
- Andrew S P Lim
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
19
|
Walker WH, Bumgarner JR, Walton JC, Liu JA, Meléndez-Fernández OH, Nelson RJ, DeVries AC. Light Pollution and Cancer. Int J Mol Sci 2020; 21:E9360. [PMID: 33302582 PMCID: PMC7764771 DOI: 10.3390/ijms21249360] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/04/2020] [Accepted: 12/06/2020] [Indexed: 01/03/2023] Open
Abstract
For many individuals in industrialized nations, the widespread adoption of electric lighting has dramatically affected the circadian organization of physiology and behavior. Although initially assumed to be innocuous, exposure to artificial light at night (ALAN) is associated with several disorders, including increased incidence of cancer, metabolic disorders, and mood disorders. Within this review, we present a brief overview of the molecular circadian clock system and the importance of maintaining fidelity to bright days and dark nights. We describe the interrelation between core clock genes and the cell cycle, as well as the contribution of clock genes to oncogenesis. Next, we review the clinical implications of disrupted circadian rhythms on cancer, followed by a section on the foundational science literature on the effects of light at night and cancer. Finally, we provide some strategies for mitigation of disrupted circadian rhythms to improve health.
Collapse
Affiliation(s)
- William H. Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA; (J.R.B.); (J.C.W.); (J.A.L.); (O.H.M.-F.); (R.J.N.); (A.C.D.)
| | - Jacob R. Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA; (J.R.B.); (J.C.W.); (J.A.L.); (O.H.M.-F.); (R.J.N.); (A.C.D.)
| | - James C. Walton
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA; (J.R.B.); (J.C.W.); (J.A.L.); (O.H.M.-F.); (R.J.N.); (A.C.D.)
| | - Jennifer A. Liu
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA; (J.R.B.); (J.C.W.); (J.A.L.); (O.H.M.-F.); (R.J.N.); (A.C.D.)
| | - O. Hecmarie Meléndez-Fernández
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA; (J.R.B.); (J.C.W.); (J.A.L.); (O.H.M.-F.); (R.J.N.); (A.C.D.)
| | - Randy J. Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA; (J.R.B.); (J.C.W.); (J.A.L.); (O.H.M.-F.); (R.J.N.); (A.C.D.)
| | - A. Courtney DeVries
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA; (J.R.B.); (J.C.W.); (J.A.L.); (O.H.M.-F.); (R.J.N.); (A.C.D.)
- Department of Medicine, Division of Oncology/Hematology, West Virginia University, Morgantown, WV 26506, USA
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
20
|
Masís-Vargas A, Hicks D, Kalsbeek A, Mendoza J. Blue light at night acutely impairs glucose tolerance and increases sugar intake in the diurnal rodent Arvicanthis ansorgei in a sex-dependent manner. Physiol Rep 2020; 7:e14257. [PMID: 31646762 PMCID: PMC6811685 DOI: 10.14814/phy2.14257] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022] Open
Abstract
In our modern society, the exposure to light at night (LAN) has increased considerably, which may impact human health negatively. Especially exposure to light at night containing short wavelength emissions (~450–500 nm) can disrupt the normal function of the biological clock, altering sleep‐wake cycles and inducing metabolic changes. Recently, we reported that light at night acutely impairs glucose tolerance in nocturnal rats. However, light at night in nocturnal rodents coincides with their activity period, in contrast to artificial light at night exposure in humans. The aim of this study was to evaluate the acute effects of blue (λ = 490 ± 20 nm) artificial light at night (bALAN) on glucose metabolism and food intake in both male and female diurnal Sudanian grass rats (Arvicanthis ansorgei) fed either regular chow or a free choice high‐fat high sucrose diet (HFHS). In both chow and HFHS fed male Arvicanthis, 1‐hour of bALAN exposure induced a higher glucose response in the oral glucose tolerance test (OGTT) accompanied by a significant decrease in plasma insulin. Furthermore, in HFHS fed animals, bALAN induced an increase in sucrose intake during the dark phase in males but not in females. Additionally, 1‐h of bALAN increased the nonfasted glucose levels together with plasma corticosterone in female grass rats. These results provide new and further evidence for the deleterious effects of exposure to short wavelength emission‐containing artificial light at night on glucose metabolism in a diurnal rodent in a sex‐dependent manner.
Collapse
Affiliation(s)
- Anayanci Masís-Vargas
- Institute of Cellular and Integrative Neurosciences (INCI), UPR-3212 CNRS, University of Strasbourg, Strasbourg, France.,Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - David Hicks
- Institute of Cellular and Integrative Neurosciences (INCI), UPR-3212 CNRS, University of Strasbourg, Strasbourg, France
| | - Andries Kalsbeek
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jorge Mendoza
- Institute of Cellular and Integrative Neurosciences (INCI), UPR-3212 CNRS, University of Strasbourg, Strasbourg, France
| |
Collapse
|
21
|
Masís‐Vargas A, Ritsema WI, Mendoza J, Kalsbeek A. Metabolic Effects of Light at Night are Time- and Wavelength-Dependent in Rats. Obesity (Silver Spring) 2020; 28 Suppl 1:S114-S125. [PMID: 32700824 PMCID: PMC7497257 DOI: 10.1002/oby.22874] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Intrinsically photosensitive retinal ganglion cells are most sensitive to short wavelengths and reach brain regions that modulate biological rhythms and energy metabolism. The increased exposure nowadays to artificial light at night (ALAN), especially short wavelengths, perturbs our synchronization with the 24-hour solar cycle. Here, the time- and wavelength dependence of the metabolic effects of ALAN are investigated. METHODS Male Wistar rats were exposed to white, blue, or green light at different time points during the dark phase. Locomotor activity, energy expenditure, respiratory exchange ratio (RER), and food intake were recorded. Brains, livers, and blood were collected. RESULTS All wavelengths decreased locomotor activity regardless of time of exposure, but changes in energy expenditure were dependent on the time of exposure. Blue and green light reduced RER at Zeitgeber time 16-18 without changing food intake. Blue light increased period 1 (Per1) gene expression in the liver, while green and white light increased Per2. Blue light decreased plasma glucose and phosphoenolpyruvate carboxykinase (Pepck) expression in the liver. All wavelengths increased c-Fos activity in the suprachiasmatic nucleus, but blue and green light decreased c-Fos activity in the paraventricular nucleus. CONCLUSIONS ALAN affects locomotor activity, energy expenditure, RER, hypothalamic c-Fos expression, and expression of clock and metabolic genes in the liver depending on the time of day and wavelength.
Collapse
Affiliation(s)
- Anayanci Masís‐Vargas
- Department of Endocrinology and MetabolismAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Hypothalamic Integration MechanismsNetherlands Institute for Neuroscience (NIN)AmsterdamThe Netherlands
- Institute of Cellular and Integrative Neurosciences (INCI)UPR‐3212 CNRSUniversity of StrasbourgStrasbourgFrance
| | - Wayne I.G.R. Ritsema
- Department of Endocrinology and MetabolismAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Hypothalamic Integration MechanismsNetherlands Institute for Neuroscience (NIN)AmsterdamThe Netherlands
| | - Jorge Mendoza
- Institute of Cellular and Integrative Neurosciences (INCI)UPR‐3212 CNRSUniversity of StrasbourgStrasbourgFrance
| | - Andries Kalsbeek
- Department of Endocrinology and MetabolismAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Hypothalamic Integration MechanismsNetherlands Institute for Neuroscience (NIN)AmsterdamThe Netherlands
| |
Collapse
|
22
|
Fleury G, Masís‐Vargas A, Kalsbeek A. Metabolic Implications of Exposure to Light at Night: Lessons from Animal and Human Studies. Obesity (Silver Spring) 2020; 28 Suppl 1:S18-S28. [PMID: 32700826 PMCID: PMC7497102 DOI: 10.1002/oby.22807] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/13/2020] [Accepted: 03/14/2020] [Indexed: 02/06/2023]
Abstract
Lately, the incidence of overweight, obesity, and type 2 diabetes has shown a staggering increase. To prevent and treat these conditions, one must look at their etiology. As life on earth has evolved under the conditions of nature's 24-hour light/dark cycle, it seems likely that exposure to artificial light at night (LAN) would affect physiology. Indeed, ample evidence has shown that LAN impacts many metabolic parameters, at least partly via the biological clock in the suprachiasmatic nucleus of the hypothalamus. This review focuses on the impact of chronic and acute effects of LAN of different wavelengths on locomotor activity, food intake, the sleep/wake cycle, body temperature, melatonin, glucocorticoids, and glucose and lipid metabolism. While chronic LAN disturbs daily rhythms in these parameters, experiments using short-term LAN exposure also have shown acute negative effects in metabolically active peripheral tissues. Experiments using LAN of different wavelengths not only have indicated an important role for melanopsin, the photopigment found in intrinsically photosensitive retinal ganglion cells, but also provided evidence that each wavelength may have a specific impact on energy metabolism. Importantly, exposure to LAN has been shown to impact glucose homeostasis also in humans and to be associated with an increased incidence of overweight, obesity, and atherosclerosis.
Collapse
Affiliation(s)
- Giulia Fleury
- Department of Endocrinology and MetabolismAmsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - Anayanci Masís‐Vargas
- Department of Endocrinology and MetabolismAmsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
- Hypothalamic Integration MechanismsNetherlands Institute for Neuroscience (NIN)Amsterdamthe Netherlands
- Institute of Cellular and Integrative Neurosciences (INCI)UPR‐3212 CNRSUniversity of StrasbourgStrasbourgFrance
| | - Andries Kalsbeek
- Department of Endocrinology and MetabolismAmsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
- Hypothalamic Integration MechanismsNetherlands Institute for Neuroscience (NIN)Amsterdamthe Netherlands
| |
Collapse
|
23
|
Angelousi A, Nasiri-Ansari N, Karapanagioti A, Kyriakopoulos G, Aggeli C, Zografos G, Choreftaki T, Parianos C, Kounadi T, Alexandraki K, Randeva HS, Kaltsas G, Papavassiliou AG, Kassi E. Expression of clock-related genes in benign and malignant adrenal tumors. Endocrine 2020; 68:650-659. [PMID: 32147772 DOI: 10.1007/s12020-020-02246-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 02/26/2020] [Indexed: 01/03/2023]
Abstract
Although the effect of the central clock system on adrenal function has been extensively studied, the role of the peripheral clock system in adrenal tumorigenesis remains largely unexplored. In this study we investigated the expression of clock-related genes in normal adrenocortical tissue and adrenocortical tumors. Twenty-seven fresh frozen human adrenal tissues including 13 cortisol secreting adenomas (CSA), seven aldosterone producing adenomas (APA), and seven adrenocortical carcinomas (ACC) were collected. CLOCK, BMAL1, PER1, CRY1, Rev-ERB, and RORα mRNA and protein expression were determined by qPCR and immunoblotting in pathological tissues and compared with the adjacent normal adrenal tissues. A significant downregulation of PER1, CRY1, and Rev-ERB compared with their normal tissue was demonstrated in CSA. All clock-related genes were overexpressed in APA compared with their normal tissue, albeit not significantly. A significant upregulation of CRY1 and PER1 and downregulation of BMAL1, RORα, and Rev-ERB compared with normal adrenal tissue was observed in ACC. BMAL1 and PER1 were significantly downregulated in APA compared with CSA. CLOCK, CRY1, and PER1 were upregulated, whereas BMAL1, RORα, and Rev-ERB were downregulated in ACC compared with CSA. Our study demonstrated the expression of CLOCK, BMAL1, PER1, CRY1, Rev-ERB, and RORα in normal and pathological human adrenal tissues. Adrenal tumors exhibited altered expression of these genes compared with normal tissue, with specific differences between benign and malignant lesions and between benign tumors arising from glomerulosa vs fasciculata zone. Further studies should clarify whether these alterations could be implicated in adrenocortical tumorigenesis.
Collapse
Affiliation(s)
- Anna Angelousi
- 1st Department of Internal Medicine, Laiko University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Narjes Nasiri-Ansari
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece
| | - Angeliki Karapanagioti
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece
| | - Georgios Kyriakopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece
| | - Chrysanthi Aggeli
- 3rd Department of Surgery, General Hospital of Athens "G. Gennimatas", Athens, Greece
| | - Giorgos Zografos
- 3rd Department of Surgery, General Hospital of Athens "G. Gennimatas", Athens, Greece
| | - Theodosia Choreftaki
- Department of Pathology, General Hospital of Athens "G. Gennimatas", Athens, Greece
| | - Christos Parianos
- 3rd Department of Surgery, General Hospital of Athens "G. Gennimatas", Athens, Greece
| | - Theodora Kounadi
- Department of Endocrinology and Diabetes Center, Athens General Hospital "G. Gennimatas", Athens, Greece
| | - Krystallenia Alexandraki
- 1st Department of Propaedeutic Internal Medicine, Laiko University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Harpal S Randeva
- Division of Translational and Experimental Medicine, Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Gregory Kaltsas
- 1st Department of Propaedeutic Internal Medicine, Laiko University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece
| | - Eva Kassi
- 1st Department of Internal Medicine, Laiko University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece.
| |
Collapse
|
24
|
Benitah SA, Welz PS. Circadian Regulation of Adult Stem Cell Homeostasis and Aging. Cell Stem Cell 2020; 26:817-831. [DOI: 10.1016/j.stem.2020.05.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
25
|
Fatima N, Rana S. Metabolic implications of circadian disruption. Pflugers Arch 2020; 472:513-526. [PMID: 32363530 DOI: 10.1007/s00424-020-02381-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 01/20/2023]
Abstract
Circadian rhythms are generated by the circadian clock, a self-sustained internal timing system that exhibits 24-h rhythms in the body. In mammals, circadian rhythms are driven by a central clock located in suprachiasmatic nucleus and various peripheral clocks located in different tissues and organs of the body. Many cellular, behavioral, and physiological processes are regulated by the circadian clock in coordination with environmental cues. The process of metabolism is also under circadian regulation. Loss of synchronization between the internal clock and environmental zeitgebers results in disruption of the circadian rhythms that seriously impacts metabolic homeostasis leading to changed eating behavior, altered glucose and lipid metabolism, and weight gain. This in turn augments the risk of having various cardio-metabolic disorders such as obesity, diabetes, metabolic syndrome, and cardiovascular disease. This review sheds light on circadian rhythms and their role in metabolism with the identification of gaps in the current knowledge that remain to be explored in these fields. In this review, the molecular mechanisms underlying circadian rhythms have been elaborated first. Then, the focus has been kept on explaining the physiological significance of circadian rhythms in regulating metabolism. Finally, the implications for metabolism when these rhythms are disrupted due to genetic mutations or social and occupational needs enforced by modern lifestyle have been discussed.
Collapse
Affiliation(s)
- Narjis Fatima
- Molecular Biology and Human Genetics Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan
| | - Sobia Rana
- Molecular Biology and Human Genetics Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
26
|
Welz PS, Benitah SA. Molecular Connections Between Circadian Clocks and Aging. J Mol Biol 2019; 432:3661-3679. [PMID: 31887285 DOI: 10.1016/j.jmb.2019.12.036] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 12/25/2022]
Abstract
The mammalian circadian clockwork has evolved as a timing system that allows the daily environmental changes to be anticipated so that behavior and tissue physiology can be adjusted accordingly. The circadian clock synchronizes the function of all cells within tissues in order to temporally separate preclusive and potentially harmful physiologic processes and to establish a coherent temporal organismal physiology. Thus, the proper functioning of the circadian clockwork is essential for maintaining cellular and tissue homeostasis. Importantly, aging reduces the robustness of the circadian clock, resulting in disturbed sleep-wake cycles, a lowered capacity to synchronize circadian rhythms in peripheral tissues, and reprogramming of the circadian clock output at the molecular function levels. These circadian clock-dependent behavioral and molecular changes in turn further accelerate the process of aging. Here we review the current knowledge about how aging affects the circadian clock, how the functional decline of the circadian clock affects aging, and how the circadian clock machinery and the molecular processes that underlie aging are intertwined.
Collapse
Affiliation(s)
- Patrick-Simon Welz
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - S A Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; ICREA, Catalan Institution for Research and Advanced Studies, Barcelona, Spain.
| |
Collapse
|
27
|
Heidari MH, Zamanian Azodi M, Zali MR, Akbari Z. Light at Night Exposure Effects on Differentiation and Cell Cycle in the Rat Liver With Autonomic Nervous System Denervation. J Lasers Med Sci 2019; 10:S43-S48. [PMID: 32021672 PMCID: PMC6983860 DOI: 10.15171/jlms.2019.s8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: N Exposure to artificial light at night (LAN) affects human health and causes several functional modifications in the body. Obesity, diabetes, and hormonal changes are reported after exposure to LAN in humans. This study aims to highlight the critical features of biological terms that are affected in the liver of rats which received autonomic nervous system denervation. Methods: The liver gene expression profiles of 8 male Wistar rats that received sympathetic plus parasympathetic hepatic denervation and were exposed to LAN from Gene Expression Omnibus (GEO) for 1 hour were compared with 5 controls. The significant differentially-expressed genes (DEGs) were screened by the protein-protein interaction (PPI) network analysis STRING database (an application of Cytoscape software). Also, CuleGO and CleuDedia, the 2 applications of Cytoscape software, were used for more analysis. Results: Among 250 DEGs, 173 characterized genes with fold change more than 2 plus 100 added relevant genes were included in the PPI network. The analysis of the main connected component (MCC) led to introducing 15 hubs and 15 bottlenecks. CCT2, COPS7A, KAT2A, and ERCC1 were determined as hub-bottlenecks. Among hubs and bottlenecks, DHX15, KAT2A, CCT2, HSP90AB1, CCNE1, DHX16, LSM2, WEE1, CWC27, BAZ1B, RAB22A, DNM2, and DHX30 were linked to each other by various kinds of actions. CCT2 and KAT2A, the 2 hub-bottlenecks, were included in the interacted genes in the action map. Four classes of biological terms including negative regulation of non-motile cilium assembly, negative regulation of transforming growth factor beta activation, alpha-tubulin acetylation, and histamine-induced gastric acid secretion were identified as the critical biochemical pathways and biological processes. Conclusion: Several essential functions such as differentiation, cell cycle, ribosome assembly, and splicing are affected by LAN in rat livers with autonomic nervous system denervation.
Collapse
Affiliation(s)
- Mohammad Hossein Heidari
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Zamanian Azodi
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Akbari
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Rumanova VS, Okuliarova M, Molcan L, Sutovska H, Zeman M. Consequences of low-intensity light at night on cardiovascular and metabolic parameters in spontaneously hypertensive rats. Can J Physiol Pharmacol 2019; 97:863-871. [DOI: 10.1139/cjpp-2019-0043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Circadian rhythms are an inherent property of physiological processes and can be disturbed by irregular environmental cycles, including artificial light at night (ALAN). Circadian disruption may contribute to many pathologies, such as hypertension, obesity, and type 2 diabetes, but the underlying mechanisms are not understood. Our study investigated the consequences of ALAN on cardiovascular and metabolic parameters in spontaneously hypertensive rats, which represent an animal model of essential hypertension and insulin resistance. Adult males were exposed to a 12 h light − 12 h dark cycle and the ALAN group experienced dim light at night (1–2 lx), either for 2 or 5 weeks. Rats on ALAN showed a loss of light–dark variability for systolic blood pressure, but not for heart rate. Moreover, a gradual increase of systolic blood pressure was recorded over 5 weeks of ALAN. Exposure to ALAN increased plasma insulin and hepatic triglyceride levels. An increased expression of metabolic transcription factors, Pparα and Pparγ, in the epididymal fat and a decreased expression of Glut4 in the heart was found in the ALAN group. Our results demonstrate that low-intensity ALAN can disturb blood pressure control and augment insulin resistance in spontaneously hypertensive rats, and may represent a serious risk factor for cardiometabolic diseases.
Collapse
Affiliation(s)
- Valentina Sophia Rumanova
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Monika Okuliarova
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Lubos Molcan
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Hana Sutovska
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Michal Zeman
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| |
Collapse
|
29
|
de Juan A, Ince LM, Pick R, Chen CS, Molica F, Zuchtriegel G, Wang C, Zhang D, Druzd D, Hessenauer MET, Pelli G, Kolbe I, Oster H, Prophete C, Hergenhan SM, Albrecht U, Ripperger J, Montanez E, Reichel CA, Soehnlein O, Kwak BR, Frenette PS, Scheiermann C. Artery-Associated Sympathetic Innervation Drives Rhythmic Vascular Inflammation of Arteries and Veins. Circulation 2019; 140:1100-1114. [PMID: 31401849 DOI: 10.1161/circulationaha.119.040232] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The incidence of acute cardiovascular complications is highly time-of-day dependent. However, the mechanisms driving rhythmicity of ischemic vascular events are unknown. Although enhanced numbers of leukocytes have been linked to an increased risk of cardiovascular complications, the role that rhythmic leukocyte adhesion plays in different vascular beds has not been studied. METHODS We evaluated leukocyte recruitment in vivo by using real-time multichannel fluorescence intravital microscopy of a tumor necrosis factor-α-induced acute inflammation model in both murine arterial and venous macrovasculature and microvasculature. These approaches were complemented with genetic, surgical, and pharmacological ablation of sympathetic nerves or adrenergic receptors to assess their relevance for rhythmic leukocyte adhesion. In addition, we genetically targeted the key circadian clock gene Bmal1 (also known as Arntl) in a lineage-specific manner to dissect the importance of oscillations in leukocytes and components of the vessel wall in this process. RESULTS In vivo quantitative imaging analyses of acute inflammation revealed a 24-hour rhythm in leukocyte recruitment to arteries and veins of the mouse macrovasculature and microvasculature. Unexpectedly, although in arteries leukocyte adhesion was highest in the morning, it peaked at night in veins. This phase shift was governed by a rhythmic microenvironment and a vessel type-specific oscillatory pattern in the expression of promigratory molecules. Differences in cell adhesion molecules and leukocyte adhesion were ablated when disrupting sympathetic nerves, demonstrating their critical role in this process and the importance of β2-adrenergic receptor signaling. Loss of the core clock gene Bmal1 in leukocytes, endothelial cells, or arterial mural cells affected the oscillations in a vessel type-specific manner. Rhythmicity in the intravascular reactivity of adherent leukocytes resulted in increased interactions with platelets in the morning in arteries and in veins at night with a higher predisposition to acute thrombosis at different times as a consequence. CONCLUSIONS Together, our findings point to an important and previously unrecognized role of artery-associated sympathetic innervation in governing rhythmicity in vascular inflammation in both arteries and veins and its potential implications in the occurrence of time-of-day-dependent vessel type-specific thrombotic events.
Collapse
Affiliation(s)
- Alba de Juan
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, BioMedical Centre, Planegg-Martinsried, Germany (A.d.J., L.M.I., R.P., C.-S.C., G.Z., D.D., M.E.T.H., S.M.H., E.M., C.A.R., C.S.)
| | - Louise Madeleine Ince
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, BioMedical Centre, Planegg-Martinsried, Germany (A.d.J., L.M.I., R.P., C.-S.C., G.Z., D.D., M.E.T.H., S.M.H., E.M., C.A.R., C.S.).,University of Geneva, Centre Médical Universitaire (CMU), Department of Pathology and Immunology, Switzerland (L.M.I., F.M., C.W., G.P., B.R. K., C.S.)
| | - Robert Pick
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, BioMedical Centre, Planegg-Martinsried, Germany (A.d.J., L.M.I., R.P., C.-S.C., G.Z., D.D., M.E.T.H., S.M.H., E.M., C.A.R., C.S.)
| | - Chien-Sin Chen
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, BioMedical Centre, Planegg-Martinsried, Germany (A.d.J., L.M.I., R.P., C.-S.C., G.Z., D.D., M.E.T.H., S.M.H., E.M., C.A.R., C.S.)
| | - Filippo Molica
- University of Geneva, Centre Médical Universitaire (CMU), Department of Pathology and Immunology, Switzerland (L.M.I., F.M., C.W., G.P., B.R. K., C.S.)
| | - Gabriele Zuchtriegel
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, BioMedical Centre, Planegg-Martinsried, Germany (A.d.J., L.M.I., R.P., C.-S.C., G.Z., D.D., M.E.T.H., S.M.H., E.M., C.A.R., C.S.)
| | - Chen Wang
- University of Geneva, Centre Médical Universitaire (CMU), Department of Pathology and Immunology, Switzerland (L.M.I., F.M., C.W., G.P., B.R. K., C.S.)
| | - Dachuan Zhang
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research and Department of Cell Biology, Albert Einstein College of Medicine, New York (D.Z., C.P., P.S.F.)
| | - David Druzd
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, BioMedical Centre, Planegg-Martinsried, Germany (A.d.J., L.M.I., R.P., C.-S.C., G.Z., D.D., M.E.T.H., S.M.H., E.M., C.A.R., C.S.)
| | - Maximilian E T Hessenauer
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, BioMedical Centre, Planegg-Martinsried, Germany (A.d.J., L.M.I., R.P., C.-S.C., G.Z., D.D., M.E.T.H., S.M.H., E.M., C.A.R., C.S.)
| | - Graziano Pelli
- University of Geneva, Centre Médical Universitaire (CMU), Department of Pathology and Immunology, Switzerland (L.M.I., F.M., C.W., G.P., B.R. K., C.S.)
| | - Isa Kolbe
- Institute of Neurobiology, University of Lübeck, Germany (I.K., H.O.)
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Germany (I.K., H.O.)
| | - Colette Prophete
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research and Department of Cell Biology, Albert Einstein College of Medicine, New York (D.Z., C.P., P.S.F.)
| | - Sophia Martina Hergenhan
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, BioMedical Centre, Planegg-Martinsried, Germany (A.d.J., L.M.I., R.P., C.-S.C., G.Z., D.D., M.E.T.H., S.M.H., E.M., C.A.R., C.S.)
| | - Urs Albrecht
- University of Freiburg, Switzerland (U.A., J.R.)
| | | | - Eloi Montanez
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, BioMedical Centre, Planegg-Martinsried, Germany (A.d.J., L.M.I., R.P., C.-S.C., G.Z., D.D., M.E.T.H., S.M.H., E.M., C.A.R., C.S.)
| | - Christoph A Reichel
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, BioMedical Centre, Planegg-Martinsried, Germany (A.d.J., L.M.I., R.P., C.-S.C., G.Z., D.D., M.E.T.H., S.M.H., E.M., C.A.R., C.S.)
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University, Munich, Germany (O.S.).,Department of Physiology and Pharmacology (FyFa) and Department of Medicine, Karolinska Institutet, Stockholm, Sweden (O.S.).,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (O.S., C.S.)
| | - Brenda R Kwak
- University of Geneva, Centre Médical Universitaire (CMU), Department of Pathology and Immunology, Switzerland (L.M.I., F.M., C.W., G.P., B.R. K., C.S.)
| | - Paul S Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research and Department of Cell Biology, Albert Einstein College of Medicine, New York (D.Z., C.P., P.S.F.)
| | - Christoph Scheiermann
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, BioMedical Centre, Planegg-Martinsried, Germany (A.d.J., L.M.I., R.P., C.-S.C., G.Z., D.D., M.E.T.H., S.M.H., E.M., C.A.R., C.S.).,University of Geneva, Centre Médical Universitaire (CMU), Department of Pathology and Immunology, Switzerland (L.M.I., F.M., C.W., G.P., B.R. K., C.S.).,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (O.S., C.S.)
| |
Collapse
|
30
|
Opperhuizen AL, Foppen E, Jonker M, Wackers P, van Faassen M, van Weeghel M, van Kerkhof L, Fliers E, Kalsbeek A. Effects of Light-at-Night on the Rat Liver - A Role for the Autonomic Nervous System. Front Neurosci 2019; 13:647. [PMID: 31281239 PMCID: PMC6596368 DOI: 10.3389/fnins.2019.00647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/05/2019] [Indexed: 12/31/2022] Open
Abstract
Exposure to light at night (LAN) has been associated with serious pathologies, including obesity, diabetes and cancer. Recently we showed that 2 h of LAN impaired glucose tolerance in rats. Several studies have suggested that the autonomic nervous system (ANS) plays an important role in communicating these acute effects of LAN to the periphery. Here, we investigated the acute effects of LAN on the liver transcriptome of male Wistar rats. Expression levels of individual genes were not markedly affected by LAN, nevertheless pathway analysis revealed clustered changes in a number of endocrine pathways. Subsequently, we used selective hepatic denervations [sympathetic (Sx), parasympathetic (Px), total (Tx, i.e., Sx plus Px), sham] to investigate the involvement of the ANS in the effects observed. Surgical removal of the sympathetic or parasympathetic hepatic branches of the ANS resulted in many, but small changes in the liver transcriptome, including a pathway involved with circadian clock regulation, but it clearly separated the four denervation groups. On the other hand, analysis of the liver metabolome was not able to separate the denervation groups, and only 6 out of 78 metabolites were significantly up- or downregulated after denervations. Finally, removal of the sympathetic and parasympathetic hepatic nerves combined with LAN exposure clearly modulated the effects of LAN on the liver transcriptome, but left most endocrine pathways unaffected. Conclusion: One-hour light-at-night acutely affects the liver transcriptome. Part of this effect is mediated via the nervous innervation, as a hepatectomy modulated and reduced the effect of LAN on liver transcripts.
Collapse
Affiliation(s)
- Anne-Loes Opperhuizen
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, Netherlands.,Laboratory of Endocrinology, Amsterdam University Medical Center, Department of Clinical Chemistry, University of Amsterdam, Amsterdam, Netherlands
| | - Ewout Foppen
- Laboratory of Endocrinology, Amsterdam University Medical Center, Department of Clinical Chemistry, University of Amsterdam, Amsterdam, Netherlands
| | - Martijs Jonker
- MAD - Dutch Genomics Service and Support Provider, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Paul Wackers
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Linda van Kerkhof
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Eric Fliers
- Amsterdam University Medical Center, Department of Endocrinology and Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Andries Kalsbeek
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, Netherlands.,Laboratory of Endocrinology, Amsterdam University Medical Center, Department of Clinical Chemistry, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam University Medical Center, Department of Endocrinology and Metabolism, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
31
|
Gnocchi D, Custodero C, Sabbà C, Mazzocca A. Circadian rhythms: a possible new player in non-alcoholic fatty liver disease pathophysiology. J Mol Med (Berl) 2019; 97:741-759. [PMID: 30953079 DOI: 10.1007/s00109-019-01780-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/10/2019] [Accepted: 03/13/2019] [Indexed: 12/16/2022]
Abstract
Over the last decades, a better knowledge of the molecular machinery supervising the regulation of circadian clocks has been achieved, and numerous findings have helped in unravelling the outstanding significance of the molecular clock for the proper regulation of our physiologic and metabolic homeostasis. Non-alcoholic fatty liver disease (NAFLD) is currently considered as one of the emerging liver pathologies in the Western countries due to the modification of eating habits and lifestyle. Although NAFLD is considered a pretty benign condition, it can progress towards non-alcoholic steatohepatitis (NASH) and eventually hepatocellular carcinoma (HCC). The pathogenic mechanisms involved in NAFLD development are complex, since this disease is a multifactorial condition. Major metabolic deregulations along with a genetic background are believed to take part in this process. In this light, the aim of this review is to give a comprehensive description of how our circadian machinery is regulated and to describe to what extent our internal clock is involved in the regulation of hormonal and metabolic homeostasis, and by extension in the development and progression of NAFLD/NASH and eventually in the onset of HCC.
Collapse
Affiliation(s)
- Davide Gnocchi
- Interdisciplinary Department of Medicine, University of Bari School of Medicine, Piazza G. Cesare, 11, 70124, Bari, Italy
| | - Carlo Custodero
- Interdisciplinary Department of Medicine, University of Bari School of Medicine, Piazza G. Cesare, 11, 70124, Bari, Italy
| | - Carlo Sabbà
- Interdisciplinary Department of Medicine, University of Bari School of Medicine, Piazza G. Cesare, 11, 70124, Bari, Italy
| | - Antonio Mazzocca
- Interdisciplinary Department of Medicine, University of Bari School of Medicine, Piazza G. Cesare, 11, 70124, Bari, Italy.
| |
Collapse
|
32
|
Mechanisms of Communication in the Mammalian Circadian Timing System. Int J Mol Sci 2019; 20:ijms20020343. [PMID: 30650649 PMCID: PMC6359556 DOI: 10.3390/ijms20020343] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 12/17/2022] Open
Abstract
24-h rhythms in physiology and behaviour are organized by a body-wide network of endogenous circadian clocks. In mammals, a central pacemaker in the hypothalamic suprachiasmatic nucleus (SCN) integrates external light information to adapt cellular clocks in all tissues and organs to the external light-dark cycle. Together, central and peripheral clocks co-regulate physiological rhythms and functions. In this review, we outline the current knowledge about the routes of communication between the environment, the main pacemakers and the downstream clocks in the body, focusing on what we currently know and what we still need to understand about the communication mechanisms by which centrally and peripherally controlled timing signals coordinate physiological functions and behaviour. We highlight recent findings that shed new light on the internal organization and function of the SCN and neuroendocrine mechanisms mediating clock-to-clock coupling. These findings have implications for our understanding of circadian network entrainment and for potential manipulations of the circadian clock system in therapeutic settings.
Collapse
|
33
|
Kervezee L, Cuesta M, Cermakian N, Boivin DB. The Phase-Shifting Effect of Bright Light Exposure on Circadian Rhythmicity in the Human Transcriptome. J Biol Rhythms 2019; 34:84-97. [DOI: 10.1177/0748730418821776] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Laura Kervezee
- Centre for Study and Treatment of Circadian Rhythms, Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Canada
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Canada
| | - Marc Cuesta
- Centre for Study and Treatment of Circadian Rhythms, Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Canada
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Canada
| | - Nicolas Cermakian
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Canada
| | - Diane B. Boivin
- Centre for Study and Treatment of Circadian Rhythms, Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Canada
| |
Collapse
|
34
|
Gastón MS, Pereyra LC, Vaira M. Artificial light at night and captivity induces differential effects on leukocyte profile, body condition, and erythrocyte size of a diurnal toad. JOURNAL OF EXPERIMENTAL ZOOLOGY PART 2018; 331:93-102. [PMID: 30320969 DOI: 10.1002/jez.2240] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/11/2018] [Accepted: 09/19/2018] [Indexed: 01/27/2023]
Abstract
Light pollution or artificial lighting at night (ALAN) is an emerging threat to biodiversity that can disrupt physiological processes and behaviors. Because ALAN stressful effects are little studied in diurnal amphibian species, we investigated if chronic ALAN exposure affects the leukocyte profile, body condition, and blood cell sizes of a diurnal toad. We hand-captured male toads of Melanophryniscus rubriventris in Angosto de Jaire (Jujuy, Argentina). We prepared blood smears from three groups of toads: "field" (toads processed in the field immediately after capture), "natural light" (toads kept in the laboratory under captivity with natural photoperiod), and "constant light" (toads kept in the laboratory under captivity with constant photoperiod/ALAN). We significantly observed higher neutrophil proportions and neutrophils to lymphocytes ratio in toads under constant light treatment. In addition, we observed significantly better body condition and higher erythrocyte size in field toads compared with captive toads. In summary, ALAN can trigger a leukocyte response to stress in males of the diurnal toad M. rubriventris. In addition, captivity can affect the body condition and erythrocyte size of these toads.
Collapse
Affiliation(s)
- María S Gastón
- Instituto de Ecorregiones Andinas (INECOA), Universidad Nacional de Jujuy, CONICET, San Salvador de Jujuy, Argentina
| | - Laura C Pereyra
- Instituto de Ecorregiones Andinas (INECOA), Universidad Nacional de Jujuy, CONICET, San Salvador de Jujuy, Argentina
| | - Marcos Vaira
- Instituto de Ecorregiones Andinas (INECOA), Universidad Nacional de Jujuy, CONICET, San Salvador de Jujuy, Argentina
| |
Collapse
|
35
|
González MMC. Dim Light at Night and Constant Darkness: Two Frequently Used Lighting Conditions That Jeopardize the Health and Well-being of Laboratory Rodents. Front Neurol 2018; 9:609. [PMID: 30116218 PMCID: PMC6084421 DOI: 10.3389/fneur.2018.00609] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 07/09/2018] [Indexed: 12/27/2022] Open
Abstract
The influence of light on mammalian physiology and behavior is due to the entrainment of circadian rhythms complemented with a direct modulation of light that would be unlikely an outcome of circadian system. In mammals, physiological and behavioral circadian rhythms are regulated by the suprachiasmatic nucleus (SCN) of the hypothalamus. This central control allows organisms to predict and anticipate environmental change, as well as to coordinate different rhythmic modalities within an individual. In adult mammals, direct retinal projections to the SCN are responsible for resetting and synchronizing physiological and behavioral rhythms to the light-dark (LD) cycle. Apart from its circadian effects, light also has direct effects on certain biological functions in such a way that the participation of the SCN would not be fundamental for this network. The objective of this review is to increase awareness, within the scientific community and commercial providers, of the fact that laboratory rodents can experience a number of adverse health and welfare outcomes attributed to commonly-used lighting conditions in animal facilities during routine husbandry and scientific procedures, widely considered as “environmentally friendly.” There is increasing evidence that exposure to dim light at night, as well as chronic constant darkness, challenges mammalian physiology and behavior resulting in disrupted circadian rhythms, neural death, a depressive-behavioral phenotype, cognitive impairment, and the deregulation of metabolic, physiological, and synaptic plasticity in both the short and long terms. The normal development and good health of laboratory rodents requires cyclical light entrainment, adapted to the solar cycle of day and night, with null light at night and safe illuminating qualities during the day. We therefore recommend increased awareness of the limited information available with regards to lighting conditions, and therefore that lighting protocols must be taken into consideration when designing experiments and duly highlighted in scientific papers. This practice will help to ensure the welfare of laboratory animals and increase the likelihood of producing reliable and reproducible results.
Collapse
Affiliation(s)
- Mónica M C González
- Sección Cronobiología y Sueño, Instituto Ferrero de Neurología y Sueño, Buenos Aires, Argentina
| |
Collapse
|
36
|
Hsieh PN, Zhang L, Jain MK. Coordination of cardiac rhythmic output and circadian metabolic regulation in the heart. Cell Mol Life Sci 2018; 75:403-416. [PMID: 28825119 PMCID: PMC5765194 DOI: 10.1007/s00018-017-2606-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 07/13/2017] [Accepted: 08/02/2017] [Indexed: 02/07/2023]
Abstract
Over the course of a 24-h day, demand on the heart rises and falls with the sleep/wake cycles of the organism. Cardiac metabolism oscillates appropriately, with the relative contributions of major energy sources changing in a circadian fashion. The cardiac peripheral clock is hypothesized to drive many of these changes, yet the precise mechanisms linking the cardiac clock to metabolism remain a source of intense investigation. Here we summarize the current understanding of circadian alterations in cardiac metabolism and physiology, with an emphasis on novel findings from unbiased transcriptomic studies. Additionally, we describe progress in elucidating the links between the cardiac peripheral clock outputs and cardiac metabolism, as well as their implications for cardiac physiology.
Collapse
Affiliation(s)
- Paishiun Nelson Hsieh
- Department of Medicine, Case Cardiovascular Research Institute, Case Western Reserve University, 2103 Cornell Road, Room 4-503, Cleveland, OH, USA
- Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Lilei Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Mukesh Kumar Jain
- Department of Medicine, Case Cardiovascular Research Institute, Case Western Reserve University, 2103 Cornell Road, Room 4-503, Cleveland, OH, USA.
- Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, Cleveland, OH, USA.
| |
Collapse
|
37
|
Rapid resetting of human peripheral clocks by phototherapy during simulated night shift work. Sci Rep 2017; 7:16310. [PMID: 29176713 PMCID: PMC5701225 DOI: 10.1038/s41598-017-16429-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 11/08/2017] [Indexed: 11/22/2022] Open
Abstract
A majority of night shift workers have their circadian rhythms misaligned to their atypical schedule. While bright light exposure at night is known to reset the human central circadian clock, the behavior of peripheral clocks under conditions of shift work is more elusive. The aim of the present study was to quantify the resetting effects of bright light exposure on both central (plasma cortisol and melatonin) and peripheral clocks markers (clock gene expression in peripheral blood mononuclear cells, PBMCs) in subjects living at night. Eighteen healthy subjects were enrolled to either a control (dim light) or a bright light group. Blood was sampled at baseline and on the 4th day of simulated night shift. In response to a night-oriented schedule, the phase of PER1 and BMAL1 rhythms in PBMCs was delayed by ~2.5–3 h (P < 0.05), while no shift was observed for the other clock genes and the central markers. Three cycles of 8-h bright light induced significant phase delays (P < 0.05) of ~7–9 h for central and peripheral markers, except BMAL1 (advanced by +5h29; P < 0.05). Here, we demonstrate in humans a lack of peripheral clock adaptation under a night-oriented schedule and a rapid resetting effect of nocturnal bright light exposure on peripheral clocks.
Collapse
|
38
|
Plano SA, Casiraghi LP, García Moro P, Paladino N, Golombek DA, Chiesa JJ. Circadian and Metabolic Effects of Light: Implications in Weight Homeostasis and Health. Front Neurol 2017; 8:558. [PMID: 29097992 PMCID: PMC5653694 DOI: 10.3389/fneur.2017.00558] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 10/04/2017] [Indexed: 12/21/2022] Open
Abstract
Daily interactions between the hypothalamic circadian clock at the suprachiasmatic nucleus (SCN) and peripheral circadian oscillators regulate physiology and metabolism to set temporal variations in homeostatic regulation. Phase coherence of these circadian oscillators is achieved by the entrainment of the SCN to the environmental 24-h light:dark (LD) cycle, coupled through downstream neural, neuroendocrine, and autonomic outputs. The SCN coordinate activity and feeding rhythms, thus setting the timing of food intake, energy expenditure, thermogenesis, and active and basal metabolism. In this work, we will discuss evidences exploring the impact of different photic entrainment conditions on energy metabolism. The steady-state interaction between the LD cycle and the SCN is essential for health and wellbeing, as its chronic misalignment disrupts the circadian organization at different levels. For instance, in nocturnal rodents, non-24 h protocols (i.e., LD cycles of different durations, or chronic jet-lag simulations) might generate forced desynchronization of oscillators from the behavioral to the metabolic level. Even seemingly subtle photic manipulations, as the exposure to a “dim light” scotophase, might lead to similar alterations. The daily amount of light integrated by the clock (i.e., the photophase duration) strongly regulates energy metabolism in photoperiodic species. Removing LD cycles under either constant light or darkness, which are routine protocols in chronobiology, can also affect metabolism, and the same happens with disrupted LD cycles (like shiftwork of jetlag) and artificial light at night in humans. A profound knowledge of the photic and metabolic inputs to the clock, as well as its endocrine and autonomic outputs to peripheral oscillators driving energy metabolism, will help us to understand and alleviate circadian health alterations including cardiometabolic diseases, diabetes, and obesity.
Collapse
Affiliation(s)
- Santiago A Plano
- Chronophysiology Laboratory, Institute for Biomedical Research (BIOMED - CONICET), School of Medical Sciences, Universidad Católica Argentina (UCA), Buenos Aires, Argentina.,Laboratorio de Cronobiología, Universidad Nacional de Quilmes - CONICET, Buenos Aires, Argentina
| | - Leandro P Casiraghi
- Laboratorio de Cronobiología, Universidad Nacional de Quilmes - CONICET, Buenos Aires, Argentina
| | - Paula García Moro
- Laboratorio de Cronobiología, Universidad Nacional de Quilmes - CONICET, Buenos Aires, Argentina
| | - Natalia Paladino
- Laboratorio de Cronobiología, Universidad Nacional de Quilmes - CONICET, Buenos Aires, Argentina
| | - Diego A Golombek
- Laboratorio de Cronobiología, Universidad Nacional de Quilmes - CONICET, Buenos Aires, Argentina
| | - Juan J Chiesa
- Laboratorio de Cronobiología, Universidad Nacional de Quilmes - CONICET, Buenos Aires, Argentina
| |
Collapse
|
39
|
Cold-sensing TRPM8 channel participates in circadian control of the brown adipose tissue. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2415-2427. [PMID: 28943398 DOI: 10.1016/j.bbamcr.2017.09.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/18/2017] [Accepted: 09/19/2017] [Indexed: 01/11/2023]
Abstract
Transient receptor potential (TRP) channels are known to regulate energy metabolism, and TRPM8 has become an interesting player in this context. Here we demonstrate the role of the cold sensor TRPM8 in the regulation of clock gene and clock controlled genes in brown adipose tissue (BAT). We investigated TrpM8 temporal profile in the eyes, suprachiasmatic nucleus and BAT; only BAT showed temporal variation of TrpM8 transcripts. Eyes from mice lacking TRPM8 lost the temporal profile of Per1 in LD cycle. This alteration in the ocular circadian physiology may explain the delay in the onset of locomotor activity in response to light pulse, as compared to wild type animals (WT). Brown adipocytes from TrpM8 KO mice exhibited a larger multilocularity in comparison to WT or TrpV1 KO mice. In addition, Ucp1 and UCP1 expression was significantly reduced in TrpM8 KO mice in comparison to WT mice. Regarding circadian components, the expression of Per1, Per2, Bmal1, Pparα, and Pparβ oscillated in WT mice kept in LD, whereas in the absence of TRPM8 the expression of clock genes was reduced in amplitude and lack temporal oscillation. Thus, our results reveal new roles for TRPM8 channel: it participates in the regulation of clock and clock-controlled genes in the eyes and BAT, and in BAT thermogenesis. Since disruption of the clock machinery has been associated with many metabolic disorders, the pharmacological modulation of TRPM8 channel may become a promising therapeutic target to counterbalance weight gain, through increased thermogenesis, energy expenditure, and clock gene activation.
Collapse
|
40
|
Opperhuizen AL, Stenvers DJ, Jansen RD, Foppen E, Fliers E, Kalsbeek A. Light at night acutely impairs glucose tolerance in a time-, intensity- and wavelength-dependent manner in rats. Diabetologia 2017; 60:1333-1343. [PMID: 28374068 PMCID: PMC5487588 DOI: 10.1007/s00125-017-4262-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/02/2017] [Indexed: 10/25/2022]
Abstract
AIMS/HYPOTHESIS Exposure to light at night (LAN) has increased dramatically in recent decades. Animal studies have shown that chronic dim LAN induced obesity and glucose intolerance. Furthermore, several studies in humans have demonstrated that chronic exposure to artificial LAN may have adverse health effects with an increased risk of metabolic disorders, including type 2 diabetes. It is well-known that acute exposure to LAN affects biological clock function, hormone secretion and the activity of the autonomic nervous system, but data on the effects of LAN on glucose homeostasis are lacking. This study aimed to investigate the acute effects of LAN on glucose metabolism. METHODS Male Wistar rats were subjected to i.v. glucose or insulin tolerance tests while exposed to 2 h of LAN in the early or late dark phase. In subsequent experiments, different light intensities and wavelengths were used. RESULTS LAN exposure early in the dark phase at ZT15 caused increased glucose responses during the first 20 min after glucose infusion (p < 0.001), whereas LAN exposure at the end of the dark phase, at ZT21, caused increased insulin responses during the first 10 min (p < 0.01), indicating that LAN immediately induces glucose intolerance in rats. Subsequent experiments demonstrated that the effect of LAN was both intensity- and wavelength-dependent. White light of 50 and 150 lx induced greater glucose responses than 5 and 20 lx, whereas all intensities other than 5 lx reduced locomotor activity. Green light induced glucose intolerance, but red and blue light did not, suggesting the involvement of a specific retina-brain pathway. CONCLUSIONS/INTERPRETATION Together, these data show that exposure to LAN has acute adverse effects on glucose metabolism in a time-, intensity- and wavelength-dependent manner.
Collapse
Affiliation(s)
- Anne-Loes Opperhuizen
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Meibergdreef 47, 1105 BA, Amsterdam, the Netherlands.
- Laboratory of Endocrinology, Department of Clinical Chemistry, Academic Medical Centre (AMC) University of Amsterdam, Amsterdam, the Netherlands.
| | - Dirk J Stenvers
- Laboratory of Endocrinology, Department of Clinical Chemistry, Academic Medical Centre (AMC) University of Amsterdam, Amsterdam, the Netherlands
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC) University of Amsterdam, Amsterdam, the Netherlands
| | - Remi D Jansen
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Meibergdreef 47, 1105 BA, Amsterdam, the Netherlands
| | - Ewout Foppen
- Laboratory of Endocrinology, Department of Clinical Chemistry, Academic Medical Centre (AMC) University of Amsterdam, Amsterdam, the Netherlands
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC) University of Amsterdam, Amsterdam, the Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC) University of Amsterdam, Amsterdam, the Netherlands
| | - Andries Kalsbeek
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Meibergdreef 47, 1105 BA, Amsterdam, the Netherlands
- Laboratory of Endocrinology, Department of Clinical Chemistry, Academic Medical Centre (AMC) University of Amsterdam, Amsterdam, the Netherlands
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC) University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
41
|
Versteeg RI, Stenvers DJ, Visintainer D, Linnenbank A, Tanck MW, Zwanenburg G, Smilde AK, Fliers E, Kalsbeek A, Serlie MJ, la Fleur SE, Bisschop PH. Acute Effects of Morning Light on Plasma Glucose and Triglycerides in Healthy Men and Men with Type 2 Diabetes. J Biol Rhythms 2017; 32:130-142. [PMID: 28470119 PMCID: PMC5423535 DOI: 10.1177/0748730417693480] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Ambient light intensity is signaled directly to hypothalamic areas that regulate energy metabolism. Observational studies have shown associations between ambient light intensity and plasma glucose and lipid levels, but human data on the acute metabolic effects of light are scarce. Since light is the main signal indicating the onset of the diurnal phase of physical activity and food intake in humans, we hypothesized that bright light would affect glucose and lipid metabolism. Therefore, we determined the acute effects of bright light on plasma glucose and lipid concentrations in 2 randomized crossover trials: (1) in 8 healthy lean men and (2) in 8 obese men with type 2 diabetes. From 0730 h, subjects were exposed to either bright light (4000 lux) or dim light (10 lux) for 5 h. After 1 h of light exposure, subjects consumed a 600-kcal mixed meal. Primary endpoints were fasting and postprandial plasma glucose levels. In healthy men, bright light did not affect fasting or postprandial plasma glucose levels. However, bright light increased fasting and postprandial plasma triglycerides. In men with type 2 diabetes, bright light increased fasting and postprandial glucose levels. In men with type 2 diabetes, bright light did not affect fasting triglyceride levels but increased postprandial triglyceride levels. We show that ambient light intensity acutely affects human plasma glucose and triglyceride levels. Our findings warrant further research into the consequences of the metabolic effects of light for the diagnosis and prevention of hyperglycemia and dyslipidemia.
Collapse
Affiliation(s)
- Ruth I Versteeg
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Dirk J Stenvers
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Dana Visintainer
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Andre Linnenbank
- Department of Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Michael W Tanck
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Gooitzen Zwanenburg
- Biosystem Data Analysis Group, University of Amsterdam, Amsterdam, The Netherlands
| | - Age K Smilde
- Biosystem Data Analysis Group, University of Amsterdam, Amsterdam, The Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Susanne E la Fleur
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Peter H Bisschop
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
42
|
Gnocchi D, Bruscalupi G. Circadian Rhythms and Hormonal Homeostasis: Pathophysiological Implications. BIOLOGY 2017; 6:biology6010010. [PMID: 28165421 PMCID: PMC5372003 DOI: 10.3390/biology6010010] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/10/2017] [Accepted: 01/23/2017] [Indexed: 02/07/2023]
Abstract
Over recent years, a deeper comprehension of the molecular mechanisms that control biological clocks and circadian rhythms has been achieved. In fact, many studies have contributed to unravelling the importance of the molecular clock for the regulation of our physiology, including hormonal and metabolic homeostasis. Here we will review the structure, organisation and molecular machinery that make our circadian clock work, and its relevance for the proper functioning of physiological processes. We will also describe the interconnections between circadian rhythms and endocrine homeostasis, as well as the underlying consequences that circadian dysregulations might have in the development of several pathologic affections. Finally, we will discuss how a better knowledge of such relationships might prove helpful in designing new therapeutic approaches for endocrine and metabolic diseases.
Collapse
Affiliation(s)
- Davide Gnocchi
- Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm 14186, Sweden.
| | - Giovannella Bruscalupi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome 00185, Italy.
| |
Collapse
|
43
|
Feeding cycle-dependent circulating insulin fluctuation is not a dominant Zeitgeber for mouse peripheral clocks except in the liver: Differences between endogenous and exogenous insulin effects. Biochem Biophys Res Commun 2017; 483:165-170. [DOI: 10.1016/j.bbrc.2016.12.173] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 12/26/2016] [Indexed: 11/20/2022]
|
44
|
Abstract
Use of artificial light resulted in relative independence from the natural light-dark (LD) cycle, allowing human subjects to shift the timing of food intake and work to convenient times. However, the increase in artificial light exposure parallels the increase in obesity prevalence. Light is the dominant Zeitgeber for the central circadian clock, which resides within the hypothalamic suprachiasmatic nucleus, and coordinates daily rhythm in feeding behaviour and metabolism. Eating during inappropriate light conditions may result in metabolic disease via changes in the biological clock. In this review, we describe the physiological role of light in the circadian timing system and explore the interaction between the circadian timing system and metabolism. Furthermore, we discuss the acute and chronic effects of artificial light exposure on food intake and energy metabolism in animals and human subjects. We propose that living in synchrony with the natural daily LD cycle promotes metabolic health and increased exposure to artificial light at inappropriate times of day has adverse effects on metabolism, feeding behaviour and body weight regulation. Reducing the negative side effects of the extensive use of artificial light in human subjects might be useful in the prevention of metabolic disease.
Collapse
|
45
|
Khan ZA, Yumnamcha T, Rajiv C, Sanjita Devi H, Mondal G, Devi SD, Bharali R, Chattoraj A. Melatonin biosynthesizing enzyme genes and clock genes in ovary and whole brain of zebrafish (Danio rerio): Differential expression and a possible interplay. Gen Comp Endocrinol 2016; 233:16-31. [PMID: 27179881 DOI: 10.1016/j.ygcen.2016.05.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 05/05/2016] [Accepted: 05/11/2016] [Indexed: 12/27/2022]
Abstract
The present study on zebrafish (Danio rerio) is the first attempt to demonstrate the circadian mRNA expression of melatonin biosynthesizing enzyme genes (Tph1a, Aanat1, Aanat2 and Hiomt) and clock associated genes (Bmal1a, Clock1a, Per1b, Per2 and Cry2a) in the ovary with a comparison to whole brain in normal (LD=12h L:12h D) and altered photic conditions (continuous dark, DD; continuous light, LL). Moreover, the present study also confirmed the ability of zebrafish ovary to biosynthesize melatonin both in vivo and in vitro with a significant difference at day and night. qRT-PCR analysis of genes revealed a dark acrophase of Aanat2 in both organs while Tph1 is in whole brain in LD condition. On the contrary, Bmal1a and Clock1a giving their peak in light, thereby showing a negative correlation with Tph1a and Aanat2. In LD-ovary, the acrophase of Tph1a, Bmal1a and Clock1a is in light and thus display a positive correlation. This trend of relationship in respect to Tph1a is not changing in altered photic conditions in both organs (except in DD-ovary). On the other hand this association for Aanat2 is varying in ovary under altered photic conditions but only in DD-whole brain. Both in LD and LL the expression of Aanat2 in brain presenting an opposite acrophase with both Bmal1a and Clock1a of ovary and consequently displaying a strong negative correlation among them. Interestingly, all ovarian clock associated genes become totally arrhythmic in DD, representing a loss of correlation between the melatonin synthesizing genes in brain and clock associated genes in ovary. The result is also indicating the formation of two heterodimers namely Clock1a:Bmal1a and Per2:Cry2a in the functioning of clock genes in both organs, irrespective of photic conditions, as they are exhibiting a strong significant positive correlation. Collectively, our data suggest that ovary of zebrafish is working as peripheral oscillator having its own melatonin biosynthesizing machinery and signifying a possible correlation with central oscillating system in various photic conditions.
Collapse
Affiliation(s)
- Zeeshan Ahmad Khan
- Biological Rhythm Laboratory, Animal Resources Programme, Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal 795 001, Manipur, India
| | - Thangal Yumnamcha
- Biological Rhythm Laboratory, Animal Resources Programme, Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal 795 001, Manipur, India
| | - Chongtham Rajiv
- Biological Rhythm Laboratory, Animal Resources Programme, Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal 795 001, Manipur, India
| | - Haobijam Sanjita Devi
- Biological Rhythm Laboratory, Animal Resources Programme, Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal 795 001, Manipur, India
| | - Gopinath Mondal
- Biological Rhythm Laboratory, Animal Resources Programme, Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal 795 001, Manipur, India
| | - Sh Dharmajyoti Devi
- Biological Rhythm Laboratory, Animal Resources Programme, Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal 795 001, Manipur, India
| | - Rupjyoti Bharali
- Department of Biotechnology, Gauhati University, Guwahati 781 014, Assam, India
| | - Asamanja Chattoraj
- Biological Rhythm Laboratory, Animal Resources Programme, Institute of Bioresources and Sustainable Development, Department of Biotechnology, Government of India, Takyelpat, Imphal 795 001, Manipur, India.
| |
Collapse
|
46
|
When to eat? The influence of circadian rhythms on metabolic health: are animal studies providing the evidence? Nutr Res Rev 2016; 29:180-193. [PMID: 27364352 DOI: 10.1017/s095442241600010x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
As obesity and metabolic diseases rise, there is need to investigate physiological and behavioural aspects associated with their development. Circadian rhythms have a profound influence on metabolic processes, as they prepare the body to optimise energy use and storage. Moreover, food-related signals confer temporal order to organs involved in metabolic regulation. Therefore food intake should be synchronised with the suprachiasmatic nucleus (SCN) to elaborate efficient responses to environmental challenges. Human studies suggest that a loss of synchrony between mealtime and the SCN promotes obesity and metabolic disturbances. Animal research using different paradigms has been performed to characterise the effects of timing of food intake on metabolic profiles. Therefore the purpose of the present review is to critically examine the evidence of animal studies, to provide a state of the art on metabolic findings and to assess whether the paradigms used in rodent models give the evidence to support a 'best time' for food intake. First we analyse and compare the current findings of studies where mealtime has been shifted out of phase from the light-dark cycle. Then, we analyse studies restricting meal times to different moments within the active period. So far animal studies correlate well with human studies, demonstrating that restricting food intake to the active phase limits metabolic disturbances produced by high-energy diets and that eating during the inactive/sleep phase leads to a worse metabolic outcome. Based on the latter we discuss the missing elements and possible mechanisms leading to the metabolic consequences, as these are still lacking.
Collapse
|
47
|
Erion R, King AN, Wu G, Hogenesch JB, Sehgal A. Neural clocks and Neuropeptide F/Y regulate circadian gene expression in a peripheral metabolic tissue. eLife 2016; 5. [PMID: 27077948 PMCID: PMC4862751 DOI: 10.7554/elife.13552] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 04/07/2016] [Indexed: 11/23/2022] Open
Abstract
Metabolic homeostasis requires coordination between circadian clocks in different tissues. Also, systemic signals appear to be required for some transcriptional rhythms in the mammalian liver and the Drosophila fat body. Here we show that free-running oscillations of the fat body clock require clock function in the PDF-positive cells of the fly brain. Interestingly, rhythmic expression of the cytochrome P450 transcripts, sex-specific enzyme 1 (sxe1) and Cyp6a21, which cycle in the fat body independently of the local clock, depends upon clocks in neurons expressing neuropeptide F (NPF). NPF signaling itself is required to drive cycling of sxe1 and Cyp6a21 in the fat body, and its mammalian ortholog, Npy, functions similarly to regulate cycling of cytochrome P450 genes in the mouse liver. These data highlight the importance of neuronal clocks for peripheral rhythms, particularly in a specific detoxification pathway, and identify a novel and conserved role for NPF/Npy in circadian rhythms. DOI:http://dx.doi.org/10.7554/eLife.13552.001 Many processes in the body follow rhythms that repeat over 24 hours and are synchronized to the cycle of day and night. Our sleep pattern is a well-known example, but others include daily fluctuations in body temperature and the production of several hormones. Internal clocks located in the brain and other organs drive these rhythms by altering the activity of certain genes depending on the time of day. Animals have specific organs that contain enzymes needed to break down toxic molecules in the body, and the levels of several of these enzymes rise and fall over each 24-hour period. In mammals, these enzymes are found in the liver, but in insects they are found in an organ called the fat body. Here, Erion, King et al. set out to determine the extent to which the internal clock in the brain influences the daily rhythms of these enzymes. The experiments show that a hormone released by the nervous system is required for the levels of the detoxifying enzymes to change in 24-hour cycles. This hormone – termed Neuropeptide F in fruit flies and Neuropeptide Y in mice – is also known to stimulate both mice and fruit flies to eat. Since toxic molecules often enter the body during feeding, Erion, King et al. speculate that it may be beneficial to link the detoxification process to feeding by using the same mechanism to control both processes. The next step following on from this work would be to find out exactly how neuropeptide F drives the 24-hour rhythms in the fat body and other organs. DOI:http://dx.doi.org/10.7554/eLife.13552.002
Collapse
Affiliation(s)
- Renske Erion
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, United States
| | - Anna N King
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, United States
| | - Gang Wu
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, United States
| | - John B Hogenesch
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, United States
| | - Amita Sehgal
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
48
|
Mazzoccoli G, Rubino R, Tiberio C, Giuliani F, Vinciguerra M, Oben J, De Cata A, Tarquini R, De Cosmo S, Liu S, Cai Y. Clock gene expression in human and mouse hepatic models shows similar periodicity but different dynamics of variation. Chronobiol Int 2016; 33:181-90. [DOI: 10.3109/07420528.2015.1132722] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
49
|
Sakakibara H, Torii Yasuda M, Shimoi K. Effects of environmental and social stressors on biological rhythms. THE JOURNAL OF PHYSICAL FITNESS AND SPORTS MEDICINE 2016. [DOI: 10.7600/jpfsm.5.143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
| | | | - Kayoko Shimoi
- School of Food and Nutritional Sciences, University of Shizuoka
| |
Collapse
|
50
|
Poletini MO, Ramos BC, Moraes MN, Castrucci AML. Nonvisual Opsins and the Regulation of Peripheral Clocks by Light and Hormones. Photochem Photobiol 2015; 91:1046-55. [DOI: 10.1111/php.12494] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/23/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Maristela O. Poletini
- Department of Physiology and Biophysics; Institute of Biological Sciences; Federal University of Minas Gerais; Belo Horizonte Brazil
- Department of Physiology; Institute of Biosciences; University of São Paulo; São Paulo Brazil
| | - Bruno C. Ramos
- Department of Physiology; Institute of Biosciences; University of São Paulo; São Paulo Brazil
| | - Maria Nathalia Moraes
- Department of Physiology; Institute of Biosciences; University of São Paulo; São Paulo Brazil
| | - Ana Maria L. Castrucci
- Department of Physiology; Institute of Biosciences; University of São Paulo; São Paulo Brazil
| |
Collapse
|