1
|
Bennett C, Pettikiriarachchi A, McLean ARD, Harding R, Blewitt ME, Seillet C, Pasricha SR. Serum iron and transferrin saturation variation are circadian regulated and linked to the harmonic circadian oscillations of erythropoiesis and hepatic Tfrc expression in mice. Am J Hematol 2024; 99:2075-2083. [PMID: 39152780 DOI: 10.1002/ajh.27447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 06/28/2024] [Accepted: 07/21/2024] [Indexed: 08/19/2024]
Abstract
Serum iron has long been thought to exhibit diurnal variation and is subsequently considered an unreliable biomarker of systemic iron status. Circadian regulation (endogenous ~24-h periodic oscillation of a biologic function) governs many critical physiologic processes. It is unknown whether serum iron levels are regulated by circadian machinery; likewise, the circadian nature of key players of iron homeostasis is unstudied. Here we show that serum iron, transferrin saturation (TSAT), hepatic transferrin receptor (TFR1) gene (Tfrc) expression, and erythropoietic activity exhibit circadian rhythms. Daily oscillations of serum iron, TSAT, hepatic Tfrc expression, and erythropoietic activity are maintained in mice housed in constant darkness, where oscillation reflects an endogenous circadian period. Oscillations of serum iron, TSAT, hepatic Tfrc, and erythropoietic activity were ablated when circadian machinery was disrupted in Bmal1 knockout mice. Interestingly, we find that circadian oscillations of erythropoietic activity and hepatic Tfrc expression are maintained in opposing phase, likely allowing for optimized usage and storage of serum iron whilst maintaining adequate serum levels and TSAT. This study provides the first confirmatory evidence that serum iron is circadian regulated, discerns circadian rhythms of TSAT, a widely used clinical marker of iron status, and uncovers liver-specific circadian regulation of TFR1, a major player in cellular iron uptake.
Collapse
Affiliation(s)
- Cavan Bennett
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Anne Pettikiriarachchi
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Alistair R D McLean
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Rebecca Harding
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Marnie E Blewitt
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Cyril Seillet
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Sant-Rayn Pasricha
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Diagnostic Haematology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- Clinical Haematology at the Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
2
|
Ono M, Burgess DE, Johnson SR, Elayi CS, Esser KA, Seward TS, Boychuk CR, Carreño AP, Stalcup RA, Prabhat A, Schroder EA, Delisle BP. Feeding behavior modifies the circadian variation in RR and QT intervals by distinct mechanisms in mice. Am J Physiol Regul Integr Comp Physiol 2024; 327:R109-R121. [PMID: 38766772 PMCID: PMC11380991 DOI: 10.1152/ajpregu.00025.2024] [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: 01/29/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024]
Abstract
Rhythmic feeding behavior is critical for regulating phase and amplitude in the ≈24-h variation of heart rate (RR intervals), ventricular repolarization (QT intervals), and core body temperature in mice. We hypothesized changes in cardiac electrophysiology associated with feeding behavior were secondary to changes in core body temperature. Telemetry was used to record electrocardiograms and core body temperature in mice during ad libitum-fed conditions and after inverting normal feeding behavior by restricting food access to the light cycle. Light cycle-restricted feeding modified the phase and amplitude of 24-h rhythms in RR and QT intervals, and core body temperature to realign with the new feeding time. Changes in core body temperature alone could not account for changes in phase and amplitude in the ≈24-h variation of the RR intervals. Heart rate variability analysis and inhibiting β-adrenergic and muscarinic receptors suggested that changes in the phase and amplitude of 24-h rhythms in RR intervals were secondary to changes in autonomic signaling. In contrast, changes in QT intervals closely mirrored changes in core body temperature. Studies at thermoneutrality confirmed that the daily variation in QT interval, but not RR interval, primarily reflected daily changes in core body temperature (even in ad libitum-fed conditions). Correcting the QT interval for differences in core body temperature helped unmask QT interval prolongation after starting light cycle-restricted feeding and in a mouse model of long QT syndrome. We conclude feeding behavior alters autonomic signaling and core body temperature to regulate phase and amplitude in RR and QT intervals, respectively.NEW & NOTEWORTHY We used time-restricted feeding and thermoneutrality to demonstrate that different mechanisms regulate the 24-h rhythms in heart rate and ventricular repolarization. The daily rhythm in heart rate reflects changes in autonomic input, whereas daily rhythms in ventricular repolarization reflect changes in core body temperature. This novel finding has major implications for understanding 24-h rhythms in mouse cardiac electrophysiology, arrhythmia susceptibility in transgenic mouse models, and interpretability of cardiac electrophysiological data acquired in thermoneutrality.
Collapse
Affiliation(s)
- Makoto Ono
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Don E Burgess
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Sidney R Johnson
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Claude S Elayi
- CHI Saint Joseph Hospital, Lexington, Kentucky, United States
| | - Karyn A Esser
- Department of Physiology and Aging, University of Florida, Gainesville, Florida, United States
| | - Tanya S Seward
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Carie R Boychuk
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Andrés P Carreño
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Rebecca A Stalcup
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Abhilash Prabhat
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| | - Elizabeth A Schroder
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
- Department of Internal Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Brian P Delisle
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
3
|
Jones AA, Marino GM, Arble DM. Time-restricted feeding reveals a role for neural respiratory clocks in optimizing daily ventilatory-metabolic coupling in mice. Am J Physiol Endocrinol Metab 2024; 327:E111-E120. [PMID: 38836780 PMCID: PMC11390118 DOI: 10.1152/ajpendo.00111.2024] [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: 03/18/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
The master circadian clock, located in the suprachiasmatic nuclei (SCN), organizes the daily rhythm in minute ventilation (V̇e). However, the extent that the daily rhythm in V̇e is secondary to SCN-imposed O2 and CO2 cycles (i.e., metabolic rate) or driven by other clock mechanisms remains unknown. Here, we experimentally shifted metabolic rate using time-restricted feeding (without affecting light-induced synchronization of the SCN) to determine the influence of metabolic rate in orchestrating the daily V̇e rhythm. Mice eating predominantly at night exhibited robust daily rhythms in O2 consumption (V̇o2), CO2 production (V̇co2), and V̇e with similar peak times (approximately ZT18) that were consistent with SCN organization. However, feeding mice exclusively during the day separated the relative timing of metabolic and ventilatory rhythms, resulting in an approximately 8.5-h advance in V̇co2 and a disruption of the V̇e rhythm, suggesting opposing circadian and metabolic influences on V̇e. To determine if the molecular clock of cells involved in the neural control of breathing contributes to the daily V̇e rhythm, we examined V̇e in mice lacking BMAL1 in Phox2b-expressing respiratory cells (i.e., BKOP mice). The ventilatory and metabolic rhythms of predominantly night-fed BKOP mice did not differ from wild-type mice. However, in contrast to wild-type mice, exclusive day feeding of BKOP mice led to an unfettered daily V̇e rhythm with a peak time aligning closely with the daily V̇co2 rhythm. Taken together, these results indicate that both daily V̇co2 changes and intrinsic circadian time-keeping within Phox2b respiratory cells are predominant orchestrators of the daily rhythm in ventilation.NEW & NOTEWORTHY The master circadian clock organizes the daily rhythm in ventilation; however, the extent that this rhythm is driven by SCN regulation of metabolic rate versus other clock mechanisms remains unknown. We report that metabolic rate alone is insufficient to explain the daily oscillation in ventilation and that neural respiratory clocks within Phox2b-expressing cells additionally optimize breathing. Collectively, these findings advance our mechanistic understanding of the circadian rhythm in ventilatory control.
Collapse
Affiliation(s)
- Aaron A Jones
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States
| | - Gabriella M Marino
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States
| | - Deanna M Arble
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States
| |
Collapse
|
4
|
Ono M, Burgess DE, Johnson SR, Elayi CS, Esser KA, Seward TS, Boychuk CR, Carreño AP, Stalcup RA, Prabhat A, Schroder EA, Delisle BP. Feeding Behavior Modifies the Circadian Variation in RR and QT intervals by Distinct Mechanisms in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.02.565372. [PMID: 37961515 PMCID: PMC10635091 DOI: 10.1101/2023.11.02.565372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Rhythmic feeding behavior is critical for regulating the phase and amplitude in the ≍24-hour variation of the heart rate (RR intervals), ventricular repolarization (QT intervals), and core body temperature in mice. We hypothesized the changes in cardiac electrophysiology associated with feeding behavior were secondary to changes in core body temperature. Telemetry was used to record electrocardiograms and core body temperature in mice during ad libitum-fed conditions and after inverting normal feeding behavior by restricting food access to the light cycle. Light cycle-restricted feeding quickly modified the phase and amplitude of the 24-hour rhythms in RR intervals, QT intervals, and core body temperature to realign with the new feeding time. Heart rate variability analysis and inhibiting β-adrenergic and muscarinic receptors suggested that the changes in the phase and amplitude of the 24-hour rhythms in RR intervals were secondary to changes in autonomic signaling. In contrast, the changes in the QT intervals closely mirrored changes in core body temperature. Studies at thermoneutrality confirmed the daily variation in the QT interval, but not the RR interval, and reflected daily changes in core body temperature (even in ad libitum-fed conditions). Correcting the QT interval for differences in core body temperature helped to unmask QT interval prolongation after starting light cycle-restricted feeding and in a mouse model of long QT syndrome. We conclude feeding behavior alters autonomic signaling and core body temperature to regulate the phase and amplitude in RR and QT intervals, respectively.
Collapse
|
5
|
Amatobi KM, Ozbek-Unal AG, Schäbler S, Deppisch P, Helfrich-Förster C, Mueller MJ, Wegener C, Fekete A. The circadian clock is required for rhythmic lipid transport in Drosophila in interaction with diet and photic condition. J Lipid Res 2023; 64:100417. [PMID: 37481037 PMCID: PMC10550813 DOI: 10.1016/j.jlr.2023.100417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 07/24/2023] Open
Abstract
Modern lifestyle is often at odds with endogenously driven rhythmicity, which can lead to circadian disruption and metabolic syndrome. One signature for circadian disruption is a reduced or altered metabolite cycling in the circulating tissue reflecting the current metabolic status. Drosophila is a well-established model in chronobiology, but day-time dependent variations of transport metabolites in the fly circulation are poorly characterized. Here, we sampled fly hemolymph throughout the day and analyzed diacylglycerols (DGs), phosphoethanolamines (PEs) and phosphocholines (PCs) using LC-MS. In wild-type flies kept on sugar-only medium under a light-dark cycle, all transport lipid species showed a synchronized bimodal oscillation pattern with maxima at the beginning and end of the light phase which were impaired in period01 clock mutants. In wild-type flies under constant dark conditions, the oscillation became monophasic with a maximum in the middle of the subjective day. In strong support of clock-driven oscillations, levels of the targeted lipids peaked once in the middle of the light phase under time-restricted feeding independent of the time of food intake. When wild-type flies were reared on full standard medium, the rhythmic alterations of hemolymph lipid levels were greatly attenuated. Our data suggest that the circadian clock aligns daily oscillations of DGs, PEs, and PCs in the hemolymph to the anabolic siesta phase, with a strong influence of light on phase and modality.
Collapse
Affiliation(s)
- Kelechi M Amatobi
- Biocenter, Julius-von-Sachs-Institute, Pharmaceutical Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany; Biocenter, Theodor-Boveri-Institute, Würzburg Insect Research (WIR), Neurobiology and Genetics, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Ayten Gizem Ozbek-Unal
- Biocenter, Julius-von-Sachs-Institute, Pharmaceutical Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Stefan Schäbler
- Biocenter, Julius-von-Sachs-Institute, Pharmaceutical Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Peter Deppisch
- Biocenter, Theodor-Boveri-Institute, Würzburg Insect Research (WIR), Neurobiology and Genetics, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Charlotte Helfrich-Förster
- Biocenter, Theodor-Boveri-Institute, Würzburg Insect Research (WIR), Neurobiology and Genetics, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Martin J Mueller
- Biocenter, Julius-von-Sachs-Institute, Pharmaceutical Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Christian Wegener
- Biocenter, Theodor-Boveri-Institute, Würzburg Insect Research (WIR), Neurobiology and Genetics, Julius-Maximilians-Universität Würzburg, Würzburg, Germany.
| | - Agnes Fekete
- Biocenter, Julius-von-Sachs-Institute, Pharmaceutical Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany.
| |
Collapse
|
6
|
Ibarra-Mendoza B, Gomez-Gil B, Betancourt-Lozano M, Raggi L, Yáñez-Rivera B. Microbial gut dysbiosis induced by xenobiotics in model organisms and the relevance of experimental criteria: a minireview. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2023; 4:e7. [PMID: 39295907 PMCID: PMC11406412 DOI: 10.1017/gmb.2023.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 09/21/2024]
Abstract
The gut microbiota is a dynamic ecosystem involved in multiple physiological processes that affect host health. Several factors affect intestinal microbial communities including dietary exposure to xenobiotics, which is highly concerning due to their widespread distribution. Current knowledge of this topic comes from culture-dependent methods, 16S rRNA amplicon fingerprinting, and metagenomics, but a standardised procedures framework remains lacking. This minireview integrates 45 studies from a systematic search using terms related to gut microbiota and its disruption. Only publications encompassing dietary-oral exposure and experimental gut microbiota assessments were included. The results were divided and described according to the biological model used and the disruption observed in the gut microbiota. An overall dysbiotic effect was unclear due to the variety of contaminants and hosts evaluated and the experimental gaps between publications. More standardised experimental designs, including WGS and physiological tests, are needed to establish how a particular xenobiotic can alter the gut microbiota and how the results can be extrapolated.
Collapse
Affiliation(s)
| | - Bruno Gomez-Gil
- CIAD, A.C. Mazatlán Unit for Aquaculture and Environmental Management, Mazatlán, Mexico
| | | | - Luciana Raggi
- Universidad Michoacana de San Nicolás de Hidalgo - CONACYT, Mexico City, Mexico
| | - Beatriz Yáñez-Rivera
- Instituto de Ciencias del Mar y Limnología, Unidad Académica Mazatlán, Universidad Nacional Autónoma de México, Mazatlán, Mexico
| |
Collapse
|
7
|
Gallop MR, Tobin SY, Chaix A. Finding balance: understanding the energetics of time-restricted feeding in mice. Obesity (Silver Spring) 2023; 31 Suppl 1:22-39. [PMID: 36513496 PMCID: PMC9877167 DOI: 10.1002/oby.23607] [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: 06/28/2022] [Revised: 08/17/2022] [Accepted: 09/06/2022] [Indexed: 12/15/2022]
Abstract
Over the course of mammalian evolution, the ability to store energy likely conferred a survival advantage when food became scarce. A long-term increase in energy storage results from an imbalance between energy intake and energy expenditure, two tightly regulated parameters that generally balance out to maintain a fairly stable body weight. Understanding the molecular determinants of this feat likely holds the key to new therapeutic development to manage obesity and associated metabolic dysfunctions. Time-restricted feeding (TRF), a dietary intervention that limits feeding to the active phase, can prevent and treat obesity and metabolic dysfunction in rodents fed a high-fat diet, likely by exerting effects on energetic balance. Even when body weight is lower in mice on active-phase TRF, food intake is generally isocaloric as compared with ad libitum fed controls. This discrepancy between body weight and energy intake led to the hypothesis that energy expenditure is increased during TRF. However, at present, there is no consensus in the literature as to how TRF affects energy expenditure and energy balance as a whole, and the mechanisms behind metabolic adaptation under TRF are unknown. This review examines our current understanding of energy balance on TRF in rodents and provides a framework for future studies to evaluate the energetics of TRF and its molecular determinants.
Collapse
Affiliation(s)
- Molly R Gallop
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT
| | - Selene Y Tobin
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT
| | - Amandine Chaix
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT
| |
Collapse
|
8
|
Kord-Varkaneh H, Salehi-Sahlabadi A, Tinsley GM, Santos HO, Hekmatdoost A. Effects of time-restricted feeding (16/8) combined with a low-sugar diet on the management of non-alcoholic fatty liver disease: A randomized controlled trial. Nutrition 2023; 105:111847. [PMID: 36257081 DOI: 10.1016/j.nut.2022.111847] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/23/2022] [Accepted: 09/05/2022] [Indexed: 10/14/2022]
Abstract
OBJECTIVES Emerging studies have employed time-restricted feeding (TRF) and a low-sugar diet alone in the management of non-alcoholic fatty liver disease (NAFLD), but their combination has not been tested. The aim of this study was to investigate the effects of TRF combined with a low-sugar diet on NAFLD parameters, cardiometabolic and inflammatory biomarkers, and body composition in patients with NAFLD. METHODS A 12-wk randomized controlled trial was performed to compare the effects of TRF (16 h fasting/8 h feeding daily [16/8]) plus a low-sugar diet versus a control diet based on traditional meal distribution in patients with NAFLD. Changes in body composition, anthropometric indices, and liver and cardiometabolic markers were investigated. RESULTS TRF 16/8 with a low-sugar diet reduced body fat (26.7 ± 5.4 to 24.2 ± 4.9 kg), body weight (83.8 ± 12.7 to 80.5 ± 12.1 kg), waist circumference (104.59 ± 10.47 to 101.91 ± 7.42 cm), and body mass index (29.1 ± 2.6 to 28 ± 2.7 kg/m2), as well as circulating levels of fasting blood glucose and liver (alanine aminotransferase, 34 ± 13.9 to 21.2 ± 5.4 U/L; aspartate aminotransferase, 26.3 ± 6.2 to 20.50 ± 4 U/L; γ-glutamyl transpeptidase, 33 ± 15 to 23.2 ± 11.1 U/L; fibrosis score, 6.3 ± 1 to 5.2 ± 1.2 kPa; and controlled attenuation parameter, 322.9 ± 34.9 to 270.9 ± 36.2 dB/m), lipids (triacylglycerols, 201.5 ± 35.3 to 133.3 ± 48.7 mg/dL; total cholesterol, 190 ± 36.6 to 157.8 ± 33.6 mg/dL; and low-density lipoprotein cholesterol, 104.6 ± 27.3 to 84 ± 26.3 mg/dL), and inflammatory markers (high-sensitivity C-reactive protein, 3.1 ± 1.1 to 2 ± 0.9 mg/L; and cytokeratin-18, 1.35 ± 0.03 to 1.16 ± 0.03 ng/mL). These results were statistically significant (P < 0.05) compared with the control group. CONCLUSIONS TRF plus a low-sugar diet can reduce adiposity and improve liver, lipid, and inflammatory markers in patients with NAFLD.
Collapse
Affiliation(s)
- Hamed Kord-Varkaneh
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ammar Salehi-Sahlabadi
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Grant M Tinsley
- Department of Kinesiology & Sport Management, Texas Tech University, Lubbock, Texas, USA
| | - Heitor O Santos
- School of Medicine, Federal University of Uberlandia (UFU), Uberlandia, Minas Gerais, Brazil
| | - Azita Hekmatdoost
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Wang H, Zhang H, Su Y. New Insights into the Diurnal Rhythmicity of Gut Microbiota and Its Crosstalk with Host Circadian Rhythm. Animals (Basel) 2022; 12:1677. [PMID: 35804575 PMCID: PMC9264800 DOI: 10.3390/ani12131677] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 12/14/2022] Open
Abstract
Unlike the strictly hierarchical organization in the circadian clock system, the gut microbiota rhythmicity has a more complex multilayer network of all taxonomic levels of microbial taxa and their metabolites. However, it is worth noting that the functionality of the gut microbiota rhythmicity is highly dependent on the host circadian clock and host physiological status. Here, we discussed the diurnal rhythmicity of the gut microbiota; its crucial role in host physiology, health, and metabolism; and the crosstalk between the gut microbial rhythmicity and host circadian rhythm. This knowledge lays the foundation for the development of chronotherapies targeting the gut microbiota. However, the formation mechanism, its beneficial effects on the host of gut microbial rhythmicity, and the dynamic microbial-host crosstalk are not yet clear and warrant further research.
Collapse
Affiliation(s)
- Hongyu Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (H.W.); (H.Z.)
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - He Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (H.W.); (H.Z.)
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (H.W.); (H.Z.)
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
10
|
Colom-Pellicer M, Rodríguez RM, Soliz-Rueda JR, de Assis LVM, Navarro-Masip È, Quesada-Vázquez S, Escoté X, Oster H, Mulero M, Aragonès G. Proanthocyanidins Restore the Metabolic Diurnal Rhythm of Subcutaneous White Adipose Tissue According to Time-Of-Day Consumption. Nutrients 2022; 14:2246. [PMID: 35684049 PMCID: PMC9182881 DOI: 10.3390/nu14112246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 02/01/2023] Open
Abstract
Consumption of grape seed proanthocyanidin extract (GSPE) has beneficial effects on the functionality of white adipose tissue (WAT). However, although WAT metabolism shows a clear diurnal rhythm, whether GSPE consumption could affect WAT rhythmicity in a time-dependent manner has not been studied. Ninety-six male Fischer rats were fed standard (STD, two groups) or cafeteria (CAF, four groups) diet for 9 weeks (n = 16 each group). From week 6 on, CAF diet animals were supplemented with vehicle or 25 mg GSPE/kg of body weight either at the beginning of the light/rest phase (ZT0) or at the beginning of the dark/active phase (ZT12). The two STD groups were also supplemented with vehicle at ZT0 or ZT12. In week 9, animals were sacrificed at 6 h intervals (n = 4) to analyze the diurnal rhythms of subcutaneous WAT metabolites by nuclear magnetic resonance spectrometry. A total of 45 metabolites were detected, 19 of which presented diurnal rhythms in the STD groups. Although most metabolites became arrhythmic under CAF diet, GSPE consumption at ZT12, but not at ZT0, restored the rhythmicity of 12 metabolites including compounds involved in alanine, aspartate, and glutamate metabolism. These results demonstrate that timed GSPE supplementation may restore, at least partially, the functional dynamics of WAT when it is consumed at the beginning of the active phase. This study opens an innovative strategy for time-dependent polyphenol treatment in obesity and metabolic diseases.
Collapse
Affiliation(s)
- Marina Colom-Pellicer
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (M.C.-P.); (R.M.R.); (J.R.S.-R.); (È.N.-M.); (M.M.)
| | - Romina M. Rodríguez
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (M.C.-P.); (R.M.R.); (J.R.S.-R.); (È.N.-M.); (M.M.)
| | - Jorge R. Soliz-Rueda
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (M.C.-P.); (R.M.R.); (J.R.S.-R.); (È.N.-M.); (M.M.)
| | - Leonardo Vinícius Monteiro de Assis
- Center of Brain, Behavior and Metabolism, Institute of Neurobiology, University of Lübeck, Marie Curie Street, 23562 Lübeck, Germany; (L.V.M.d.A.); (H.O.)
| | - Èlia Navarro-Masip
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (M.C.-P.); (R.M.R.); (J.R.S.-R.); (È.N.-M.); (M.M.)
| | - Sergio Quesada-Vázquez
- Unitat de Nutrició i Salut, Centre Tecnològic de Catalunya, Eurecat, 43204 Reus, Spain; (S.Q.-V.); (X.E.)
| | - Xavier Escoté
- Unitat de Nutrició i Salut, Centre Tecnològic de Catalunya, Eurecat, 43204 Reus, Spain; (S.Q.-V.); (X.E.)
| | - Henrik Oster
- Center of Brain, Behavior and Metabolism, Institute of Neurobiology, University of Lübeck, Marie Curie Street, 23562 Lübeck, Germany; (L.V.M.d.A.); (H.O.)
| | - Miquel Mulero
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (M.C.-P.); (R.M.R.); (J.R.S.-R.); (È.N.-M.); (M.M.)
| | - Gerard Aragonès
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (M.C.-P.); (R.M.R.); (J.R.S.-R.); (È.N.-M.); (M.M.)
| |
Collapse
|
11
|
Tsurudome Y, Akamine T, Horiguchi M, Wada Y, Fujimura A, Ushijima K. Potential mechanism of hepatic lipid accumulation during a long-term rest phase restricted feeding in mice. Chronobiol Int 2022; 39:1132-1143. [PMID: 35603436 DOI: 10.1080/07420528.2022.2077746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Eating during a rest phase disrupts the biological clock system and leads to obesity and metabolic diseases. Although a rest phase restricted feeding (RF) is reported to enhance hepatic lipid accumulation, the mechanism(s) of the phenomenon is still unknown. This study evaluated the potential involvement of the CD36-related transport of lipids into the liver in mice with the RF procedure. This study showed that hepatic lipid accumulation was more significant in the RF group compared with mice under an active phase restricted feeding (AF). The RF procedure also elevated the expression of CD36 mRNA and its protein on the cellular membrane throughout the day. The transcription factor profiling array revealed that the RF activated the proliferator-activated receptor-γ (PPARγ), one of the CD36 transcript enhancers. In the liver of RF mice, the expression of miR-27b-3p, which is known to interfere with PPARγ gene expression, significantly decreased. These results suggest that the RF procedure inhibits the expression of miR-27b-3p in the liver and subsequently elevates PPARγ activity. Activated PPARγ might lead to CD36 upregulation, which, in turn, stimulates the transport of lipids into the liver.
Collapse
Affiliation(s)
- Yuya Tsurudome
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi, Japan
| | - Takahiro Akamine
- Department of Pharmacology, Faculty of Medicine, Oita University, Oita, Japan
| | - Michiko Horiguchi
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi, Japan
| | - Yukiyo Wada
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi, Japan
| | - Akio Fujimura
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi, Japan.,Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Tochigi, Japan
| | - Kentaro Ushijima
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi, Japan.,Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
12
|
Lee JH, Moon E, Park J, Oh CE, Hong YR, Yoon M. Optimization of Analysis of Circadian Rest-Activity Rhythm Using Cosinor Analysis in Mice. Psychiatry Investig 2022; 19:380-385. [PMID: 35620823 PMCID: PMC9136527 DOI: 10.30773/pi.2021.0395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/05/2022] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE Data processing in analysis of circadian rhythm was performed in various ways. However, there was a lack of evidence for the optimal analysis of circadian rest-activity rhythm. Therefore, we aimed to perform mathematical simulations of data processing to investigate proper evidence for the optimal analysis of circadian rest-activity rhythm. METHODS Locomotor activities of 20 ICR male mice were measured by infrared motion detectors. The data of locomotor activities was processed using data summation, data average, and data moving average methods for cosinor analysis. Circadian indices were estimated according to time block, respectively. Also, statistical F and p-values were calculated by zero-amplitude test. RESULTS The data moving average result showed well-fitted cosine curves independent of data processing time. Meanwhile, the amplitude, MESOR, and acrophase were properly estimated within 800 seconds in data summation and data average methods. CONCLUSION These findings suggest that data moving average would be an optimal method for data processing in a cosinor analysis and data average within 800-second data processing time might be adaptable. The results of this study can be helpful to analyze circadian restactivity rhythms and integrate the results of the studies using different data processing methods.
Collapse
Affiliation(s)
- Jung Hyun Lee
- Department of Pediatrics, Kosin University College of Medicine, Busan, Republic of Korea
| | - Eunsoo Moon
- Department of Psychiatry and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea.,Department of Psychiatry, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jeonghyun Park
- Department of Psychiatry and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Chi Eun Oh
- Department of Pediatrics, Kosin University College of Medicine, Busan, Republic of Korea
| | - Yoo Rha Hong
- Department of Pediatrics, Kosin University College of Medicine, Busan, Republic of Korea
| | - Min Yoon
- Department of Applied Mathematics, Pukyung National University, Busan, Republic of Korea
| |
Collapse
|
13
|
Randomized controlled trial for time-restricted eating in healthy volunteers without obesity. Nat Commun 2022; 13:1003. [PMID: 35194047 PMCID: PMC8864028 DOI: 10.1038/s41467-022-28662-5] [Citation(s) in RCA: 113] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/03/2022] [Indexed: 12/24/2022] Open
Abstract
Time-restricted feeding (TRF) improves metabolic health. Both early TRF (eTRF, food intake restricted to the early part of the day) and mid-day TRF (mTRF, food intake restricted to the middle of the day) have been shown to have metabolic benefits. However, the two regimens have yet to be thoroughly compared. We conducted a five-week randomized trial to compare the effects of the two TRF regimens in healthy individuals without obesity (ChiCTR2000029797). The trial has completed. Ninety participants were randomized to eTRF (n=30), mTRF (n=30), or control groups (n=30) using a computer-based random-number generator. Eighty-two participants completed the entire five-week trial and were analyzed (28 in eTRF, 26 in mTRF, 28 in control groups). The primary outcome was the change in insulin resistance. Researchers who assessed the outcomes were blinded to group assignment, but participants and care givers were not. Here we show that eTRF was more effective than mTRF at improving insulin sensitivity. Furthermore, eTRF, but not mTRF, improved fasting glucose, reduced total body mass and adiposity, ameliorated inflammation, and increased gut microbial diversity. No serious adverse events were reported during the trial. In conclusion, eTRF showed greater benefits for insulin resistance and related metabolic parameters compared with mTRF. Clinical Trial Registration URL: http://www.chictr.org.cn/showproj.aspx?proj=49406. Time-restricted eating, both early (eTRF) and mid-day (mTRF), have been shown to have metabolic benefits. Here the authors report a randomized controlled trial to compare the effects of eTRF and mTRF in healthy volunteers without obesity, and find that eTRF is more effective in improving the primary outcome insulin sensitivity.
Collapse
|
14
|
Fernández-Pérez A, Sanz-Magro A, Moratalla R, Vallejo M. Restricting feeding to dark phase fails to entrain circadian activity and energy expenditure oscillations in Pitx3-mutant Aphakia mice. Cell Rep 2022; 38:110241. [PMID: 35021074 DOI: 10.1016/j.celrep.2021.110241] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 11/19/2021] [Accepted: 12/16/2021] [Indexed: 01/01/2023] Open
Abstract
Metabolic homeostasis is under circadian regulation to adapt energy requirements to light-dark cycles. Feeding cycles are regulated by photic stimuli reaching the suprachiasmatic nucleus via retinohypothalamic axons and by nutritional information involving dopaminergic neurotransmission. Previously, we reported that Pitx3-mutant Aphakia mice with altered development of the retinohypothalamic tract and the dopaminergic neurons projecting to the striatum, are resistant to locomotor and metabolic entrainment by time-restricted feeding. In their Matters Arising article, Scarpa et al. (2022) challenge this conclusion using mice from the same strain but following a different experimental paradigm involving calorie restriction. Here, we address their concerns by extending the analyses of our previous data, by identifying important differences in the experimental design between both studies and by presenting additional results on the dopaminergic deficit in the brain of Aphakia mice. This Matters Arising Response article addresses the Matters Arising article by Scarpa et al. (2022), published concurrently in Cell Reports.
Collapse
Affiliation(s)
- Antonio Fernández-Pérez
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas /Universidad Autónoma de Madrid, Calle Arturo Duperier 4, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Madrid, Spain
| | - Adrián Sanz-Magro
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, and CIBERNED, Instituto de Salud Carlos III, 28002 Madrid, Spain
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, and CIBERNED, Instituto de Salud Carlos III, 28002 Madrid, Spain
| | - Mario Vallejo
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas /Universidad Autónoma de Madrid, Calle Arturo Duperier 4, 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Madrid, Spain.
| |
Collapse
|
15
|
Wang W, Huang Z, Huang L, Gao L, Cui L, Cowley M, Guo L, Chen C. Time-Restricted Feeding Restored Insulin-Growth Hormone Balance and Improved Substrate and Energy Metabolism in MC4RKO Obese Mice. Neuroendocrinology 2022; 112:174-185. [PMID: 33735897 DOI: 10.1159/000515960] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/01/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Dysregulation of metabolic regulatory hormones often occurs during the progress of obesity. Key regulatory hormone insulin-growth hormone (GH) balance has recently been proposed to maintain metabolism profiles. Time-restricted feeding (TRF) is an effective strategy against obesity without detailed research on pulsatile GH releasing patterns. METHODS TRF was performed in an over-eating melanocortin 4 receptor-knockout (MC4RKO) obese mouse model using normal food. Body weight and food intake were measured. Series of blood samples were collected for 6-h pulsatile GH profile, glucose tolerance test, and insulin tolerance test at 5, 8, and 9 weeks of TRF, respectively. Indirect calorimetric recordings were performed by the Phenomaster system at 6 weeks for 1 week, and body composition was measured by nuclear magnetic resonance spectroscopy (NMR). Substrate- and energy metabolism-related gene expressions were measured in terminal liver and subcutaneous white adipose tissues. RESULTS TRF increased pulsatile GH secretion in dark phase and suppressed hyperinsulinemia in MC4RKO obese mice to reach a reduced insulin/GH ratio. This was accompanied by the improvement in insulin sensitivity, metabolic flexibility, glucose tolerance, and decreased glucose fluctuation, together with appropriate modification of gene expression involved in substrate metabolism and adipose tissue browning. NMR measurement showed that TRF decreased fat mass but increased lean mass. Indirect calorimeter recording indicated that TRF decreased the respiratory exchange ratio (RER) reflecting consumption of more fatty acid in energy production in light phase and increased the oxygen consumption during activities in dark phase. CONCLUSIONS TRF effectively decreases hyperinsulinemia and restores pulsatile GH secretion in the overeating obese mice with significant improvement in substrate and energy metabolism and body composition without reducing total caloric intake.
Collapse
Affiliation(s)
- Weihao Wang
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhengxiang Huang
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Lili Huang
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Lyn Gao
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Ling Cui
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Michael Cowley
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Lixin Guo
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
16
|
Rodrigues LGF, de Araujo LD, Roa SLR, Bueno AC, Uchoa ET, Antunes-Rodrigues J, Moreira AC, Elias LLK, de Castro M, Martins CS. Restricted feeding modulates peripheral clocks and nutrient sensing pathways in rats. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2021; 65:549-561. [PMID: 34591411 PMCID: PMC10528573 DOI: 10.20945/2359-3997000000407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 06/29/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE Feeding restriction in rats alters the oscillators in suprachiasmatic, paraventricular, and arcuate nuclei, hypothalamic areas involved in food intake. In the present study, using the same animals and experimental protocol, we aimed to analyze if food restriction could reset clock genes (Clock, Bmal1) and genes involved in lipid metabolism (Pgc1a, Pparg, Ucp2) through nutrient-sensing pathways (Sirt1, Ampk, Nampt) in peripheral tissues. METHODS Rats were grouped according to food access: Control group (CG, food ad libitum), Restricted night-fed (RF-n, food access during 2 h at night), Restricted day-fed (RF-d, food access during 2 h in the daytime), and Day-fed (DF, food access during 12 h in the daytime). After 21 days, rats were decapitated at ZT3 (0900-1000 h), ZT11 (1700-1800 h), or ZT17 (2300-2400 h). Blood, liver, brown (BAT) and peri-epididymal (PAT) adipose tissues were collected. Plasma corticosterone and gene expression were evaluated by radioimmunoassay and qPCR, respectively. RESULTS In the liver, the expression pattern of Clock and Bmal1 shifted when food access was dissociated from rat nocturnal activity; this phenomenon was attenuated in adipose tissues. Daytime feeding also inverted the profile of energy-sensing and lipid metabolism-related genes in the liver, whereas calorie restriction induced a pre-feeding increased expression of these genes. In adipose tissues, Sirt1 expression was modified by daytime feeding and calorie restriction, with concomitant expression of Pgc1a, Pparg, and Ucp2 but not Ampk and Nampt. CONCLUSION Feeding restriction reset clock genes and genes involved in lipid metabolism through nutrient-sensing-related genes in rat liver, brown, and peri-epididymal adipose tissues.
Collapse
Affiliation(s)
- Luis Guilherme F Rodrigues
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Leonardo D de Araujo
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Silvia L R Roa
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Ana C Bueno
- Departamento de Pediatria, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Ernane T Uchoa
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - José Antunes-Rodrigues
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Ayrton C Moreira
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Lucila L K Elias
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Margaret de Castro
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Clarissa S Martins
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil,
| |
Collapse
|
17
|
Nishide S, Suzuki Y, Ono D, Honma S, Honma KI. The Food-entrainable Oscillator Is a Complex of Non-SCN Activity Bout Oscillators Uncoupled From the SCN Circadian Pacemaker. J Biol Rhythms 2021; 36:575-588. [PMID: 34634956 DOI: 10.1177/07487304211047937] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The food-entrainable oscillator, which underlies the prefeeding activity peak developed by restricted daily feeding (RF) in rodents, does not depend on the circadian pacemaker in the suprachiasmatic nucleus (SCN) or on the known clock genes. In the present study, to clarify the roles of SCN circadian pacemaker and nutrient conditions on the development of prefeeding activity peak, RF of 3-h daily feeding was imposed on four groups of adult male mice for 10 cycles at different circadian times, zeitgeber time (ZT)2, ZT8, ZT14, and ZT20, where ZT0 is the time of lights-on in LD12:12. Seven days after the termination of RF session with ad libitum feeding in between, total food deprivation (FD) for 72 h was imposed. Wheel-running activity and core body temperature were measured throughout the experiment. Immediately after the RF or FD session, the PER2::LUC rhythms were measured in the cultured SCN slices and peripheral tissues. Not only the buildup process and magnitude of the prefeeding activity peak, but also the percentages of nocturnal activity and hypothermia developed under RF were significantly different among the four groups, indicating the involvement of light entrained circadian pacemaker. The buildup of prefeeding activity peak was accomplished by either phase-advance or phase-delay shifts (or both) of activity bouts comprising a nocturnal band. Hypothermia under FD was less prominent in RF-exposed mice than in naïve counterparts, indicating that restricted feeding increases tolerance to caloric restriction as well as to the heat loss mechanism. RF phase-shifted the peripheral clocks but FD did not affect the clocks in any tissue examined. These findings are better understood by assuming multiple bout oscillators, which are located outside the SCN and directly drive activity bouts uncoupled from the circadian pacemaker by RF or hypothermia.
Collapse
Affiliation(s)
- Shinya Nishide
- Department of Physiology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Physiology Laboratory, Department of Occupational Therapy, School of Rehabilitation Sciences, Health Sciences University of Hokkaido, Tobetsu, Japan
| | - Yohko Suzuki
- Department of Chronomedicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Daisuke Ono
- Photonic Bioimaging Section, Research Center for Cooperative Projects, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Sato Honma
- Department of Chronomedicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Ken-Ichi Honma
- Department of Chronomedicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
18
|
Healy KL, Morris AR, Liu AC. Circadian Synchrony: Sleep, Nutrition, and Physical Activity. FRONTIERS IN NETWORK PHYSIOLOGY 2021; 1:732243. [PMID: 35156088 PMCID: PMC8830366 DOI: 10.3389/fnetp.2021.732243] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/20/2021] [Indexed: 08/01/2023]
Abstract
The circadian clock in mammals regulates the sleep/wake cycle and many associated behavioral and physiological processes. The cellular clock mechanism involves a transcriptional negative feedback loop that gives rise to circadian rhythms in gene expression with an approximately 24-h periodicity. To maintain system robustness, clocks throughout the body must be synchronized and their functions coordinated. In mammals, the master clock is located in the suprachiasmatic nucleus (SCN) of the hypothalamus. The SCN is entrained to the light/dark cycle through photic signal transduction and subsequent induction of core clock gene expression. The SCN in turn relays the time-of-day information to clocks in peripheral tissues. While the SCN is highly responsive to photic cues, peripheral clocks are more sensitive to non-photic resetting cues such as nutrients, body temperature, and neuroendocrine hormones. For example, feeding/fasting and physical activity can entrain peripheral clocks through signaling pathways and subsequent regulation of core clock genes and proteins. As such, timing of food intake and physical activity matters. In an ideal world, the sleep/wake and feeding/fasting cycles are synchronized to the light/dark cycle. However, asynchronous environmental cues, such as those experienced by shift workers and frequent travelers, often lead to misalignment between the master and peripheral clocks. Emerging evidence suggests that the resulting circadian disruption is associated with various diseases and chronic conditions that cause further circadian desynchrony and accelerate disease progression. In this review, we discuss how sleep, nutrition, and physical activity synchronize circadian clocks and how chronomedicine may offer novel strategies for disease intervention.
Collapse
|
19
|
Gutierrez Lopez DE, Lashinger LM, Weinstock GM, Bray MS. Circadian rhythms and the gut microbiome synchronize the host's metabolic response to diet. Cell Metab 2021; 33:873-887. [PMID: 33789092 DOI: 10.1016/j.cmet.2021.03.015] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 01/22/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022]
Abstract
The molecular circadian clock and symbiotic host-microbe relationships both evolved as mechanisms that enhance metabolic responses to environmental challenges. The gut microbiome benefits the host by breaking down diet-derived nutrients indigestible by the host and generating microbiota-derived metabolites that support host metabolism. Similarly, cellular circadian clocks optimize organismal physiology to the environment by influencing the timing and coordination of metabolic processes. Host-microbe interactions are influenced by dietary quality and timing, as well as daily light/dark cycles that entrain circadian rhythms in the host. Together, the gut microbiome and the molecular circadian clock play a coordinated role in neural processing, metabolism, adipogenesis, inflammation, and disease initiation and progression. This review examines the bidirectional interactions between the circadian clock, gut microbiota, and host metabolic systems and their effects on obesity and energy homeostasis. Directions for future research and the development of therapies that leverage these systems to address metabolic disease are highlighted.
Collapse
Affiliation(s)
- Diana E Gutierrez Lopez
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Laura M Lashinger
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - George M Weinstock
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Storrs, CT 06032, USA
| | - Molly S Bray
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
20
|
Guo Y, Wang QJ, Zhang KH, Yao CY, Huang J, Li Q, Liu ZY, Zhang Y, Shan CH, Liu P, Wang MZ, An L, Tian JH, Wu ZH. Night-restricted feeding improves locomotor activity rhythm and modulates nutrient utilization to accelerate growth in rabbits. FASEB J 2020; 35:e21166. [PMID: 33184921 DOI: 10.1096/fj.202001265rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 10/19/2020] [Accepted: 10/23/2020] [Indexed: 11/11/2022]
Abstract
An unfavorable lifestyle disrupts the circadian rhythm, leading to metabolic dysfunction in adult humans and animals. Increasing evidence suggests that night-restricted feeding (NRF) can effectively prevent ectopic fat deposition caused by circadian rhythm disruption, and reduce the risk of metabolic diseases. However, previous studies have mainly focused on the prevention of obesity in adults by regulating dietary patterns, whereas limited attention has been paid to the effect of NRF on metabolism during growth and development. Here, we used weaning rabbits as models and found that NRF increased body weight gain without increasing feed intake, and promoted insulin-mediated protein synthesis through the mTOR/S6K pathway and muscle formation by upregulating MYOG. NRF improved the circadian clock, promoted PDH-regulated glycolysis and CPT1B-regulated fatty-acid β-oxidation, and reduced fat content in the serum and muscles. In addition, NRF-induced body temperature oscillation might be partly responsible for the improvement in the circadian clock and insulin sensitivity. Time-restricted feeding could be used as a nondrug intervention to prevent obesity and accelerate growth in adolescents.
Collapse
Affiliation(s)
- Yao Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qiang-Jun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ke-Hao Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Chun-Yan Yao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jie Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qin Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhong-Ying Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yu Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Chun-Hua Shan
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Peng Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Mei-Zhi Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lei An
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jian-Hui Tian
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhong-Hong Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
21
|
Kobayashi A, Suzuki Y, Sugai S. Specificity of transaminase activities in the prediction of drug-induced hepatotoxicity. J Toxicol Sci 2020; 45:515-537. [PMID: 32879252 DOI: 10.2131/jts.45.515] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The activities of the transaminases (aminotransferases) alanine aminotransferase and aspartate aminotransferase in the blood (serum or plasma) are widely used as sensitive markers of possible tissue damage and, in particular for liver toxicity. On the other hand, an increase in transaminase activities is not always accompanied by findings suggestive of hepatotoxicity. Transaminases are some of the key enzymes in the gluconeogenesis and glycolysis pathways and exist in many organs and tissues which have high activities of the gluconeogenesis and glycolysis. The activities of transaminases are altered not only in the liver but also in other organs by modification of gluconeogenesis by nutritional or hormonal factors and this phenomenon leads to alteration of transaminase activity in the blood. Drugs, which are considered to directly or secondarily modify gluconeogenesis through lowering blood glucose levels or activating lipid metabolism, such as α-glucosidase inhibitors and fibrates, slightly increase transaminase activities in the blood but there is little evidence that the phenomenon is related to drug-induced liver injury (DILI). This type of elevations can be called pharmacology-related elevation. The pharmacology-related elevation of transaminase activities sometimes makes it difficult to assess precisely the potential hepatotoxicity of new investigational drugs. Considering the characteristic of transaminases, concomitant use of new biomarkers more specific to hepatic injury is needed in the assessment of DILI both in non-clinical and clinical studies. In this review, we will discuss the specificity of transaminases to DILI and future perspectives for transaminases in the estimation of risk of DILI.
Collapse
Affiliation(s)
- Akio Kobayashi
- Toxicology Research Lab., Central Pharmaceutical Research Institute, JAPAN TOBACCO INC
| | - Yusuke Suzuki
- Toxicology Research Lab., Central Pharmaceutical Research Institute, JAPAN TOBACCO INC
| | - Shoichiro Sugai
- Toxicology Research Lab., Central Pharmaceutical Research Institute, JAPAN TOBACCO INC
| |
Collapse
|
22
|
Refinetti R. Circadian rhythmicity of body temperature and metabolism. Temperature (Austin) 2020; 7:321-362. [PMID: 33251281 PMCID: PMC7678948 DOI: 10.1080/23328940.2020.1743605] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/19/2022] Open
Abstract
This article reviews the literature on the circadian rhythms of body temperature and whole-organism metabolism. The two rhythms are first described separately, each description preceded by a review of research methods. Both rhythms are generated endogenously but can be affected by exogenous factors. The relationship between the two rhythms is discussed next. In endothermic animals, modulation of metabolic activity can affect body temperature, but the rhythm of body temperature is not a mere side effect of the rhythm of metabolic thermogenesis associated with general activity. The circadian system modulates metabolic heat production to generate the body temperature rhythm, which challenges homeothermy but does not abolish it. Individual cells do not regulate their own temperature, but the relationship between circadian rhythms and metabolism at the cellular level is also discussed. Metabolism is both an output of and an input to the circadian clock, meaning that circadian rhythmicity and metabolism are intertwined in the cell.
Collapse
Affiliation(s)
- Roberto Refinetti
- Department of Psychology, University of New Orleans, New Orleans, LA, USA
| |
Collapse
|
23
|
Pickel L, Sung HK. Feeding Rhythms and the Circadian Regulation of Metabolism. Front Nutr 2020; 7:39. [PMID: 32363197 PMCID: PMC7182033 DOI: 10.3389/fnut.2020.00039] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/20/2020] [Indexed: 12/16/2022] Open
Abstract
The molecular circadian clock regulates metabolic processes within the cell, and the alignment of these clocks between tissues is essential for the maintenance of metabolic homeostasis. The possibility of misalignment arises from the differential responsiveness of tissues to the environmental cues that synchronize the clock (zeitgebers). Although light is the dominant environmental cue for the master clock of the suprachiasmatic nucleus, many other tissues are sensitive to feeding and fasting. When rhythms of feeding behavior are altered, for example by shift work or the constant availability of highly palatable foods, strong feedback is sent to the peripheral molecular clocks. Varying degrees of phase shift can cause the systemic misalignment of metabolic processes. Moreover, when there is a misalignment between the endogenous rhythms in physiology and environmental inputs, such as feeding during the inactive phase, the body's ability to maintain homeostasis is impaired. The loss of phase coordination between the organism and environment, as well as internal misalignment between tissues, can produce cardiometabolic disease as a consequence. The aim of this review is to synthesize the work on the mechanisms and metabolic effects of circadian misalignment. The timing of food intake is highlighted as a powerful environmental cue with the potential to destroy or restore the synchrony of circadian rhythms in metabolism.
Collapse
Affiliation(s)
- Lauren Pickel
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Hoon-Ki Sung
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Mindikoglu AL, Abdulsada MM, Jain A, Choi JM, Jalal PK, Devaraj S, Mezzari MP, Petrosino JF, Opekun AR, Jung SY. Intermittent fasting from dawn to sunset for 30 consecutive days is associated with anticancer proteomic signature and upregulates key regulatory proteins of glucose and lipid metabolism, circadian clock, DNA repair, cytoskeleton remodeling, immune system and cognitive function in healthy subjects. J Proteomics 2020; 217:103645. [PMID: 31927066 DOI: 10.1016/j.jprot.2020.103645] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 12/13/2019] [Accepted: 01/08/2020] [Indexed: 02/06/2023]
Abstract
Murine studies showed that disruption of circadian clock rhythmicity could lead to cancer and metabolic syndrome. Time-restricted feeding can reset the disrupted clock rhythm, protect against cancer and metabolic syndrome. Based on these observations, we hypothesized that intermittent fasting for several consecutive days without calorie restriction in humans would induce an anticarcinogenic proteome and the key regulatory proteins of glucose and lipid metabolism. Fourteen healthy subjects fasted from dawn to sunset for over 14 h daily. Fasting duration was 30 consecutive days. Serum samples were collected before 30-day intermittent fasting, at the end of 4th week during 30-day intermittent fasting, and one week after 30-day intermittent fasting. An untargeted serum proteomic profiling was performed using ultra high-performance liquid chromatography/tandem mass spectrometry. Our results showed that 30-day intermittent fasting was associated with an anticancer serum proteomic signature, upregulated key regulatory proteins of glucose and lipid metabolism, circadian clock, DNA repair, cytoskeleton remodeling, immune system, and cognitive function, and resulted in a serum proteome protective against cancer, metabolic syndrome, inflammation, Alzheimer's disease, and several neuropsychiatric disorders. These findings suggest that fasting from dawn to sunset for 30 consecutive days can be preventive and adjunct therapy in cancer, metabolic syndrome, and several cognitive and neuropsychiatric diseases. SIGNIFICANCE: Our study has important clinical implications. Our results showed that intermittent fasting from dawn to sunset for over 14 h daily for 30 consecutive days was associated with an anticancer serum proteomic signature and upregulated key regulatory proteins of glucose and lipid metabolism, insulin signaling, circadian clock, DNA repair, cytoskeleton remodeling, immune system, and cognitive function, and resulted in a serum proteome protective against cancer, obesity, diabetes, metabolic syndrome, inflammation, Alzheimer's disease, and several neuropsychiatric disorders. Importantly, these findings occurred in the absence of any calorie restriction and significant weight loss. These findings suggest that intermittent fasting from dawn to sunset can be a preventive and adjunct therapy in cancer, metabolic syndrome and Alzheimer's disease and several neuropsychiatric diseases.
Collapse
Affiliation(s)
- Ayse L Mindikoglu
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, United States of America; Michael E. DeBakey Department of Surgery, Division of Abdominal Transplantation, Baylor College of Medicine, Houston, TX, United States of America.
| | - Mustafa M Abdulsada
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, United States of America
| | - Antrix Jain
- Advanced Technology Core, Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, United States of America
| | - Jong Min Choi
- Advanced Technology Core, Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, United States of America
| | - Prasun K Jalal
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, United States of America; Michael E. DeBakey Department of Surgery, Division of Abdominal Transplantation, Baylor College of Medicine, Houston, TX, United States of America
| | - Sridevi Devaraj
- Clinical Chemistry and Point of Care Technology, Texas Children's Hospital and Health Centers, Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States of America
| | - Melissa P Mezzari
- The Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, United States of America
| | - Joseph F Petrosino
- The Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, United States of America
| | - Antone R Opekun
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, United States of America; Department of Pediatrics, Division of Gastroenterology, Nutrition and Hepatology, Baylor College of Medicine, Houston, TX, United States of America
| | - Sung Yun Jung
- Advanced Technology Core, Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, United States of America; Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America
| |
Collapse
|
25
|
del Río-Martín A, Pérez-Taboada I, Fernández-Pérez A, Moratalla R, de la Villa P, Vallejo M. Hypomorphic Expression of Pitx3 Disrupts Circadian Clocks and Prevents Metabolic Entrainment of Energy Expenditure. Cell Rep 2019; 29:3678-3692.e4. [DOI: 10.1016/j.celrep.2019.11.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 05/13/2019] [Accepted: 11/06/2019] [Indexed: 12/11/2022] Open
|
26
|
Greenwell BJ, Trott AJ, Beytebiere JR, Pao S, Bosley A, Beach E, Finegan P, Hernandez C, Menet JS. Rhythmic Food Intake Drives Rhythmic Gene Expression More Potently than the Hepatic Circadian Clock in Mice. Cell Rep 2019; 27:649-657.e5. [PMID: 30995463 DOI: 10.1016/j.celrep.2019.03.064] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/03/2019] [Accepted: 03/15/2019] [Indexed: 02/02/2023] Open
Abstract
Every mammalian tissue exhibits daily rhythms in gene expression to control the activation of tissue-specific processes at the most appropriate time of the day. Much of this rhythmic expression is thought to be driven cell autonomously by molecular circadian clocks present throughout the body. By manipulating the daily rhythm of food intake in the mouse, we here show that more than 70% of the cycling mouse liver transcriptome loses rhythmicity under arrhythmic feeding. Remarkably, core clock genes are not among the 70% of genes losing rhythmic expression, and their expression continues to exhibit normal oscillations in arrhythmically fed mice. Manipulation of rhythmic food intake also alters the timing of key signaling and metabolic pathways without altering the hepatic clock oscillations. Our findings thus demonstrate that systemic signals driven by rhythmic food intake significantly contribute to driving rhythms in liver gene expression and metabolic functions independently of the cell-autonomous hepatic clock.
Collapse
Affiliation(s)
- Ben J Greenwell
- Program of Genetics, Texas A&M University, College Station, TX 77843, USA; Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Alexandra J Trott
- Program of Genetics, Texas A&M University, College Station, TX 77843, USA; Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | | | - Shanny Pao
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Alexander Bosley
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Erin Beach
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Patrick Finegan
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | | | - Jerome S Menet
- Program of Genetics, Texas A&M University, College Station, TX 77843, USA; Department of Biology, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
27
|
Role of the Circadian Clock in the Metabolic Syndrome and Nonalcoholic Fatty Liver Disease. Dig Dis Sci 2018; 63:3187-3206. [PMID: 30121811 DOI: 10.1007/s10620-018-5242-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/06/2018] [Indexed: 12/20/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in industrialized nations and is strongly associated with the metabolic syndrome. The prevalence of NAFLD continues to rise along with the epidemic of the metabolic syndrome. Metabolic homeostasis is linked to the circadian clock (rhythm), with multiple signaling pathways in organs regulated by circadian clock genes, and recent studies of circadian clock gene functions suggest that disruption of the circadian rhythm is associated with significant morbidity and mortality, including the metabolic syndrome. In the industrialized world, various human behaviors and activities such as work and eating patterns, jet lag, and sleep deprivation interfere with the circadian rhythm, leading to perturbations in metabolism and development of the metabolic syndrome. In this review, we discuss how disruption of the circadian rhythm is associated with various metabolic conditions that comprise the metabolic syndrome and NAFLD.
Collapse
|
28
|
Gasmi M, Sellami M, Denham J, Padulo J, Kuvacic G, Selmi W, Khalifa R. Time-restricted feeding influences immune responses without compromising muscle performance in older men. Nutrition 2018; 51-52:29-37. [PMID: 29571007 DOI: 10.1016/j.nut.2017.12.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/20/2017] [Accepted: 12/04/2017] [Indexed: 12/24/2022]
Abstract
OBJECTIVE This study examined the effect of 12 wk of time-restricted feeding (TRF) on complete blood cell counts, natural killer cells, and muscle performance in 20- and 50-year-old men. METHODS Forty active and healthy participants were randomly divided into young experimental, young control, aged experimental, and aged control group. Experimental groups participated in TRF. Before (P1) and after (P2) TRF, participants performed a maximal exercise test to quantify muscle power. Resting venous blood samples were collected for blood count calculation. RESULTS No changes were identified in muscle power in all groups after TRF (P > 0.05). At P1, red cells, hemoglobin, and hematocrit were significantly higher in young participants compared with elderly participants (P < 0.05). At P2, this age effect was not found in red cells between the young experimental group and the aged experimental group (P > 0.05). At P1, white blood cells and neutrophils were significantly higher in young participants compared with elderly participants (P < 0.05). At P2, only neutrophils decreased significantly (P < 0.05) in experimental groups without significant (P > 0.05) difference among them. Lymphocytes decreased significantly in the aged experimental group at P2 (P < 0.05), whereas NKCD16+ and NKCD56+ decreased significantly in experimental groups at P2 (P < 0.05). TRF had no effect on CD3, CD4+, and CD8+ levels (P > 0.05). CONCLUSION TRF decreases hematocrit, total white blood cells, lymphocytes, and neutrophils in young and older men. TRF may be effective in preventing inflammation by decreasing natural killer cells. As such, TRF could be a lifestyle strategy to reduce systemic low-grade inflammation and age-related chronic diseases linked to immunosenescence, without compromising physical performance.
Collapse
Affiliation(s)
- Maha Gasmi
- Higher Institute of Sport and Physical Education of Ksar said, Tunis, Tunisia
| | - Maha Sellami
- University of Split, Faculty of Kinesiology, Split, Croatia; Tunisian Research Laboratory Sport Performance Optimization, National Center of Medicine and Science in Sports, Tunis, Tunisia.
| | - Joshua Denham
- School of Science and Technology, University of New England, Armidale, NSW, Australia
| | - Johnny Padulo
- University of Split, Faculty of Kinesiology, Split, Croatia; Tunisian Research Laboratory Sport Performance Optimization, National Center of Medicine and Science in Sports, Tunis, Tunisia; University e-campus, Novedrate, Italy
| | - Goran Kuvacic
- University of Split, Faculty of Kinesiology, Split, Croatia
| | - Walid Selmi
- Higher Institute of Sport and Physical Education of Ksar said, Tunis, Tunisia
| | - Riadh Khalifa
- Higher Institute of Sport and Physical Education of Ksar said, Tunis, Tunisia
| |
Collapse
|
29
|
Anton SD, Moehl K, Donahoo WT, Marosi K, Lee S, Mainous AG, Leeuwenburgh C, Mattson MP. Flipping the Metabolic Switch: Understanding and Applying the Health Benefits of Fasting. Obesity (Silver Spring) 2018; 26:254-268. [PMID: 29086496 PMCID: PMC5783752 DOI: 10.1002/oby.22065] [Citation(s) in RCA: 352] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 09/14/2017] [Accepted: 09/26/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Intermittent fasting (IF) is a term used to describe a variety of eating patterns in which no or few calories are consumed for time periods that can range from 12 hours to several days, on a recurring basis. This review is focused on the physiological responses of major organ systems, including the musculoskeletal system, to the onset of the metabolic switch: the point of negative energy balance at which liver glycogen stores are depleted and fatty acids are mobilized (typically beyond 12 hours after cessation of food intake). RESULTS AND CONCLUSIONS Emerging findings suggest that the metabolic switch from glucose to fatty acid-derived ketones represents an evolutionarily conserved trigger point that shifts metabolism from lipid/cholesterol synthesis and fat storage to mobilization of fat through fatty acid oxidation and fatty acid-derived ketones, which serve to preserve muscle mass and function. Thus, IF regimens that induce the metabolic switch have the potential to improve body composition in overweight individuals. Moreover, IF regimens also induce the coordinated activation of signaling pathways that optimize physiological function, enhance performance, and slow aging and disease processes. Future randomized controlled IF trials should use biomarkers of the metabolic switch (e.g., plasma ketone levels) as a measure of compliance and of the magnitude of negative energy balance during the fasting period.
Collapse
Affiliation(s)
- Stephen D. Anton
- Department of Aging and Geriatric Research, Institute on Aging, Department of Clinical and Health Psychology, University of Florida, Gainesville, FL 32610
| | - Keelin Moehl
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD 21224
| | - William T. Donahoo
- Division of Endocrinology, Diabetes and Metabolism, University of Florida College of Medicine, Gainesville, FL 32610
| | - Krisztina Marosi
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD 21224
| | - Stephanie Lee
- Department of Aging and Geriatric Research, Institute on Aging, Department of Clinical and Health Psychology, University of Florida, Gainesville, FL 32610
| | - Arch G. Mainous
- Department of Health Services Research, Management and Policy; Department of Community Health and Family Medicine, University of Florida, Gainesville, FL 32610
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, Institute on Aging, Department of Clinical and Health Psychology, University of Florida, Gainesville, FL 32610
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD 21224
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
30
|
Impact of Time-Restricted Feeding and Dawn-to-Sunset Fasting on Circadian Rhythm, Obesity, Metabolic Syndrome, and Nonalcoholic Fatty Liver Disease. Gastroenterol Res Pract 2017; 2017:3932491. [PMID: 29348746 PMCID: PMC5733887 DOI: 10.1155/2017/3932491] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 10/01/2017] [Accepted: 10/12/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity now affects millions of people and places them at risk of developing metabolic syndrome, nonalcoholic fatty liver disease (NAFLD), and even hepatocellular carcinoma. This rapidly emerging epidemic has led to a search for cost-effective methods to prevent the metabolic syndrome and NAFLD as well as the progression of NAFLD to cirrhosis and hepatocellular carcinoma. In murine models, time-restricted feeding resets the hepatic circadian clock and enhances transcription of key metabolic regulators of glucose and lipid homeostasis. Studies of the effect of dawn-to-sunset Ramadan fasting, which is akin to time-restricted feeding model, have also identified significant improvement in body mass index, serum lipid profiles, and oxidative stress parameters. Based on the findings of studies conducted on human subjects, dawn-to-sunset fasting has the potential to be a cost-effective intervention for obesity, metabolic syndrome, and NAFLD.
Collapse
|
31
|
Mullegama SV, Alaimo JT, Fountain MD, Burns B, Balog AH, Chen L, Elsea SH. RAI1 Overexpression Promotes Altered Circadian Gene Expression and Dyssomnia in Potocki-Lupski Syndrome. J Pediatr Genet 2017; 6:155-164. [PMID: 28794907 DOI: 10.1055/s-0037-1599147] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 01/17/2017] [Indexed: 12/22/2022]
Abstract
Retinoic acid induced 1 ( RAI1 ) encodes a dosage-sensitive gene that when haploinsufficient results in Smith-Magenis syndrome (SMS) and when overexpressed results in Potocki-Lupski syndrome (PTLS). Phenotypic and molecular evidence illustrates that haploinsufficiency of RAI1 disrupts circadian rhythm through the dysregulation of the master circadian regulator, circadian locomotor output cycles kaput ( CLOCK) , and other core circadian components, contributing to prominent sleep disturbances in SMS. However, the phenotypic and molecular characterization of sleep features in PTLS has not been elucidated. Using the Pittsburgh Sleep Quality Index (PSQI), caregivers of 15 school-aged children with PTLS reported difficulties in initiating sleep. Indeed, more than 70% of individuals manifested moderate to severe sleep latency, as defined by the PSQI. Moreover, these individuals manifested difficulties in sleep maintenance, with middle of the night and early morning awakenings. When assessing daytime sleepiness through the Epworth Sleepiness Scale, approximately 21% of the individuals manifested excessive daytime somnolence. This indicates that mild dyssomnia characterizes the majority of the sleep phenotype, with occasionally problematic daytime somnolence, a phenotype different than that expressed by individuals with SMS, where daytime sleepiness is a chronic problem. Gene expression analysis of the core circadian machinery in the hypothalamus of the PTLS mouse model ( Rai1 -Tg) found significant dysregulation of the transcriptional activators, Clock and Arntl , and the transcriptional repressors, Per1-3 and Cry1/2 , during both light and dark phases. These findings suggest a partial loss of circadian entrainment typically evoked by environmental photic cues. Examination of circadian clock gene expression in the Rai1- Tg mouse heart, liver, and kidney found unchanged expression of Clock and most of its downstream targets during both light and dark phases, suggesting an asynchronized circadian rhythm. Furthermore, examination of circadian gene expression in synchronized PTLS lymphoblasts revealed reduced transcripts of the Period ( PER1-3 ) family and normal expression of CRY1/2 . The finding that central circadian gene expression was altered while many peripheral circadian components were intact suggests a tissue-specific circadian uncoupling of the circadian machinery due to Rai1 overexpression. Overall, our results demonstrate that overexpression of RAI1 results in sleep deficiencies in individuals with PTLS due to a lack of properly regulated circadian machinery gene expression and highlight the importance of evaluating sleep concerns in individuals with PTLS.
Collapse
Affiliation(s)
- Sureni V Mullegama
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States.,Department of Pathology and Laboratory Medicine, University of California, Los Angeles, California, United States
| | - Joseph T Alaimo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Michael D Fountain
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Brooke Burns
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
| | - Amanda Hebert Balog
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
| | - Li Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States.,Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Sarah H Elsea
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States.,Department of Pathology and Laboratory Medicine, University of California, Los Angeles, California, United States.,Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
| |
Collapse
|
32
|
Late feeding in the active period decreases slow-wave activity. Life Sci 2016; 160:18-26. [DOI: 10.1016/j.lfs.2016.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/06/2016] [Accepted: 07/15/2016] [Indexed: 01/15/2023]
|
33
|
Verwey M, Dhir S, Amir S. Circadian influences on dopamine circuits of the brain: regulation of striatal rhythms of clock gene expression and implications for psychopathology and disease. F1000Res 2016; 5. [PMID: 27635233 PMCID: PMC5007753 DOI: 10.12688/f1000research.9180.1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2016] [Indexed: 12/18/2022] Open
Abstract
Circadian clock proteins form an autoregulatory feedback loop that is central to the endogenous generation and transmission of daily rhythms in behavior and physiology. Increasingly, circadian rhythms in clock gene expression are being reported in diverse tissues and brain regions that lie outside of the suprachiasmatic nucleus (SCN), the master circadian clock in mammals. For many of these extra-SCN rhythms, however, the region-specific implications are still emerging. In order to gain important insights into the potential behavioral, physiological, and psychological relevance of these daily oscillations, researchers have begun to focus on describing the neurochemical, hormonal, metabolic, and epigenetic contributions to the regulation of these rhythms. This review will highlight important sites and sources of circadian control within dopaminergic and striatal circuitries of the brain and will discuss potential implications for psychopathology and disease
. For example, rhythms in clock gene expression in the dorsal striatum are sensitive to changes in dopamine release, which has potential implications for Parkinson’s disease and drug addiction. Rhythms in the ventral striatum and limbic forebrain are sensitive to psychological and physical stressors, which may have implications for major depressive disorder. Collectively, a rich circadian tapestry has emerged that forces us to expand traditional views and to reconsider the psychopathological, behavioral, and physiological importance of these region-specific rhythms in brain areas that are not immediately linked with the regulation of circadian rhythms.
Collapse
Affiliation(s)
- Michael Verwey
- Center for Studies in Behavioural Neurobiology, FRQS Groupe de Recherche en Neurobiologie Comportementale, Concorida University, Montreal, Quebec, Canada
| | | | - Shimon Amir
- Center for Studies in Behavioural Neurobiology, FRQS Groupe de Recherche en Neurobiologie Comportementale, Concorida University, Montreal, Quebec, Canada
| |
Collapse
|
34
|
Targeting of the circadian clock via CK1δ/ε to improve glucose homeostasis in obesity. Sci Rep 2016; 6:29983. [PMID: 27439882 PMCID: PMC4954991 DOI: 10.1038/srep29983] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/24/2016] [Indexed: 12/21/2022] Open
Abstract
Growing evidence indicates that disruption of our internal timing system contributes to the incidence and severity of metabolic diseases, including obesity and type 2 diabetes. This is perhaps not surprising since components of the circadian clockwork are tightly coupled to metabolic processes across the body. In the current study, we assessed the impact of obesity on the circadian system in mice at a behavioural and molecular level, and determined whether pharmacological targeting of casein kinase 1δ and ε (CK1δ/ε), key regulators of the circadian clock, can confer metabolic benefit. We demonstrate that although behavioural rhythmicity was maintained in diet-induced obesity (DIO), gene expression profiling revealed tissue-specific alteration to the phase and amplitude of the molecular clockwork. Clock function was most significantly attenuated in visceral white adipose tissue (WAT) of DIO mice, and was coincident with elevated tissue inflammation, and dysregulation of clock-coupled metabolic regulators PPARα/γ. Further, we show that daily administration of a CK1δ/ε inhibitor (PF-5006739) improved glucose tolerance in both DIO and genetic (ob/ob) models of obesity. These data further implicate circadian clock disruption in obesity and associated metabolic disturbance, and suggest that targeting of the clock represents a therapeutic avenue for the treatment of metabolic disorders.
Collapse
|
35
|
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: 2.4] [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
|
36
|
Hutchison AT, Heilbronn LK. Metabolic impacts of altering meal frequency and timing – Does when we eat matter? Biochimie 2016; 124:187-197. [DOI: 10.1016/j.biochi.2015.07.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 07/25/2015] [Indexed: 12/26/2022]
|
37
|
Casiraghi LP, Alzamendi A, Giovambattista A, Chiesa JJ, Golombek DA. Effects of chronic forced circadian desynchronization on body weight and metabolism in male mice. Physiol Rep 2016; 4:4/8/e12743. [PMID: 27125665 PMCID: PMC4848717 DOI: 10.14814/phy2.12743] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 02/29/2016] [Indexed: 02/05/2023] Open
Abstract
Metabolic functions are synchronized by the circadian clock setting daily patterns of food intake, nutrient delivery, and behavioral activity. Here, we study the impact of chronic jet‐lag (CJL) on metabolism, and test manipulations aimed to overcome potential alterations. We recorded weight gain in C57Bl/6 mice under chronic 6 h advances or delays of the light–dark cycle every 2 days (ChrA and ChrD, respectively). We have previously reported ChrA, but not ChrD, to induce forced desynchronization of locomotor activity rhythms in mice (Casiraghi et al. 2012). Body weight was rapidly increased under ChrA, with animals tripling the mean weight gain observed in controls by day 10, and doubling it by day 30 (6% vs. 2%, and 15% vs. 7%, respectively). Significant increases in retroperitoneal and epidydimal adipose tissue masses (172% and 61%, respectively), adipocytes size (28%), and circulating triglycerides (39%) were also detected. Daily patterns of food and water intake were abolished under ChrA. In contrast, ChrD had no effect on body weight. Wheel‐running, housing of animals in groups, and restriction of food availability to hours of darkness prevented abnormal increase in body weight under ChrA. Our findings suggest that the observed alterations under ChrA may arise either from a direct effect of circadian disruption on metabolism, from desynchronization between feeding and metabolic rhythms, or both. Direction of shifts, timing of feeding episodes, and other reinforcing signals deeply affect the outcome of metabolic function under CJL. Such features should be taken into account in further studies of shift working schedules in humans.
Collapse
Affiliation(s)
- Leandro P Casiraghi
- Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes - CONICET. Bernal, Buenos Aires, Argentina
| | - Ana Alzamendi
- Unidad de Neuroendocrinología, IMBICE (CONICET-CICPBA), La Plata, Argentina
| | | | - Juan J Chiesa
- Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes - CONICET. Bernal, Buenos Aires, Argentina
| | - Diego A Golombek
- Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes - CONICET. Bernal, Buenos Aires, Argentina
| |
Collapse
|
38
|
Abstract
Robust circadian rhythms in metabolic processes have been described in both humans and animal models, at the whole body, individual organ, and even cellular level. Classically, these time-of-day-dependent rhythms have been considered secondary to fluctuations in energy/nutrient supply/demand associated with feeding/fasting and wake/sleep cycles. Renewed interest in this field has been fueled by studies revealing that these rhythms are driven, at least in part, by intrinsic mechanisms and that disruption of metabolic synchrony invariably increases the risk of cardiometabolic disease. The objectives of this paper are to provide a comprehensive review regarding rhythms in glucose, lipid, and protein/amino acid metabolism, the relative influence of extrinsic (eg, neurohumoral factors) versus intrinsic (eg, cell autonomous circadian clocks) mediators, the physiologic roles of these rhythms in terms of daily fluctuations in nutrient availability and activity status, as well as the pathologic consequences of dyssynchrony.
Collapse
Affiliation(s)
- Graham R McGinnis
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
39
|
Milev NB, Reddy AB. Circadian redox oscillations and metabolism. Trends Endocrinol Metab 2015; 26:430-7. [PMID: 26113283 PMCID: PMC5122445 DOI: 10.1016/j.tem.2015.05.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 05/26/2015] [Accepted: 05/27/2015] [Indexed: 12/12/2022]
Abstract
Circadian rhythms are 24h oscillations in physiology and behavior which allow organisms to anticipate and adapt to daily demands associated with the day/night cycle. The currently accepted model of the molecular clockwork is described as a transcriptional process composed of negative regulatory feedback loops. However, ample evidence underlines the important contribution of non-transcriptional and metabolic oscillations to cellular timekeeping. We summarize recent evidence pointing to the relationship between the transcriptional oscillator and metabolic redox state, with particular emphasis on the potential nodes of interaction. We highlight the intrinsic difficulty in segregating these two tightly coupled and interdependent processes, in living systems, and how disruption of their synchronicity impacts upon (patho)physiological processes as diverse as cardiovascular and metabolic disorders, aging, and cancer.
Collapse
Affiliation(s)
- Nikolay B Milev
- Department of Clinical Neurosciences, University of Cambridge Metabolic Research Laboratories, National Institute for Health Research (NIHR) Biomedical Research Centre, Wellcome Trust-Medical Research Council (MRC) Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Akhilesh B Reddy
- Department of Clinical Neurosciences, University of Cambridge Metabolic Research Laboratories, National Institute for Health Research (NIHR) Biomedical Research Centre, Wellcome Trust-Medical Research Council (MRC) Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK.
| |
Collapse
|
40
|
Dridi I, Ben-Cherif W, Aouam K, Haouas Z, Ben-Attia M, Reinberg A, Boughattas NA. Circadian variation in hepatic toxicity of the immunosuppressive agent “Mycophenolate Mofetil” in rats. BIOL RHYTHM RES 2015. [DOI: 10.1080/09291016.2015.1052648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
41
|
Abstract
Circadian rhythm, or daily oscillation, of behaviors and biological processes is a fundamental feature of mammalian physiology that has developed over hundreds of thousands of years under the continuous evolutionary pressure of energy conservation and efficiency. Evolution has fine-tuned the body's clock to anticipate and respond to numerous environmental cues in order to maintain homeostatic balance and promote survival. However, we now live in a society in which these classic circadian entrainment stimuli have been dramatically altered from the conditions under which the clock machinery was originally set. A bombardment of artificial lighting, heating, and cooling systems that maintain constant ambient temperature; sedentary lifestyle; and the availability of inexpensive, high-calorie foods has threatened even the most powerful and ancient circadian programming mechanisms. Such environmental changes have contributed to the recent staggering elevation in lifestyle-influenced pathologies, including cancer, cardiovascular disease, depression, obesity, and diabetes. This review scrutinizes the role of the body's internal clocks in the hard-wiring of circadian networks that have evolved to achieve energetic balance and adaptability, and it discusses potential therapeutic strategies to reset clock metabolic control to modern time for the benefit of human health.
Collapse
Affiliation(s)
- Zachary Gerhart-Hines
- Section for Metabolic Receptology (Z.G.-H.), Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; and Division of Endocrinology, Diabetes, and Metabolism (M.A.L.), Department of Medicine, Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Mitchell A Lazar
- Section for Metabolic Receptology (Z.G.-H.), Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; and Division of Endocrinology, Diabetes, and Metabolism (M.A.L.), Department of Medicine, Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
42
|
Wang K, Sun Y, Lin P, Song J, Zhao R, Li W, Hou X, Wang C, Wang L, Zhu P, Chen L. Liraglutide Activates AMPK Signaling and Partially Restores Normal Circadian Rhythm and Insulin Secretion in Pancreatic Islets in Diabetic Mice. Biol Pharm Bull 2015; 38:1142-9. [DOI: 10.1248/bpb.b15-00024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Kexin Wang
- Department of General Surgery, Qilu Hospital of Shandong University
| | - Yu Sun
- Department of Endocrinology, Qilu Hospital of Shandong University
| | - Peng Lin
- Department of Endocrinology, Qilu Hospital of Shandong University
| | - Jun Song
- Department of Endocrinology, Qilu Hospital of Shandong University
| | - Ruxing Zhao
- Department of Endocrinology, Qilu Hospital of Shandong University
| | - Wenjuan Li
- Department of Endocrinology, Qilu Hospital of Shandong University
| | - Xinguo Hou
- Department of Endocrinology, Qilu Hospital of Shandong University
| | - Chuan Wang
- Department of Endocrinology, Qilu Hospital of Shandong University
| | - Lingshu Wang
- Department of Endocrinology, Qilu Hospital of Shandong University
| | - Ping Zhu
- Department of Ophthalmology, College of Medicine, University of Florida
| | - Li Chen
- Department of Endocrinology, Qilu Hospital of Shandong University
- Institute of Endocrinology and Metabolism, Shandong University
| |
Collapse
|
43
|
Metabolic consequences of timed feeding in mice. Physiol Behav 2014; 128:188-201. [DOI: 10.1016/j.physbeh.2014.02.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/29/2014] [Accepted: 02/06/2014] [Indexed: 01/02/2023]
|
44
|
Navigatore-Fonzo LS, Delgado SM, Golini RS, Anzulovich AC. Circadian rhythms of locomotor activity and hippocampal clock genes expression are dampened in vitamin A–deficient rats. Nutr Res 2014; 34:326-35. [DOI: 10.1016/j.nutres.2014.02.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 01/29/2014] [Accepted: 02/03/2014] [Indexed: 11/24/2022]
|
45
|
Sunderram J, Sofou S, Kamisoglu K, Karantza V, Androulakis IP. Time-restricted feeding and the realignment of biological rhythms: translational opportunities and challenges. J Transl Med 2014; 12:79. [PMID: 24674294 PMCID: PMC3973614 DOI: 10.1186/1479-5876-12-79] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 03/10/2014] [Indexed: 12/15/2022] Open
Abstract
It has been argued that circadian dysregulation is not only a critical inducer and promoter of adverse health effects, exacerbating symptom burden, but also hampers recovery. Therefore understanding the health-promoting roles of regulating (i.e., restoring) circadian rhythms, thus suppressing harmful effects of circadian dysregulation, would likely improve treatment. At a critical care setting it has been argued that studies are warranted to determine whether there is any use in restoring circadian rhythms in critically ill patients, what therapeutic goals should be targeted, and how these could be achieved. Particularly interesting are interventional approaches aiming at optimizing the time of feeding in relation to individualized day-night cycles for patients receiving enteral nutrition, in an attempt to re-establish circadian patterns of molecular expression. In this short review we wish to explore the idea of transiently imposing (appropriate, but yet to be determined) circadian rhythmicity via regulation of food intake as a means of exploring rhythm-setting properties of metabolic cues in the context of improving immune response. We highlight some of the key elements associated with his complex question particularly as they relate to: a) stress and rhythmic variability; and b) metabolic entrainment of peripheral tissues as a possible intervention strategy through time-restricted feeding. Finally, we discuss the challenges and opportunities for translating these ideas to the bedside.
Collapse
Affiliation(s)
- Jag Sunderram
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Rutgers - Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Stavroula Sofou
- Biomedical Engineering Department, Rutgers University, Piscataway, NJ 08854, USA
- Chemical & Biochemical Engineering Department, Rutgers University, Piscataway, NJ 08854, USA
| | - Kubra Kamisoglu
- Chemical & Biochemical Engineering Department, Rutgers University, Piscataway, NJ 08854, USA
| | - Vassiliki Karantza
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick 08901, USA
| | - Ioannis P Androulakis
- Biomedical Engineering Department, Rutgers University, Piscataway, NJ 08854, USA
- Chemical & Biochemical Engineering Department, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
46
|
Abstract
Animal studies of delayed eating have provided useful information regarding the potential relationship between nighttime eating and increased weight and metabolic dysregulation, which occur in the absence of increased locomotion or increased caloric intake. We first review recent studies detailing these relationships and possible mechanisms in rodents. We then examine human data showing that sleep restriction leads to increased energy intake and weight gain, followed by a review of the human phenotype of delayed eating, night eating syndrome, and its relation to weight and metabolism. Finally, we examine human experimental studies of delayed eating and discuss preliminary data that show slight weight gain, dysfunction in energy expenditure, and abnormalities in the circadian rhythms of appetitive, stress, and sleep hormones. Well-controlled, longer-term experimental studies in humans are warranted to test the effect of delayed eating without sleep restriction to clarify whether limiting or eliminating nighttime eating could lead to weight loss and significantly improve related disorders, such as diabetes and heart disease, over time.
Collapse
Affiliation(s)
- Kelly C Allison
- Center for Weight and Eating Disorders, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 3535 Market St., Suite 3027, Philadelphia, PA, 19104, USA.
| | - Namni Goel
- Division of Sleep and Chronobiology, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 1017 Blockley Hall, 423 Guardian Drive, Philadelphia, PA, 19104, USA.
| | - Rexford S Ahima
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
47
|
Mirzaei K, Xu M, Qi Q, de Jonge L, Bray GA, Sacks F, Qi L. Variants in glucose- and circadian rhythm-related genes affect the response of energy expenditure to weight-loss diets: the POUNDS LOST Trial. Am J Clin Nutr 2014; 99:392-9. [PMID: 24335056 PMCID: PMC3893729 DOI: 10.3945/ajcn.113.072066] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Circadian rhythm has been shown to be related to glucose metabolism and risk of diabetes, probably through effects on energy balance. Recent genome-wide association studies identified variants in circadian rhythm-related genes (CRY2 and MTNR1B) associated with glucose homeostasis. OBJECTIVE We tested whether CRY2 and MTNR1B genotypes affected changes in measures of energy expenditure in response to a weight-loss diet intervention in a 2-y randomized clinical trial, the POUNDS (Preventing Overweight Using Novel Dietary Strategies) LOST Trial. DESIGN The variants CRY2 rs11605924 (n = 721) and MTNR1B rs10830963 (n = 722) were genotyped in overweight or obese adults who were randomly assigned to 1 of 4 weight-loss diets that differed in their proportions of macronutrients. Respiratory quotient (RQ) and resting metabolic rate (RMR) were measured. RESULTS By 2 y of diet intervention, the A allele of CRY2 rs11605924 was significantly associated with a greater reduction in RQ (P = 0.03) and a greater increase in RMR and RMR/kg (both P = 0.04). The G allele of MTNR1B rs10830963 was significantly associated with a greater increase in RQ (P = 0.01) but was not related to changes in RMR and RMR/kg. In addition, we found significant gene-diet fat interactions for both CRY2 (P-interaction = 0.02) and MTNR1B (P-interaction < 0.001) in relation to 2-y changes in RQ. CONCLUSIONS Our data indicate that variants in the circadian-related genes CRY2 and MTNR1B may affect long-term changes in energy expenditure, and dietary fat intake may modify the genetic effects. This trial was registered at www.clinicaltrials.gov as NCT00072995.
Collapse
Affiliation(s)
- Khadijeh Mirzaei
- Department of Nutrition, Harvard School of Public Health, Boston, MA (KM, MX, QQ, FS, and LQ); the Pennington Biomedical Research Center of the Louisiana State University System, Baton Rouge, LA (LdJ and GAB); and the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (LQ)
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
In mammals, a network of circadian clocks regulates 24-h rhythms of behavior and physiology. Circadian disruption promotes obesity and the development of obesity-associated disorders, but it remains unclear to which extent peripheral tissue clocks contribute to this effect. To reveal the impact of the circadian timing system on lipid metabolism, blood and adipose tissue samples from wild-type, ClockΔ19, and Bmal1(-/-) circadian mutant mice were subjected to biochemical assays and gene expression profiling. We show diurnal variations in lipolysis rates and release of free fatty acids (FFAs) and glycerol into the blood correlating with rhythmic regulation of two genes encoding the lipolysis pacemaker enzymes, adipose triglyceride (TG) lipase and hormone-sensitive lipase, by self-sustained adipocyte clocks. Circadian clock mutant mice show low and nonrhythmic FFA and glycerol blood content together with decreased lipolysis rates and increased sensitivity to fasting. Instead circadian clock disruption promotes the accumulation of TGs in white adipose tissue (WAT), leading to increased adiposity and adipocyte hypertrophy. In summary, circadian modulation of lipolysis rates regulates the availability of lipid-derived energy during the day, suggesting a role for WAT clocks in the regulation of energy homeostasis.
Collapse
Affiliation(s)
- Anton Shostak
- Circadian Rhythms Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Judit Meyer-Kovac
- Circadian Rhythms Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Henrik Oster
- Circadian Rhythms Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
- Medical Department I, University of Lübeck, Lübeck, Germany
- Corresponding author: Henrik Oster,
| |
Collapse
|
49
|
Hibi M, Masumoto A, Naito Y, Kiuchi K, Yoshimoto Y, Matsumoto M, Katashima M, Oka J, Ikemoto S. Nighttime snacking reduces whole body fat oxidation and increases LDL cholesterol in healthy young women. Am J Physiol Regul Integr Comp Physiol 2013; 304:R94-R101. [DOI: 10.1152/ajpregu.00115.2012] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The increase in obesity and lipid disorders in industrialized countries may be due to irregular eating patterns. Few studies have investigated the effects of nighttime snacking on energy metabolism. We examined the effects of nighttime snacking for 13 days on energy metabolism. Eleven healthy women (means ± SD; age: 23 ± 1 yr; body mass index: 20.6 ± 2.6 kg/m2) participated in this randomized crossover trial for a 13-day intervention period. Subjects consumed a specified snack (192.4 ± 18.3 kcal) either during the daytime (10:00) or the night time (23:00) for 13 days. On day 14, energy metabolism was measured in a respiratory chamber without snack consumption. An oral glucose tolerance test was performed on day 15. Relative to daytime snacking, nighttime snacking significantly decreased fat oxidation (daytime snacking: 52.0 ± 13.6 g/day; nighttime snacking: 45.8 ± 14.0 g/day; P = 0.02) and tended to increase the respiratory quotient (daytime snacking: 0.878 ± 0.022; nighttime snacking: 0.888 ± 0.021; P = 0.09). The frequency of snack intake and energy intake, body weight, and energy expenditure were not affected. Total and low-density lipoprotein (LDL) cholesterol significantly increased after nighttime snacking (152 ± 26 mg/dl and 161 ± 29 mg/dl; P = 0.03 and 76 ± 20 mg/dl and 83 ± 24 mg/dl; P = 0.01, respectively), but glucose and insulin levels after the glucose load were not affected. Nighttime snacking increased total and LDL cholesterol and reduced fat oxidation, suggesting that eating at night changes fat metabolism and increases the risk of obesity.
Collapse
Affiliation(s)
- Masanobu Hibi
- Health Care Food Research Laboratories, Kao Corporation, Tokyo, Japan
| | - Ayumi Masumoto
- Department of Nutrition and Food Science, Ochanomizu University, Tokyo, Japan
| | - Yuri Naito
- Department of Nutrition and Food Science, Ochanomizu University, Tokyo, Japan
| | - Kahori Kiuchi
- Department of Nutrition and Food Science, Ochanomizu University, Tokyo, Japan
| | - Yayoi Yoshimoto
- Health Care Food Research Laboratories, Kao Corporation, Tokyo, Japan
| | - Mai Matsumoto
- Department of Human Nutrition, Seitoku University, Chiba, Japan
| | | | - Jun Oka
- Department of Home Economics, Tokyo Kasei University, Tokyo, Japan; and
| | - Shinji Ikemoto
- Department of Human Nutrition, Seitoku University, Chiba, Japan
| |
Collapse
|
50
|
Klöting I, Bahr J, Wilke B, Lange J. Light rhythm and diet differently influence facets of the metabolic syndrome in WOKW rats. Lab Anim 2013; 47:31-5. [PMID: 23287511 DOI: 10.1258/la.2012.011095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
It has previously been shown that high-calorie diet alters the function of the mammalian circadian clock and that obesity has an influence on circadian organization of hormone secretion. That prompted us to test whether inbred Wistar Ottawa Karlsburg W (RT1(u)) (WOKW) rats developing facets of the metabolic syndrome show changes in their metabolic profiles under different feeding conditions (high-fat, high-sugar versus control diet) and under two different 12 h:12 h light-dark (LD) cycles. At the age of four weeks, these rats were divided into four groups. Groups 1 and 2 were kept under initial LD cycle (lights on at 05:00 h). Group 1 was fed with a normal rat diet while group 2 received a high-fat, high-sugar diet from 10 up to the age of 21 weeks. Groups 3 and 4 were kept under a shifted LD cycle (lights on at 11:00 h). Group 3 was given a normal diet while group 4 received a high-fat, high-sugar diet from an age like groups 1 and 2. Several metabolic traits were studied during the observation period of 21 weeks. The blood samples were obtained 2 h before lights off. Body weight gain (P < 0.001), leptin (P < 0.001), triglycerides (P < 0.001) and cholesterol (P < 0.05) were significantly reduced in group 4 versus group 2, but comparable between control groups (1 versus 3). The insulin concentrations were reduced in groups 3 and 4 versus groups 1 and 2 without effect of diet. In conclusion, the results provide evidence that light conditions influence diet induced changes in phenotypic traits like body weight gain, lipids as well as hormone levels (insulin and leptin) in WOKW rats.
Collapse
Affiliation(s)
- Ingrid Klöting
- Department of Laboratory Animal Science, Medical Faculty, University of Greifswald, D-17495 Karlsburg, Germany.
| | | | | | | |
Collapse
|