1
|
Ehichioya DE, Masud I, Taufique ST, Shen M, Farah S, Yamazaki S. Multiple oscillators underlie circadian food anticipation in mice. Neurobiol Sleep Circadian Rhythms 2025; 18:100116. [PMID: 40124171 PMCID: PMC11929000 DOI: 10.1016/j.nbscr.2025.100116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 02/21/2025] [Accepted: 02/23/2025] [Indexed: 03/25/2025] Open
Abstract
Circadian pacemakers orchestrate behavioral and physiological rhythms, enabling organisms to anticipate daily reoccurring environmental events such as light and dark, temperature changes, and food availability. When nocturnal rodents are subjected to time-restricted feeding during the day, they typically display food anticipatory activity several hours before mealtime. Upon releasing mice to ad libitum feeding, this anticipatory activity is abolished immediately but, following food deprivation, reappears at approximately the same time. However, the mechanism by which rodents retain this time memory of food availability during ad libitum feeding has remained elusive. We utilized the open-source Feeding Experimentation Device 3 (FED3) to measure food-seeking nose-poking behavior. We programmed the FED3 to dispense a pellet by a single left nose-poke, but not by right poke. During daytime restricted feeding, mice exhibited strong anticipatory nose-poking a few hours prior to the daytime meal in both rewarded left and unrewarded right pokes. In addition, mice also exhibited elevation of both rewarded and unrewarded pokes at night, coinciding with mice's previous habitual feeding time. Following ad libitum feeding, rewarded daytime nose-poking gradually moved back to habitual nighttime. However, following food deprivation, anticipatory poking immediately reappeared during the day and night, coinciding with the times of previous daytime restricted feeding and nighttime habitual feeding. Under ad libitum feeding, db/db mice didn't exhibit a clear daily rhythm in food intake. However, these mice exhibited robust food anticipation in both nose-pokes and activity during daytime restricted feeding. Following release back to ad libitum feeding, db/db mice poked sporadically during the day and night, and following food deprivation, anticipation promptly reappeared. These data suggest that there are at least two oscillators underlying food anticipation: one oscillator with a phase that changes according to food availability, and another oscillator with a phase unaffected by feeding conditions. In db/db mice, the first oscillator is likely impaired, and the second oscillator is unaffected.
Collapse
Affiliation(s)
- David E. Ehichioya
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ishrat Masud
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
- Jack E. Singley Academy, 4601 N MacArthur Blvd, Irving, TX, USA
| | | | - Melody Shen
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sofia Farah
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
| | - Shin Yamazaki
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
- Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
2
|
Zhang Q, Litwin C, Dietert K, Tsialtas I, Chen WH, Li Z, Koronowski KB. Frequent Shifts During Chronic Jet Lag Uncouple Liver Rhythms From the Light Cycle in Male Mice. J Biol Rhythms 2025; 40:194-207. [PMID: 39773136 PMCID: PMC11915764 DOI: 10.1177/07487304241311328] [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] [Indexed: 01/11/2025]
Abstract
Circadian disruption is pervasive in modern society and associated with increased risk of disease. Chronic jet lag paradigms are popular experimental tools aiming to emulate human circadian disruption experienced during rotating and night shift work. Chronic jet lag induces metabolic phenotypes tied to liver and systemic functions, yet lack of a clear definition for how rhythmic physiology is impaired under these conditions hinders the ability to identify the underlying molecular mechanisms. Here, we compared 2 common chronic jet lag paradigms and found that neither induced arrythmicity of the liver and each had distinct effects on rhythmicity. Instead, more frequent 8-h forward shifts of the light schedule induced more severe misalignment and non-fasted hyperglycemia. Every other day shifts eventually uncoupled behavioral and hepatic rhythms from the light cycle, reminiscent of free-running conditions. These results point to misalignment, not arrhythmicity, as the initial disturbance tied to metabolic dysfunction in environmental circadian disruption and highlight considerations for the interpretation and design of chronic jet lag studies.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, Texas
| | - Christopher Litwin
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, Texas
| | - Kristi Dietert
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, Texas
| | - Ioannis Tsialtas
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, Texas
| | - Wan Hsi Chen
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, Texas
| | - Zhihong Li
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, Texas
| | - Kevin B Koronowski
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
3
|
Chaikin CA, Thakkar AV, Steffeck AWT, Pfrender EM, Hung K, Zhu P, Waldeck NJ, Nozawa R, Song W, Futtner CR, Quattrocelli M, Bass J, Ben-Sahra I, Peek CB. Control of circadian muscle glucose metabolism through the BMAL1-HIF axis in obesity. Proc Natl Acad Sci U S A 2025; 122:e2424046122. [PMID: 40127275 DOI: 10.1073/pnas.2424046122] [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: 11/18/2024] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
Disruptions of circadian rhythms are widespread in modern society and lead to accelerated and worsened symptoms of metabolic syndrome. In healthy mice, the circadian clock factor BMAL1 is required for skeletal muscle function and metabolism. However, the importance of muscle BMAL1 in the development of metabolic diseases, such as diet-induced obesity (DIO), remains unclear. Here, we demonstrate that skeletal muscle-specific BMAL1-deficient mice exhibit worsened glucose tolerance upon high-fat diet feeding, despite no evidence of increased weight gain. Metabolite profiling from Bmal1-deficient muscles revealed impaired glucose utilization specifically at early steps in glycolysis that dictate the switch between anabolic and catabolic glucose fate. We provide evidence that this is due to abnormal control of the nutrient stress-responsive hypoxia-inducible factor (HIF) pathway. Genetic HIF1α stabilization in muscle Bmal1-deficient mice restores glucose tolerance and expression of 217/736 dysregulated genes during DIO, including glycolytic enzymes. Together, these data indicate that during DIO, skeletal muscle BMAL1 is an important regulator of HIF-driven glycolysis and metabolic flexibility, which influences the development of high-fat-diet-induced glucose intolerance.
Collapse
Affiliation(s)
- Claire A Chaikin
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Abhishek V Thakkar
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Adam W T Steffeck
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Eric M Pfrender
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Kaitlyn Hung
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Pei Zhu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Nathan J Waldeck
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Rino Nozawa
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Weimin Song
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Christopher R Futtner
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Mattia Quattrocelli
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Clara B Peek
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
4
|
Li H, Hu X, Zhang Y, Hou W, Wang W, Sun H. Relationship between dietary energy and macronutrient intake at dinner versus breakfast and biological aging and premature mortality: Assessment of 2003-2014 National Health and Nutrition Examination Survey participants. J Affect Disord 2025; 380:466-473. [PMID: 40154808 DOI: 10.1016/j.jad.2025.03.083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND The impact of dietary patterns on health and lifespan is well-established, yet the effects of meal timing on the aging process and risk of premature death remain unclear. This study aimed to investigate the association between the difference in energy and macronutrient intake at dinner versus breakfast and the risk of premature mortality and biological aging. METHODS Utilizing data from the National Health and Nutrition Examination Survey (NHANES) between 2003 and 2014, a cohort of 27,261 adults was examined. Dietary data were collected through 24-h dietary recalls, and Cox proportional hazards models and binary logistic regression models were used to assess the risk of premature death and indicators of biological aging. RESULTS Individuals with higher energy and protein intake at dinner compared to breakfast exhibited an increased risk of premature death and higher biological aging indicators. Isocaloric substitution of energy and macronutrients from breakfast to dinner significantly increased the risk of aging. CONCLUSION The difference in energy and macronutrient intake at dinner versus breakfast is closely associated with the risk of premature death and biological aging. The findings underscore the potential impact of meal timing on metabolic health and lifespan extension.
Collapse
Affiliation(s)
- Hui Li
- Department of Epidemiology, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin 150081, Heilongjiang, China
| | - Xierong Hu
- Department of Epidemiology, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin 150081, Heilongjiang, China
| | - Yue Zhang
- Department of Epidemiology, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin 150081, Heilongjiang, China
| | - Wanying Hou
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin 150081, Heilongjiang, China
| | - Weiqi Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin 150081, Heilongjiang, China
| | - Hongru Sun
- Department of Epidemiology, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin 150081, Heilongjiang, China.
| |
Collapse
|
5
|
Gazit Shimoni N, Tose AJ, Seng C, Jin Y, Lukacsovich T, Yang H, Verharen JPH, Liu C, Tanios M, Hu E, Read J, Tang LW, Lim BK, Tian L, Földy C, Lammel S. Changes in neurotensin signalling drive hedonic devaluation in obesity. Nature 2025:10.1038/s41586-025-08748-y. [PMID: 40140571 DOI: 10.1038/s41586-025-08748-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/06/2025] [Indexed: 03/28/2025]
Abstract
Calorie-rich foods, particularly those that are high in fat and sugar, evoke pleasure in both humans and animals1. However, prolonged consumption of such foods may reduce their hedonic value, potentially contributing to obesity2-4. Here we investigated this phenomenon in mice on a chronic high-fat diet (HFD). Although these mice preferred high-fat food over regular chow in their home cages, they showed reduced interest in calorie-rich foods in a no-effort setting. This paradoxical decrease in hedonic feeding has been reported previously3-7, but its neurobiological basis remains unclear. We found that in mice on regular diet, neurons in the lateral nucleus accumbens (NAcLat) projecting to the ventral tegmental area (VTA) encoded hedonic feeding behaviours. In HFD mice, this behaviour was reduced and uncoupled from neural activity. Optogenetic stimulation of the NAcLat→VTA pathway increased hedonic feeding in mice on regular diet but not in HFD mice, though this behaviour was restored when HFD mice returned to a regular diet. HFD mice exhibited reduced neurotensin expression and release in the NAcLat→VTA pathway. Furthermore, neurotensin knockout in the NAcLat and neurotensin receptor blockade in the VTA each abolished optogenetically induced hedonic feeding behaviour. Enhancing neurotensin signalling via overexpression normalized aspects of diet-induced obesity, including weight gain and hedonic feeding. Together, our findings identify a neural circuit mechanism that links the devaluation of hedonic foods with obesity.
Collapse
Affiliation(s)
- Neta Gazit Shimoni
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Amanda J Tose
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Charlotte Seng
- Brain Research Institute, Faculties of Medicine and Science, University of Zurich, Zürich, Switzerland
| | - Yihan Jin
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Davis, CA, USA
- Max Planck Florida Institute For Neuroscience, Jupiter, FL, USA
| | - Tamás Lukacsovich
- Brain Research Institute, Faculties of Medicine and Science, University of Zurich, Zürich, Switzerland
| | - Hongbin Yang
- Department of Neurobiology and Department of Affiliated Mental Health Center of Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Jeroen P H Verharen
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Christine Liu
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Michael Tanios
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Eric Hu
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Jonathan Read
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Lilly W Tang
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Byung Kook Lim
- Division of Biological Sciences, University of California San Diego, San Diego, CA, USA
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Davis, CA, USA
- Max Planck Florida Institute For Neuroscience, Jupiter, FL, USA
| | - Csaba Földy
- Brain Research Institute, Faculties of Medicine and Science, University of Zurich, Zürich, Switzerland
| | - Stephan Lammel
- Department of Neuroscience and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA.
| |
Collapse
|
6
|
Leinweber B, Pilorz V, Olejniczak I, Skrum L, Begemann K, Heyde I, Stenger S, Sadik CD, Oster H. Bmal1 deficiency in neutrophils alleviates symptoms induced by high-fat diet. iScience 2025; 28:112038. [PMID: 40124497 PMCID: PMC11930374 DOI: 10.1016/j.isci.2025.112038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/24/2024] [Accepted: 02/12/2025] [Indexed: 03/25/2025] Open
Abstract
Physiological processes, including metabolism and immune responses, are generated by the circadian clock, driven by clock genes. Disrupting circadian rhythms through a high-fat diet promotes obesity and inflammation. Studies show that deleting the clock gene, brain, and muscle ARNT-like 1 (Bmal1) in adipose tissue leads to overeating and weight gain. We now show that Bmal1 deletion in neutrophils protects against diet-induced obesity and reduces inflammatory macrophage infiltration into epididymal white adipose tissue (eWAT), despite increased food intake over 20 weeks of a high-fat diet. This protection is linked to enhanced energy expenditure, increased UCP1 expression in iBAT, improved insulin sensitivity, and altered expression of genes encoding chemokine receptors CXCR2, CXCR4, and the ligand Cxcl2 in eWAT. Our findings reveal a key role of Bmal1 in neutrophils in regulating high-fat diet-induced adipose inflammation and emphasize circadian regulation's importance in immuno-metabolic function.
Collapse
Affiliation(s)
- Brinja Leinweber
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Violetta Pilorz
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Iwona Olejniczak
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Ludmila Skrum
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Kimberly Begemann
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Isabel Heyde
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Sarah Stenger
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Christian David Sadik
- University of Lübeck, Department of Dermatology, Allergy, and Venereology Ratzeburger Allee, 23562 Luebeck, Germany
| | - Henrik Oster
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| |
Collapse
|
7
|
Douglass AM, Kucukdereli H, Madara JC, Wang D, Wu C, Lowenstein ED, Tao J, Lowell BB. Acute and circadian feedforward regulation of agouti-related peptide hunger neurons. Cell Metab 2025; 37:708-722.e5. [PMID: 39719709 PMCID: PMC11885038 DOI: 10.1016/j.cmet.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/26/2024] [Accepted: 11/12/2024] [Indexed: 12/26/2024]
Abstract
When food is freely available, eating occurs without energy deficit. While agouti-related peptide (AgRP) neurons are likely involved, their activation is thought to require negative energy balance. To investigate this, we implemented long-term, continuous in vivo fiber-photometry recordings in mice. We discovered new forms of AgRP neuron regulation, including fast pre-ingestive decreases in activity and unexpectedly rapid activation by fasting. Furthermore, AgRP neuron activity has a circadian rhythm that peaks concurrent with the daily feeding onset. Importantly, this rhythm persists when nutrition is provided via constant-rate gastric infusions. Hence, it is not secondary to a circadian feeding rhythm. The AgRP neuron rhythm is driven by the circadian clock, the suprachiasmatic nucleus (SCN), as SCN ablation abolishes the circadian rhythm in AgRP neuron activity and feeding. The SCN activates AgRP neurons via excitatory afferents from thyrotrophin-releasing hormone-expressing neurons in the dorsomedial hypothalamus (DMHTrh neurons) to drive daily feeding rhythms.
Collapse
Affiliation(s)
- Amelia M Douglass
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Hakan Kucukdereli
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Joseph C Madara
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Daqing Wang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Chen Wu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Elijah D Lowenstein
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jenkang Tao
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Program in Neuroscience, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Duan J, Karri SS, Forouzesh K, Mortimer T, Plikus MV, Benitah SA, Takahashi JS, Andersen B. Designing and Evaluating Circadian Experiments on Mouse Skin. J Invest Dermatol 2025; 145:484-493. [PMID: 39891645 DOI: 10.1016/j.jid.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 02/03/2025]
Abstract
All skin layers and cutaneous appendages harbor a robust circadian clock, whose phase is under the influence of light through the central clock in the suprachiasmatic nucleus. The skin clock coordinates fundamental biological processes, including metabolism and stem cell activation. It also prominently modulates activity of skin-resident immune cells and the inflammatory response. Numerous diurnally regulated genes in the skin have been implicated in skin diseases in GWASs. Therefore, the mouse skin is a powerful model for understanding the diverse roles of circadian biology in maintaining tissue health and the initiation and propagation of disease states. When planning experiments to study the circadian biology of mouse skin, multiple technical and biological factors must be carefully considered. In this paper, we provide comprehensive guidance on the general circadian experimental design and associated housing for the mice. We highlight the importance of aligning sample collection with the desired hair cycle stage and animal age. We introduce methods to disrupt the clock in the skin, including altering light and feeding schedules as well as using transgenic mouse models. Finally, we discuss the use of transcriptomic data, both bulk and single cell, for circadian studies.
Collapse
Affiliation(s)
- Junyan Duan
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA; The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA
| | - Satya Swaroop Karri
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California, USA; Division of Endocrinology, Department of Medicine, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Kiarash Forouzesh
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California, USA; Division of Endocrinology, Department of Medicine, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Thomas Mortimer
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Maksim V Plikus
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA; The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA; Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona, Spain; Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bogi Andersen
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA; Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California, USA; Division of Endocrinology, Department of Medicine, School of Medicine, University of California, Irvine, Irvine, California, USA.
| |
Collapse
|
9
|
Deota S, Pendergast JS, Kolthur-Seetharam U, Esser KA, Gachon F, Asher G, Dibner C, Benitah SA, Escobar C, Muoio DM, Zhang EE, Hotamışlıgil GS, Bass J, Takahashi JS, Rabinowitz JD, Lamia KA, de Cabo R, Kajimura S, Longo VD, Xu Y, Lazar MA, Verdin E, Zierath JR, Auwerx J, Drucker DJ, Panda S. The time is now: accounting for time-of-day effects to improve reproducibility and translation of metabolism research. Nat Metab 2025; 7:454-468. [PMID: 40097742 DOI: 10.1038/s42255-025-01237-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
The constant expansion of the field of metabolic research has led to more nuanced and sophisticated understanding of the complex mechanisms that underlie metabolic functions and diseases. Collaborations with scientists of various fields such as neuroscience, immunology and drug discovery have further enhanced the ability to probe the role of metabolism in physiological processes. However, many behaviours, endocrine and biochemical processes, and the expression of genes, proteins and metabolites have daily ~24-h biological rhythms and thus peak only at specific times of the day. This daily variation can lead to incorrect interpretations, lack of reproducibility across laboratories and challenges in translating preclinical studies to humans. In this Review, we discuss the biological, environmental and experimental factors affecting circadian rhythms in rodents, which can in turn alter their metabolic pathways and the outcomes of experiments. We recommend that these variables be duly considered and suggest best practices for designing, analysing and reporting metabolic experiments in a circadian context.
Collapse
Affiliation(s)
- Shaunak Deota
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Ullas Kolthur-Seetharam
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
- Tata Institute of Fundamental Research, Hyderabad, India
| | - Karyn A Esser
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - Frédéric Gachon
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Gad Asher
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Charna Dibner
- Department of Surgery and Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute for Science and Technology, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Carolina Escobar
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Deborah M Muoio
- Departments of Medicine and Pharmacology & Cancer Biology, Duke Molecular Physiology Institute, Durham, NC, USA
| | | | - Gökhan S Hotamışlıgil
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Katja A Lamia
- Department of Molecular and Cellular Biology and Department of Molecular Medicine, the Scripps Research Institute, La Jolla, CA, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Shingo Kajimura
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, USA
| | - Valter D Longo
- Longevity Institute, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- AIRC Institute of Molecular Oncology, Italian Foundation for Cancer Research Institute of Molecular Oncology, Milan, Italy
| | - Ying Xu
- CAM-SU Genomic Resource Center, Soochow University, Suzhou, China
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity and Metabolism and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Daniel J Drucker
- The Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and the Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
10
|
Qiu J, Khedr MA, Pan M, Ferreira CR, Chen J, Snyder MM, Ajuwon KM, Yue F, Kuang S. Ablation of FAM210A in Brown Adipocytes of Mice Exacerbates High-Fat Diet-Induced Metabolic Dysfunction. Diabetes 2025; 74:282-294. [PMID: 39602358 PMCID: PMC11842609 DOI: 10.2337/db24-0294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
Thermogenesis of brown adipose tissue (BAT) provides metabolic benefits against pathologic conditions, such as type 2 diabetes, obesity, cardiovascular disease, and cancer. The thermogenic function of BAT relies on mitochondria, but whether mitochondrial remodeling is required for the beneficial effects of BAT remains unclear. We recently identified FAM210A as a BAT-enriched mitochondrial protein essential for cold-induced thermogenesis through the modulation of OPA1-dependent cristae remodeling. Here, we report a key role of FAM210A in the systemic response to a high-fat diet (HFD). We discovered that an HFD suppressed FAM210A expression, associated with excessive OPA1 cleavage in BAT. Ucp1-Cre-driven BAT-specific Fam210a knockout (Fam210aUKO) similarly elevated OPA1 cleavage, accompanied by whitening of BAT. When subjected to an HFD, Fam210aUKO mice gained similar fat mass as sibling control mice but developed glucose intolerance, insulin resistance, and liver steatosis. The metabolic dysfunction was associated with overall increased lipid content in both the liver and BAT. Additionally, Fam210aUKO leads to inflammation in white adipose tissue. These data demonstrate that FAM210A in BAT is necessary for counteracting HFD-induced metabolic dysfunction but not obesity. ARTICLE HIGHLIGHTS FAM210A regulates cold-induced mitochondrial remodeling through control of OPA1 cleavage, but whether it also plays a role in high-fat diet (HFD)-induced cristae remodeling is unknown. We asked if an HFD would alter the FAM210A level and OPA1 cleavage in brown adipose tissue (BAT) and how FAM210A loss of function would affect diet-induced obesity in mice. We found that an HFD diminished FAM210A expression and accelerated OPA1 cleavage in BAT, and Fam210a knockout exacerbated HFD-induced whitening of BAT, cold intolerance, liver steatosis, white adipose tissue inflammation, and metabolic dysfunction. Our work reveals a physiologic role of FAM210A-mediated BAT mitochondrial remodeling in systemic adaptation to an HFD and suggests that BAT mitochondria may be targeted to treat diet-induced metabolic dysfunction.
Collapse
Affiliation(s)
- Jiamin Qiu
- Department of Animal Sciences, Purdue University, West Lafayette, IN
| | - Mennatallah A. Khedr
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Orthopaedic Surgery, School of Medicine, Duke University, Durham, NC
| | - Meijin Pan
- Department of Animal Sciences, Purdue University, West Lafayette, IN
| | | | - Jingjuan Chen
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Orthopaedic Surgery, School of Medicine, Duke University, Durham, NC
| | - Madigan M. Snyder
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Biological Sciences, Purdue University, West Lafayette, IN
| | - Kolapo M. Ajuwon
- Department of Animal Sciences, Purdue University, West Lafayette, IN
| | - Feng Yue
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Animal Sciences, University of Florida, Gainesville, FL
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Orthopaedic Surgery, School of Medicine, Duke University, Durham, NC
- Institute for Cancer Research, Purdue University, West Lafayette, IN
| |
Collapse
|
11
|
Sagun E, Akyol A, Kaymak C. Chrononutrition in Critical Illness. Nutr Rev 2025; 83:e1146-e1157. [PMID: 38904422 DOI: 10.1093/nutrit/nuae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024] Open
Abstract
Circadian rhythms in humans are biological rhythms that regulate various physiological processes within a 24-hour time frame. Critical illness can disrupt the circadian rhythm, as can environmental and clinical factors, including altered light exposure, organ replacement therapies, disrupted sleep-wake cycles, noise, continuous enteral feeding, immobility, and therapeutic interventions. Nonpharmacological interventions, controlling the ICU environment, and pharmacological treatments are among the treatment strategies for circadian disruption. Nutrition establishes biological rhythms in metabolically active peripheral tissues and organs through appropriate synchronization with endocrine signals. Therefore, adhering to a feeding schedule based on the biological clock, a concept known as "chrononutrition," appears to be vitally important for regulating peripheral clocks. Chrononutritional approaches, such as intermittent enteral feeding that includes overnight fasting and consideration of macronutrient composition in enteral solutions, could potentially restore circadian health by resetting peripheral clocks. However, due to the lack of evidence, further studies on the effect of chrononutrition on clinical outcomes in critical illness are needed. The purpose of this review was to discuss the role of chrononutrition in regulating biological rhythms in critical illness, and its impact on clinical outcomes.
Collapse
Affiliation(s)
- Eylul Sagun
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Hacettepe University, Ankara, 06100, Turkey
| | - Asli Akyol
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Hacettepe University, Ankara, 06100, Turkey
| | - Cetin Kaymak
- Gülhane Faculty of Medicine, Department of Anesthesiology and Reanimation, University of Health Sciences, Ankara Training and Research Hospital, Intensive Care Unit, Ankara, 06230, Turkey
| |
Collapse
|
12
|
Gou R, Chen L, Cheng Z, Cun J, Li G. Association of dietary intake of saturated fatty acids with obstructive sleep apnea: mediating effects of Life's Crucial 9. Front Nutr 2025; 12:1503815. [PMID: 40034735 PMCID: PMC11872719 DOI: 10.3389/fnut.2025.1503815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Obstructive sleep apnea (OSA) is a global public health issue. Life's Crucial 9 (LC9) is recognized as a powerful tool for assessing cardiovascular health. Although the etiology of OSA remains unclear, saturated fatty acids (SFAs) and cardiovascular health are increasingly regarded as a non-negligible element. This study aims to assess the association between dietary intake of SFAs and the risk of OSA, and the mediating effect of LC9. Methods Based on the National Health and Nutrition Examination Survey (NHANES), dietary questionnaires of participant were collected, and the average values of 24-h dietary recall data over 2 days were obtained. A continuous cross-sectional analysis with dietary energy adjustment was employed. Weighted multivariable logistic regression models were used to estimate the weighted odds ratios (ORs) and their 95% confidence intervals (CIs) for SFAs and OSA. Evaluate the mediating role of LC9 in the relationship between SFAs and OSA. Results A total of 13,563 participants aged 20 years and above were included in this study. The intakes of Sfa 4.0 and LC9 among participants with OSA were significantly lower than those in the normal population. After adjusting for confounding factors, total SFAs could increase the risk of OSA [Model 1, Q3, 0.03, 1.49 (1.03, 2.15); Model 2, Q3, 0.04, 1.47 (1.01, 2.13)]. It was emphasized that dietary intake of Sfa 12.0, Sfa 14.0, and Sfa 16.0 were protective factors for OSA, especially among participants aged 45-64 years and white individuals. Moreover, Sfa 12.0 exhibited a better protective effect in female participants [Q3, 0.04, 0.66 (0.45, 0.99)]. In addition, the cardiovascular health score - LC9 had a mediating effect in Sfa4.0 on OSA [Proportion of mediation: -0.035, 95% CI: (-0.058, -0.01); p= 0.002]. There was a nonlinear relationship between dietary intake of Sfa 12.0, Sfa 16.0, and Sfa 18.0 and OSA (P-Nonlinear = 0.013). Discussion These findings suggest that dietary mixtures of saturated fatty acids increase the risk of OSA. Among them, SFA 4:0 can increase the risk of OSA through the level of cardiovascular health. However, contrary to traditional beliefs, long-chain saturated fatty acids can reduce the risk of OSA.
Collapse
Affiliation(s)
- Ruoyu Gou
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Lili Chen
- Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Zeyi Cheng
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiawei Cun
- School of First Clinical Medical, Ningxia, Medical University, Yinchuan, Ningxia, China
| | - Guanghua Li
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, China
- School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
13
|
Duez H, Staels B. Circadian Disruption and the Risk of Developing Obesity. Curr Obes Rep 2025; 14:20. [PMID: 39939483 PMCID: PMC11821678 DOI: 10.1007/s13679-025-00610-6] [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] [Accepted: 01/22/2025] [Indexed: 02/14/2025]
Abstract
PURPOSE OF THE REVIEW This review summarizes recent evidence for a role of the clock in adipose tissue physiology and the impact of circadian desynchrony on the development of obesity. RECENT FINDINGS Circadian disruptions due to shift work, late time eating and nighttime light exposure are associated with obesity and its metabolic and cardiovascular consequences. Studies in mice harboring tissue-specific gain/loss of function mutations in clock genes revealed that the circadian clock acts on multiple pathways to control adipogenesis, lipogenesis/lipolysis and thermogenesis. Time-restricted eating (TRE), aligning feeding with the active period to restore clock function, represents a promising strategy to curb obesity. While TRE has shown clear benefits, especially in participants at higher cardiometabolic risk, current studies are limited in size and duration. Larger, well-controlled studies are warranted to conclusively assess the effects of TRE in relation to the metabolic status and gender. Field studies in shift-workers, comparing permanent night shift versus rotating shifts, are also necessary to identify the optimal time window for TRE.
Collapse
Affiliation(s)
- Hélène Duez
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000, Lille, France.
| | - Bart Staels
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000, Lille, France.
| |
Collapse
|
14
|
Yao N, Kinouchi K, Katoh M, Ashtiani KC, Abdelkarim S, Morimoto H, Torimitsu T, Kozuma T, Iwahara A, Kosugi S, Komuro J, Kato K, Tonomura S, Nakamura T, Itoh A, Yamaguchi S, Yoshino J, Irie J, Hashimoto H, Yuasa S, Satoh A, Mikami Y, Uchida S, Ueki T, Nomura S, Baldi P, Hayashi K, Itoh H. Maternal circadian rhythms during pregnancy dictate metabolic plasticity in offspring. Cell Metab 2025; 37:395-412.e6. [PMID: 39814018 PMCID: PMC11872692 DOI: 10.1016/j.cmet.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 04/29/2024] [Accepted: 12/04/2024] [Indexed: 01/18/2025]
Abstract
Tissue-level oscillation is achieved by tissue-intrinsic clocks along with network-dependent signals originating from distal organs and organismal behavior. Yet, it remains unexplored whether maternal circadian rhythms during pregnancy influence fetal rhythms and impact long-term susceptibility to dietary challenges in offspring. Here, we demonstrate that circadian disruption during pregnancy decreased placental and neonatal weight yet retained transcriptional and structural maturation. Intriguingly, diet-induced obesity was exacerbated in parallel with arrhythmic feeding behavior, hypothalamic leptin resistance, and hepatic circadian reprogramming in offspring of chronodisrupted mothers. In utero circadian desynchrony altered the phase-relationship between the mother and fetus and impacted placental efficiency. Temporal feeding restriction in offspring failed to fully prevent obesity, whereas the circadian alignment of caloric restriction with the onset of the active phase virtually ameliorated the phenotype. Thus, maternal circadian rhythms during pregnancy confer adaptive properties to metabolic functions in offspring and provide insights into the developmental origins of health and disease.
Collapse
Affiliation(s)
- Na Yao
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kenichiro Kinouchi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Manami Katoh
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Frontier Cardiovascular Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Sherif Abdelkarim
- Department of Computer Science, University of California, Irvine, Irvine, CA 92697, USA
| | - Hiroyuki Morimoto
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takuto Torimitsu
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takahide Kozuma
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akihide Iwahara
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shotaro Kosugi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; Health Center, Keio University, Yokohama, Japan
| | - Jin Komuro
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Kyosuke Kato
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shun Tonomura
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Toshifumi Nakamura
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Arata Itoh
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shintaro Yamaguchi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Jun Yoshino
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo, Japan; The Center for Integrated Kidney Research and Advance (IKRA), Faculty of Medicine, Shimane University, Izumo, Japan
| | - Junichiro Irie
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hisayuki Hashimoto
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Shinsuke Yuasa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan; Department of Cardiovascular Medicine, Academic Field, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Akiko Satoh
- Department of Integrative Physiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan; Department of Integrative Physiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shusaku Uchida
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takatoshi Ueki
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Frontier Cardiovascular Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Pierre Baldi
- Department of Computer Science, University of California, Irvine, Irvine, CA 92697, USA
| | - Kaori Hayashi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Itoh
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; Center for Preventive Medicine, Keio University, Tokyo, Japan.
| |
Collapse
|
15
|
Yang MY, Lin HYH, Chen YYM, Hu ML, Chen IY, Yang CH. Chronic low-dose REV-ERBs agonist SR9009 mitigates constant light-induced weight gain and insulin resistance via adipogenesis modulation. Biomed J 2025:100830. [PMID: 39800061 DOI: 10.1016/j.bj.2025.100830] [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: 11/08/2024] [Revised: 12/24/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Obesity and circadian rhythm disruption are significant global health concerns, contributing to an increased risk of metabolic disorders. Both adipose tissue and circadian rhythms play critical roles in maintaining energy homeostasis, and their dysfunction is closely linked to obesity. This study aimed to assess the effects of chronic low-dose SR9009, a REV-ERB ligand, on circadian disruption induced by constant light exposure in mice. MATERIAL AND METHODS Mice were exposed to constant light for eight weeks (LL mice), resulting in increased body weight, insulin resistance, white fat mass, and altered circadian clock gene expression. Low-dose SR9009 (10 mg/kg daily) was administered chronically to assess its impact on these metabolic disruptions. RESULTS LL mice treated with SR9009 for eight weeks showed reduced weight gain, insulin resistance, and white fat mass but no significant impact on overall energy homeostasis. SR9009 suppressed Bmal1 expression and restored Rev-erbα and Rev-erbβ expression in white and brown adipose tissue (WAT and BAT). In vitro studies using 3T3-L1 cells indicated that SR9009 inhibited adipogenesis, leading to further investigation in vivo. SR9009 restored ChREBP1a and Srebp-1c expression in BAT but did not affect inflammatory cytokine or adipokine gene expression, nor did it restore Fasn, Pparγ, and Prom1 expression in both WAT and BAT. CONCLUSIONS These findings suggest that SR9009 may be a potential therapeutic for preventing weight gain and insulin resistance caused by circadian disruptions, likely through adipogenesis inhibition, though its effects on other metabolic pathways remain limited at low doses.
Collapse
Affiliation(s)
- Ming-Yu Yang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hugo Y-H Lin
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan; Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Ywan M Chen
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Ming-Luen Hu
- Division of Hepatogestroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - I-Ya Chen
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chao-Hui Yang
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan.
| |
Collapse
|
16
|
Braunsperger A, Bauer M, Brahim CB, Seep L, Tischer D, Peitzsch M, Hasenauer J, Figueroa SH, Worthmann A, Heeren J, Dyar KA, Koehler K, Soriano-Arroquia A, Schönfelder M, Wackerhage H. Effects of time-of-day on the noradrenaline, adrenaline, cortisol and blood lipidome response to an ice bath. Sci Rep 2025; 15:1263. [PMID: 39779795 PMCID: PMC11711488 DOI: 10.1038/s41598-025-85304-8] [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: 09/08/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025] Open
Abstract
While the effect of time-of-day (morning versus evening) on hormones, lipids and lipolysis has been studied in relation to meals and exercise, there are no studies that have investigated the effects of time-of-day on ice bath induced hormone and lipidome responses. In this crossover-designed study, a group of six women and six men, 26 ± 5 years old, 176 ± 7 cm tall, weighing 75 ± 10 kg, and a BMI of 23 ± 2 kg/m2 had an ice bath (8-12 °C for 5 min) both in the morning and evening on separate days. Absence from intense physical exercise, nutrient intake and meal order was standardized in the 24 h prior the ice baths to account for confounders such as diet or exercise. We collected venous blood samples before and after (5 min and 30 min) the ice baths to measure hormones (noradrenaline, adrenaline, and cortisol) and lipid levels in plasma via liquid chromatography mass spectrometry shotgun lipidomics. We found that ice baths in the morning increase plasma fatty acids more than in the evening. Overall plasma lipid composition significantly differed in-between the morning and evening, and only in the morning ice bathing is accompanied by significantly increased plasma fatty acids from 5.1 ± 2.2% to 6.0 ± 2.4% (P = 0.029) 5 min after and to 6.3 ± 3.1% (P = 0.008) 30 min after. Noradrenaline was not affected by time-of-day and increased significantly immediately after the ice baths in the morning by 127 ± 2% (pre: 395 ± 158 pg/ml, post 5 min: 896 ± 562 pg/ml, P = 0.025) and in the evening by 144 ± 2% (pre: 385 ± 146 pg/ml, post 5 min: 937 ± 547 pg/ml, P = 0.015). Cortisol was generally higher in the morning than in the evening (pre: 179 ± 108 pg/ml versus 91 ± 59 pg/ml, P = 0.013; post 5 min: 222 ± 96 pg/ml versus 101 ± 52 pg/ml, P = 0.001; post 30 min: 190 ± 96 pg/ml versus 98 ± 54 pg/ml, P = 0.009). There was no difference in the hormonal and lipidome response to an ice bath between women and men. The main finding of the study was that noradrenaline, adrenaline, cortisol and plasma lipidome responses are similar after an ice bath in the morning and evening. However, ice baths in the morning increase plasma fatty acids more than in the evening.
Collapse
Affiliation(s)
- Alexander Braunsperger
- Professorship of Exercise Biology, Department Health and Sport Sciences, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany.
| | - Maximilian Bauer
- Professorship of Exercise Biology, Department Health and Sport Sciences, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Chaima Ben Brahim
- Professorship of Exercise Biology, Department Health and Sport Sciences, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Lea Seep
- Computational Biology, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Dominik Tischer
- Institute for Pharmacology and Toxicology, Biomedical Center, University of Bonn, Bonn, Germany
| | - Mirko Peitzsch
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Jan Hasenauer
- Computational Biology, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Helmholtz Center Munich, German Research Center for Environmental Health, Computational Health Center, Munich, Germany
| | - Sieglinde Hechenbichler Figueroa
- Professorship of Exercise, Nutrition and Health, Department Health and Sport Sciences, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Anna Worthmann
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kenneth A Dyar
- Metabolic Physiology, Institute for Diabetes and Cancer, Helmholtz Diabetes Center, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Karsten Koehler
- Professorship of Exercise, Nutrition and Health, Department Health and Sport Sciences, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Ana Soriano-Arroquia
- Institute for Pharmacology and Toxicology, Biomedical Center, University of Bonn, Bonn, Germany
| | - Martin Schönfelder
- Professorship of Exercise Biology, Department Health and Sport Sciences, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Henning Wackerhage
- Professorship of Exercise Biology, Department Health and Sport Sciences, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| |
Collapse
|
17
|
Pati P, De Miguel C, Paul JR, Zhang D, Colson J, Allan JM, Edell CJ, Rhoads MK, Dunaway LS, Biswal SN, Zhong Y, Sedaka R, Millender-Swain T, Bailey SM, Gamble KL, Pollock DM, Pollock JS. Time-restricted feeding reduces cardiovascular disease risk in obese mice. JCI Insight 2025; 10:e160257. [PMID: 39812779 PMCID: PMC11949066 DOI: 10.1172/jci.insight.160257] [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: 03/24/2022] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Disrupted feeding and fasting cycles as well as chronic high-fat diet-induced (HFD-induced) obesity are associated with cardiovascular disease risk factors. We designed studies that determined whether 2 weeks of time-restricted feeding (TRF) intervention in mice fed a chronic HFD would reduce cardiovascular disease risk factors. Mice were fed a normal diet (ND; 10% fat) ad libitum or HFD (45% fat) for 18 weeks ad libitum to establish diet-induced obesity. ND or HFD mice were continued on ad libitum diet or subjected to TRF (limiting food availability to 12 hours only during the dark phase) during the final 2 weeks of the feeding protocol. TRF improved whole-body metabolic diurnal rhythms without a change in body weight. HFD mice showed reduced blood pressure dipping compared with ND, which was restored by TRF. Further, TRF reduced aortic wall thickness, decreased aortic stiffness, as well as increased kidney tubular brush border integrity, decreased renal medullary fibrosis, and reduced renal medullary T cell inflammation in HFD mice. These findings indicate that TRF may be an effective intervention for improving vascular and kidney health in a model of established diet-induced obesity.
Collapse
Affiliation(s)
- Paramita Pati
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Carmen De Miguel
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Jodi R. Paul
- Division of Behavioral Neurobiology, Department of Psychiatry; and
| | - Dingguo Zhang
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Jackson Colson
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - John Miller Allan
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Claudia J. Edell
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Megan K. Rhoads
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Luke S. Dunaway
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Sara N. Biswal
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Yihan Zhong
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Randee Sedaka
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Telisha Millender-Swain
- Division of Molecular and Cellular Pathology, Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Shannon M. Bailey
- Division of Molecular and Cellular Pathology, Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Karen L. Gamble
- Division of Behavioral Neurobiology, Department of Psychiatry; and
| | - David M. Pollock
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Jennifer S. Pollock
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| |
Collapse
|
18
|
Wong GC, Bearzatto B, Gala JL, Delzenne NM, Van Hul M, Cani PD. Obesity phenotype and gut microbiota alterations are not associated with anxiety-like behaviour in high-fat diet-fed mice. Food Funct 2025; 16:268-281. [PMID: 39655876 DOI: 10.1039/d4fo04461d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Anxiety is a common co-morbidity with obesity and metabolic disease, and can lead to a significant impact on quality of life. The vast differences in the gut microbiota between obese and control individuals provide a potential avenue for therapeutic intervention. A high-fat diet (HFD) in rodent models have been shown to induce anxiety-like behaviour and has been tested through an array of distinct behavioural tests such as the elevated plus maze test, light-dark test and open field test. Despite differences in testing and assessment parameters, the behavioural outcomes have previously yielded similar results. Recent evidence suggests that HFD has an anxiolytic effect on mice, complicating the model. Here, we aimed to confirm whether HFD-fed mice are more susceptible to presenting anxiety-like behaviours. Our findings showed no significant differences in behaviour, plasma corticosterone and inflammation markers between HFD and control diet (CTD) mice, despite considerable differences in adiposity and faecal microbial communities. Additionally, daily oral gavage is one of the most common methods for testing bacterial probiotics in rodent models, but this handling could potentially also cause stress to the mice. Thus, we investigated if daily oral gavage could mask differences in HFD and CTD mice. We found no significant differences in weight, fat mass or anxiety-like behaviour in CTD-fed mice with or without daily oral gavage.
Collapse
Affiliation(s)
- Giselle C Wong
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), 1200 Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Bertrand Bearzatto
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research (IREC), Centre des Technologies Moléculaires Appliquées (CTMA), 1200 Brussels, Belgium
| | - Jean-Luc Gala
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research (IREC), Centre des Technologies Moléculaires Appliquées (CTMA), 1200 Brussels, Belgium
| | - Nathalie M Delzenne
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), 1200 Brussels, Belgium.
| | - Matthias Van Hul
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), 1200 Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Patrice D Cani
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), 1200 Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO Department, WEL Research Institute, Wavre, Belgium
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research (IREC), 1200 Brussels, Belgium
| |
Collapse
|
19
|
Soliz-Rueda JR, López-Fernández-Sobrino R, Schellekens H, Bravo FI, Suárez M, Mulero M, Muguerza B. Clock system disruption in male Fischer 344 rats fed cafeteria diet and administered sweet treats at different times: The zeitgeber role of grape seed flavanols. Biofactors 2025; 51:e70000. [PMID: 39832727 DOI: 10.1002/biof.70000] [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: 09/24/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Current lifestyles include calorie-dense diets and late-night food intake, which can lead to circadian misalignment. Our group recently demonstrated that sweet treats before bedtime alter the clock system in healthy rats, increasing metabolic risk factors. Therefore, we aimed to assess the impact of the sweet treat consumption time on the clock system in rats fed a cafeteria diet (CAF). Moreover, since flavanols have demonstrated beneficial effects in metabolic disorders and clock gene modulation, we also investigated whether these phenolic compounds can restore the circadian disruption caused by these altered dietary patterns. For this, 64 Fisher rats were fed CAF for 9 weeks. In the last 4 weeks, animals were daily administered a low dose of sugar (160 mg/kg) as a sweet treat at 8 a.m. (ZT0) or 8 p.m. (ZT12). Two other groups received 25 mg/kg of grape seed flavanols in addition to sweet treats. Finally, the animals were sacrificed at different time points (9 a.m., 3 p.m., 9 p.m., and 3 a.m.). The results showed that metabolic and circadian disturbances by CAF may be influenced by the time of sugar administration, slightly reinforcing the alterations in diurnal rhythmicity of serum biochemical parameters, hormones, and hypothalamic genes with bedtime snacking. Flavanols improved metabolic health and restored the oscillation of biochemical parameters, hormones, and clock and appetite-signaling genes, showing greater effects at ZT12. These results highlight the importance of meal timing in influencing physiological and metabolic outcomes, even under calorie-dense diets. Moreover, they also suggest the zeitgeber role of flavanols, modulating the clock system and contributing to an improved metabolic profile under different feeding pattern conditions.
Collapse
Affiliation(s)
- Jorge R Soliz-Rueda
- Departament de Bioquimica i Biotecnologia, Universitat Rovira i Virgili, Nutrigenomics Research Group, Tarragona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), Tarragona, Spain
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Raúl López-Fernández-Sobrino
- Departament de Bioquimica i Biotecnologia, Universitat Rovira i Virgili, Nutrigenomics Research Group, Tarragona, Spain
| | - Harriët Schellekens
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Francisca Isabel Bravo
- Departament de Bioquimica i Biotecnologia, Universitat Rovira i Virgili, Nutrigenomics Research Group, Tarragona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), Tarragona, Spain
| | - Manuel Suárez
- Departament de Bioquimica i Biotecnologia, Universitat Rovira i Virgili, Nutrigenomics Research Group, Tarragona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), Tarragona, Spain
| | - Miquel Mulero
- Departament de Bioquimica i Biotecnologia, Universitat Rovira i Virgili, Nutrigenomics Research Group, Tarragona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), Tarragona, Spain
| | - Begoña Muguerza
- Departament de Bioquimica i Biotecnologia, Universitat Rovira i Virgili, Nutrigenomics Research Group, Tarragona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), Tarragona, Spain
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
20
|
Soliz-Rueda JR, Cuesta-Marti C, O'Mahony SM, Clarke G, Schellekens H, Muguerza B. Gut microbiota and eating behaviour in circadian syndrome. Trends Endocrinol Metab 2025; 36:15-28. [PMID: 39095231 DOI: 10.1016/j.tem.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/04/2024] [Accepted: 07/12/2024] [Indexed: 08/04/2024]
Abstract
Eating behaviour and circadian rhythms are closely related. The type, timing, and quantity of food consumed, and host circadian rhythms, directly influence the intestinal microbiota, which in turn impacts host circadian rhythms and regulates food intake beyond homeostatic eating. This Opinion discusses the impact of food intake and circadian disruptions induced by an obesogenic environment on gut-brain axis signalling. We also explore potential mechanisms underlying the effects of altered gut microbiota on food intake behaviour and circadian rhythmicity. Understanding the crosstalk between gut microbiota, circadian rhythms, and unhealthy eating behaviour is crucial to addressing the obesity epidemic, which remains one of the biggest societal challenges of our time.
Collapse
Affiliation(s)
- Jorge R Soliz-Rueda
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira I Virgili, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain; Center of Environmental, Food and Toxicological Technology (TecnATox), Tarragona, Spain
| | - Cristina Cuesta-Marti
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, Cork, Ireland
| | - Siobhain M O'Mahony
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Harriët Schellekens
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, Cork, Ireland.
| | - Begoña Muguerza
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira I Virgili, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain; Center of Environmental, Food and Toxicological Technology (TecnATox), Tarragona, Spain
| |
Collapse
|
21
|
Ono M, Dai Y, Fujiwara T, Fujiwara H, Daikoku T, Ando H, Kuji N, Nishi H. Influence of lifestyle and the circadian clock on reproduction. Reprod Med Biol 2025; 24:e12641. [PMID: 40078335 PMCID: PMC11897534 DOI: 10.1002/rmb2.12641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Background The biological reproductive process requires the precise coordination of annual and daily signals to adapt to environmental shifts. Humans and animals have developed shared neuroendocrine systems that have adapted to process daily and seasonal light signals within the hypothalamic-pituitary -gonadal axis. However, the stability of circadian and seasonal biological processes is at risk due to industrialization and contemporary round-the-clock lifestyles. These threats include skipping breakfast, excessive artificial illumination during inappropriate hours because of irregular work schedules, nighttime urban lighting, and widespread environmental pollution from endocrine-disrupting chemicals. This review aimed to explore the interplay between lifestyle factors, circadian rhythms, and reproductive functions. Methods This review examined the reciprocal influences of circadian clocks on reproductive hormones, exploring the underlying mechanisms and their implications for fertility and reproductive health. We emphasized key findings regarding molecular clock components, endocrine pathways, and the critical importance of synchronizing circadian rhythms with hormonal cycles. Main Findings The intersection of reproductive endocrinology and circadian biology reveals complex interactions between hormonal regulation and circadian rhythms. Circadian rhythm misalignments due to environmental factors, including late-night work and skipping breakfast, negatively impact endocrine and reproductive functions. Conclusions More strategies are needed to mitigate the effects of circadian disruption on reproductive functions.
Collapse
Affiliation(s)
- Masanori Ono
- Department of Obstetrics and GynecologyTokyo Medical UniversityTokyoJapan
| | - Yidan Dai
- Department of Obstetrics and GynecologyTokyo Medical UniversityTokyoJapan
| | - Tomoko Fujiwara
- Department of Human Life EnvironmentsKyoto Notre Dame UniversityKyotoJapan
| | - Hiroshi Fujiwara
- Ochi Yume ClinicNagoyaJapan
- School of Veterinary MedicineAzabu UniversitySagamiharaJapan
| | - Takiko Daikoku
- Division of Animal Disease Model, Research Center for Experimental Modeling of Human Disease, Graduate School of Medical ScienceKanazawa UniversityKanazawaJapan
| | - Hitoshi Ando
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical ScienceKanazawa UniversityKanazawaJapan
| | - Naoaki Kuji
- Department of Obstetrics and GynecologyTokyo Medical UniversityTokyoJapan
| | - Hirotaka Nishi
- Department of Obstetrics and GynecologyTokyo Medical UniversityTokyoJapan
| |
Collapse
|
22
|
Fu M, Lu S, Gong L, Zhou Y, Wei F, Duan Z, Xiang R, Gonzalez FJ, Li G. Intermittent fasting shifts the diurnal transcriptome atlas of transcription factors. Mol Cell Biochem 2025; 480:491-504. [PMID: 38528297 DOI: 10.1007/s11010-024-04928-y] [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: 10/04/2023] [Accepted: 01/05/2024] [Indexed: 03/27/2024]
Abstract
Intermittent fasting remains a safe and effective strategy to ameliorate various age-related diseases, but its specific mechanisms are not fully understood. Considering that transcription factors (TFs) determine the response to environmental signals, here, we profiled the diurnal expression of 600 samples across four metabolic tissues sampled every 4 over 24 h from mice placed on five different feeding regimens to provide an atlas of TFs in biological space, time, and feeding regimen. Results showed that 1218 TFs exhibited tissue-specific and temporal expression profiles in ad libitum mice, of which 974 displayed significant oscillations at least in one tissue. Intermittent fasting triggered more than 90% (1161 in 1234) of TFs to oscillate somewhere in the body and repartitioned their tissue-specific expression. A single round of fasting generally promoted TF expression, especially in skeletal muscle and adipose tissues, while intermittent fasting mainly suppressed TF expression. Intermittent fasting down-regulated aging pathway and upregulated the pathway responsible for the inhibition of mammalian target of rapamycin (mTOR). Intermittent fasting shifts the diurnal transcriptome atlas of TFs, and mTOR inhibition may orchestrate intermittent fasting-induced health improvements. This atlas offers a reference and resource to understand how TFs and intermittent fasting may contribute to diurnal rhythm oscillation and bring about specific health benefits.
Collapse
Affiliation(s)
- Min Fu
- Department of Neurology, The Fourth Hospital of Changsha, Affiliated Changsha Hospital of Hunan Normal University, Changsha, 410006, Hunan, China
| | - Siyu Lu
- Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Lijun Gong
- Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Yiming Zhou
- Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Fang Wei
- Department of Neurology, The Fourth Hospital of Changsha, Affiliated Changsha Hospital of Hunan Normal University, Changsha, 410006, Hunan, China.
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
| | - Zhigui Duan
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Rong Xiang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 41001, Hunan, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Guolin Li
- Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China.
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
| |
Collapse
|
23
|
Egea MB, Pierce G, Shay N. Intake of S-Methylmethionine Alters Glucose Metabolism and Hepatic Gene Expression in C57BL/6J High-Fat-Fed Mice. Foods 2024; 14:34. [PMID: 39796324 PMCID: PMC11720019 DOI: 10.3390/foods14010034] [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: 11/14/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
A diet containing foods that are sources of S-methylmethionine (SMM), and its use as a dietary supplement, have demonstrated beneficial health effects. Thus, the objective of this work was to evaluate the inclusion of SMM as a dietary supplement in C57BL/6J high-fat-fed mice to verify whether this compound alone would be responsible for these positive effects. Mice were divided into three groups: LF (low-fat diet), HF (high-fat diet), and HF+SMM (high-fat diet plus SMM), and maintained for 10 weeks with water and food provided ad libitum. Body weight and food intake were measured weekly, and food efficiency was calculated. In addition, at week 9, fasting glucose was measured and, after necropsy, at week 10, liver, inguinal adipose, and kidney weights were measured; triglycerides, histology, liver gene expression, serum insulin, and MCP-1 levels were also determined. Final body weight, average weight gain, and the liver/body weight of the SMM group showed a significant difference with the LF group. HF+SMM-fed mice show improved regulation in glucose metabolism, demonstrated by the assessment of fasting glucose, insulin concentration, and HOMA-IR, compared with the HF-fed group. Liver triglycerides and MCP-1 levels showed no significant differences between fed groups. By the positive gene regulation of Sult1e1, Phlda1, and Ciart, we hypothesized that SMM administration to mice may have regulated xenobiotic, glucose, and circadian rhythm pathways.
Collapse
Affiliation(s)
- Mariana Buranelo Egea
- Campus Rio Verde, Goiano Federal Institute of Education, Science and Technology, Rio Verde 75901-970, GO, Brazil
- Department of Food Science and Technology, Oregon State University, Corvallis, OR 97331, USA;
| | - Gavin Pierce
- Department of Food Science and Technology, Oregon State University, Corvallis, OR 97331, USA;
| | - Neil Shay
- Department of Food Science and Technology, Oregon State University, Corvallis, OR 97331, USA;
| |
Collapse
|
24
|
Young MJ, Heanue S, Kanki M, Moneghetti KJ. Circadian disruption and its impact on the cardiovascular system. Trends Endocrinol Metab 2024:S1043-2760(24)00316-3. [PMID: 39706759 DOI: 10.1016/j.tem.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/11/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
Circadian rhythms are highly conserved biorhythms of ~24 h that govern many fundamental biological processes, including cardiovascular (CV) homeostasis. Disrupting the timing of cellular oscillators promotes cellular stress, and induction of pathogenic pathways underpins the pathogenesis of many CV diseases (CVDs). Thus, shift work, late eating, sleep disturbances, and other disruptors can result in an elevated risk of heart disease and increased incidence of adverse CV events. Here, we discuss the importance of circadian rhythms for CV homeostasis, recent developments in understanding the impact of disrupted circadian rhythms on CV health and disease progression, and how understanding the interactions between circadian and CV physiology is crucial for improving interventions to mitigate CVD, especially in populations impacted by disrupted circadian rhythms.
Collapse
Affiliation(s)
- Morag J Young
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia.
| | - Seamus Heanue
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Medicine, Central Clinical School, Monash University, Clayton, VIC, Australia
| | - Monica Kanki
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Kegan J Moneghetti
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
25
|
Woodie LN, Alberto AJ, Krusen BM, Melink LC, Lazar MA. Genetic synchronization of the brain and liver molecular clocks defend against chrono-metabolic disease. Proc Natl Acad Sci U S A 2024; 121:e2417678121. [PMID: 39665757 DOI: 10.1073/pnas.2417678121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/12/2024] [Indexed: 12/13/2024] Open
Abstract
Nearly every cell of the body contains a circadian clock mechanism that is synchronized with the light-entrained clock in the suprachiasmatic nucleus (SCN). Desynchrony between the SCN and the external environment leads to metabolic dysfunction in shift workers. Similarly, mice with markedly shortened endogenous period due to the deletion of circadian REV-ERBα/β nuclear receptors in the SCN (SCN DKO) exhibit increased sensitivity to diet-induced obesity (DIO) on a 24 h light:dark cycle while mice with REV-ERBs deleted in hepatocytes (HepDKO) display exacerbated hepatosteatosis in response to a high-fat diet. Here, we show that inducing deletion of hepatocyte REV-ERBs in SCN DKO mice (Hep-SCN DDKO) rescued the exacerbated DIO and hepatic triglyceride accumulation, without affecting the shortened behavioral period. These findings suggest that metabolic disturbances due to environmental desynchrony with the central clock are due to effects on peripheral clocks which can be mitigated by matching peripheral and central clock periods even in a desynchronous environment. Thus, maintaining synchrony within an organism, rather than between endogenous and exogenous clocks, may be a viable target for the treatment of metabolic disorders associated with circadian disruption.
Collapse
Affiliation(s)
- Lauren N Woodie
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Ahren J Alberto
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Brianna M Krusen
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Lily C Melink
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Mitchell A Lazar
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
26
|
Jia Y, Ma S, Chen X, Chen Z, Yang X, Li H, Jiang L, Du L, Liu L, Ge J. Higher intake energy, protein, and polyunsaturated fatty acids at dinner versus breakfast increase the risk of hyperhomocysteinemia among adults in the USA. Eur J Nutr 2024; 64:47. [PMID: 39680148 DOI: 10.1007/s00394-024-03567-9] [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: 04/09/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE This study analyzed the relation of energy and macronutrient intake at dinner versus breakfast with the risk of hyperhomocysteinemia (Hhcy). METHODS Up to 12,474 adults, in which 1,387 with Hhcy, completed a questionnaire about energy and macronutrient intake in the National Health and Nutrition Examination. The differences (Δ) in that between dinner and breakfast (Δ = dinner - breakfast) were categorized into quartiles. Logistic regression analyses or restrictive cubic spline regressions were conducted to determine the relation in Δ and the risk of Hhcy, as well as the change in risk when 5% energy at dinner was substituted with those at breakfast through isocaloric substitution models. RESULTS After adjusted the confounders, results showed that compared to the research objects in the lowest quartile, those in the highest quartile were more prone to get Hhcy (odds ratio (OR)Δ energy = 1.26, 95% CI = 1.03-1.56; ORΔ protein = 1.25, 95% CI = 1.01-1.55; ORΔ PUFA = 1.22, 95% CI = 1.01-1.49, respectively). Isocalorically replacing 5% energy at dinner with energy at breakfast was related to 5% lower Hhcy risk. Replacing 5% of energy provided by protein at dinner with that by protein or PUFA at breakfast was related to 10% and 11% lower Hhcy risk, respectively. Replacing 5% energy provided by PUFA at dinner with that by protein or PUFA at breakfast were associated with 8% and 6% lower Hhcy risk, respectively. CONCLUSION The optimal intake period for energy, protein, and polyunsaturated fatty acid intake for reducing Hhcy risk in adults was the morning.
Collapse
Affiliation(s)
- Yuehui Jia
- School of Public Health, Qiqihar Medical University, Qiqihar, 161000, China
| | - Shuli Ma
- School of Public Health, Qiqihar Medical University, Qiqihar, 161000, China
| | - Xiaoting Chen
- Sanitary Analysis Center, Scientific Research Office, Qiqihar Medical University, Qiqihar, 161000, China
| | - Zhe Chen
- School of Public Health, Qiqihar Medical University, Qiqihar, 161000, China
| | - Xiaolei Yang
- School of Public Health, Qiqihar Medical University, Qiqihar, 161000, China
| | - Hongjie Li
- School of Public Health, Qiqihar Medical University, Qiqihar, 161000, China
| | - Libo Jiang
- The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, China
| | - Linlin Du
- School of Public Health, Qiqihar Medical University, Qiqihar, 161000, China
| | - Lei Liu
- Modern Educational Technology Center, Qiqihar Medical University, Qiqihar, 161000, China
| | - Jie Ge
- School of Public Health, Qiqihar Medical University, Qiqihar, 161000, China.
| |
Collapse
|
27
|
Blumstein DM, MacManes MD. Impacts of dietary fat on multi tissue gene expression in the desert-adapted cactus mouse. J Exp Biol 2024; 227:jeb247978. [PMID: 39676723 PMCID: PMC11698062 DOI: 10.1242/jeb.247978] [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: 05/07/2024] [Accepted: 11/05/2024] [Indexed: 12/17/2024]
Abstract
Understanding the relationship between dietary fat and physiological responses is crucial in species adapted to arid environments where water scarcity is common. In this study, we present a comprehensive exploration of gene expression across five tissues (kidney, liver, lung, gastrointestinal tract and hypothalamus) and 17 phenotypic measurements, investigating the effects of dietary fat in the desert-adapted cactus mouse (Peromyscus eremicus). We show impacts on immune function, circadian gene regulation and mitochondrial function for mice fed a lower-fat diet compared with mice fed a higher-fat diet. In arid environments with severe water scarcity, even subtle changes in organismal health and water balance can affect physical performance, potentially impacting survival and reproductive success. This study sheds light on the complex interplay between diet, physiological processes and environmental adaptation, providing valuable insights into the multifaceted impacts of dietary choices on organismal well-being and adaptation strategies in arid habitats.
Collapse
Affiliation(s)
- Danielle M. Blumstein
- University of New Hampshire, Molecular, Cellular, and Biomedical Sciences Department, Durham, NH 03824, USA
| | - Matthew D. MacManes
- University of New Hampshire, Molecular, Cellular, and Biomedical Sciences Department, Durham, NH 03824, USA
| |
Collapse
|
28
|
Verdelho Machado M. Circadian Deregulation: Back Facing the Sun Toward Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) Development. Nutrients 2024; 16:4294. [PMID: 39770915 PMCID: PMC11679855 DOI: 10.3390/nu16244294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Earth's rotation around its axis has pressured its inhabitants to adapt to 24 h cycles of day and night. Humans adapted their own circadian rhythms to the Earth's rhythms with a light-aligned awake-sleep cycle. As a consequence, metabolism undergoes drastic changes throughout the circadian cycle and needs plasticity to cope with opposing conditions in the day (when there is an increase in energy demands and food availability), and during the night (when prolonged fasting couples with cyclic changes in the energy demands across the sleep stages). In the last century, human behavior changed dramatically with a disregard for the natural circadian cycles. This misalignment in sleep and eating schedules strongly modulates the metabolism and energy homeostasis, favoring the development of obesity, metabolic syndrome, and metabolic dysfunction-associated steatotic liver disease (MASLD). This review summarizes the effects of circadian disruption, with a particular focus on the feeding and sleep cycles in the development of MASLD and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Mariana Verdelho Machado
- Gastroenterology Department, Hospital de Vila Franca de Xira, 2600-009 Vila Franca de Xira, Portugal; ; Tel.: +351-912620306
- Clínica Universitária de Gastrenterologia, Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
29
|
Kovynev A, Ying Z, Zhang S, Olgiati E, Lambooij JM, Visentin C, Guigas B, Ducarmon QR, Rensen PCN, Schönke M. Timing Matters: Late, but Not Early, Exercise Training Ameliorates MASLD in Part by Modulating the Gut-Liver Axis in Mice. J Pineal Res 2024; 76:e70003. [PMID: 39539028 DOI: 10.1111/jpi.70003] [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: 09/17/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) affects two billion people worldwide and is currently mostly treatable via lifestyle interventions, such as exercise training. However, it is unclear whether the positive effects of exercise are restricted to unique circadian windows. We therefore aimed to study whether the timing of exercise training differentially modulates MASLD development. Twenty weeks old male APOE*3-Leiden.CETP mice were fed a high fat-high cholesterol diet to induce MASLD and treadmill-trained for 1 h five times per week for 12 weeks either early (ZT13; E-RUN) or late (ZT22; L-RUN) in the dark phase while corresponding sedentary groups (E-SED and L-SED) did not. Late, but not early exercise training decreased the MASLD score, body weight, fat mass, and liver triglycerides, accompanied by an altered composition of the gut microbiota. Specifically, only late exercise training increased the abundance of short-chain fatty acid-producing bacterial families and genera, such as Akkermansia, Lachnospiraceae, and Rikenella. To assess the role of the gut microbiota in training-induced effects, the study was repeated and trained (ZT22 only, RUN) or sedentary mice (SED) served as fecal donors for sedentary recipient mice (RUN FMT and SED FMT). Fecal microbiota transplantation reduced liver weight and plasma triglycerides in RUN FMT compared to SED FMT and tended to lower the MASLD score and liver triglycerides. Timing of exercise training is a critical factor for the positive effect on MASLD in this preclinical model, and the effect of late exercise is partially mediated via the gut-liver axis.
Collapse
Affiliation(s)
- Artemiy Kovynev
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Zhixiong Ying
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Sen Zhang
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Emanuele Olgiati
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Joost M Lambooij
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Clara Visentin
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Bruno Guigas
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands
| | - Quinten R Ducarmon
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Milena Schönke
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
30
|
Douglas A, Stevens B, Rendas M, Kane H, Lynch E, Kunkemoeller B, Wessendorf-Rodriguez K, Day EA, Sutton C, Brennan M, O'Brien K, Kohlgruber AC, Prendeville H, Garza AE, O'Neill LAJ, Mills KHG, Metallo CM, Veiga-Fernandes H, Lynch L. Rhythmic IL-17 production by γδ T cells maintains adipose de novo lipogenesis. Nature 2024; 636:206-214. [PMID: 39478228 PMCID: PMC11618085 DOI: 10.1038/s41586-024-08131-3] [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: 05/26/2022] [Accepted: 09/27/2024] [Indexed: 11/06/2024]
Abstract
The circadian rhythm of the immune system helps to protect against pathogens1-3; however, the role of circadian rhythms in immune homeostasis is less well understood. Innate T cells are tissue-resident lymphocytes with key roles in tissue homeostasis4-7. Here we use single-cell RNA sequencing, a molecular-clock reporter and genetic manipulations to show that innate IL-17-producing T cells-including γδ T cells, invariant natural killer T cells and mucosal-associated invariant T cells-are enriched for molecular-clock genes compared with their IFNγ-producing counterparts. We reveal that IL-17-producing γδ (γδ17) T cells, in particular, rely on the molecular clock to maintain adipose tissue homeostasis, and exhibit a robust circadian rhythm for RORγt and IL-17A across adipose depots, which peaks at night. In mice, loss of the molecular clock in the CD45 compartment (Bmal1∆Vav1) affects the production of IL-17 by adipose γδ17 T cells, but not cytokine production by αβ or IFNγ-producing γδ (γδIFNγ) T cells. Circadian IL-17 is essential for de novo lipogenesis in adipose tissue, and mice with an adipocyte-specific deficiency in IL-17 receptor C (IL-17RC) have defects in de novo lipogenesis. Whole-body metabolic analysis in vivo shows that Il17a-/-Il17f-/- mice (which lack expression of IL-17A and IL-17F) have defects in their circadian rhythm for de novo lipogenesis, which results in disruptions to their whole-body metabolic rhythm and core-body-temperature rhythm. This study identifies a crucial role for IL-17 in whole-body metabolic homeostasis and shows that de novo lipogenesis is a major target of IL-17.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Adipose Tissue/metabolism
- Adipose Tissue/immunology
- Circadian Rhythm/genetics
- Circadian Rhythm/immunology
- Homeostasis
- Interferon-gamma/metabolism
- Interleukin-17/genetics
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Lipogenesis
- Mice, Inbred C57BL
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Single-Cell Gene Expression Analysis
- Biological Clocks/genetics
- Biological Clocks/immunology
- Receptors, Interleukin-17/deficiency
- Receptors, Interleukin-17/metabolism
- Body Temperature
- Leukocyte Common Antigens/metabolism
- ARNTL Transcription Factors/genetics
- ARNTL Transcription Factors/metabolism
Collapse
Affiliation(s)
- Aaron Douglas
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Brenneth Stevens
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Miguel Rendas
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Harry Kane
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Evan Lynch
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | - Emily A Day
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Caroline Sutton
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Martin Brennan
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Katie O'Brien
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | | | - Hannah Prendeville
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Amanda E Garza
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Kingston H G Mills
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Christian M Metallo
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Lydia Lynch
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland.
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Molecular Biology, Princeton University, Princeton, NJ, USA.
- Ludwig Cancer Research Institute, Princeton Branch, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
31
|
Burns ME, Contini FM, Michaud JM, Waring CT, Price JC, McFarland AT, Burke SG, Murphy CA, Guindon GE, Krevosky MK, Seggio JA. Obesity alters circadian and behavioral responses to constant light in male mice. Physiol Behav 2024; 287:114711. [PMID: 39395627 DOI: 10.1016/j.physbeh.2024.114711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/14/2024]
Abstract
Exposure to artificial light during the night is known to promote disruption to the biological clock, which can lead to impaired mood and metabolism. Metabolic hormone secretion is modulated by the circadian pacemaker and recent research has shown that hormones such as insulin and leptin can also directly affect behavioral outcomes and the circadian clock. In turn, obesity itself is known to modulate the circadian rhythm and alter emotionality. This study investigated the behavioral and metabolic effects of constant light exposure in two models of obesity - a leptin null mutant (OB) and diet-induced obesity via high-fat diet. For both experiments, mice were placed into either a standard Light:Dark cycle (LD) or constant light (LL) and their circadian locomotor rhythms were continuously monitored. After 10 weeks of exposure to their respective lighting conditions, all mice were subjected to an open field assay to assess their explorative behaviors. Their metabolic hormone levels and inflammation levels were also measured. Behaviorally, exposure to constant light led to increased period lengthening and open field activity in the lean mice compared to both obesity models. Metabolically, LL led to increased cytokine levels and poorer metabolic outcomes in both lean and obese mice, sometimes exacerbating the metabolic issues in the obese mice, independent of weight gain. This study illustrates that LL can produce altered behavioral and physiological outcomes, even in lean mice. These results also indicate that obesity induced by different reasons can lead to shortened circadian rhythmicity and exploratory activity when exposed to chronic light.
Collapse
Affiliation(s)
- Meredith E Burns
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA
| | - Fernanda Medeiros Contini
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA; Now at Harvard University Medical School, Neurobiology Department
| | - Julie M Michaud
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA
| | - Caitlin T Waring
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA; Now at Colorado State University, College of Veterinary Medicine & Biomedical Sciences
| | - John C Price
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA
| | - Alexander T McFarland
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA; Now at Georgia Southern University, Department of Biology
| | - Samantha G Burke
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA; Now at Cummings School of Veterinary Medicine at Tufts University
| | - Cloey A Murphy
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA
| | - Grace E Guindon
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA
| | - Merideth K Krevosky
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA
| | - Joseph A Seggio
- Department of Biological Sciences, Bridgewater State University, 24 Park Ave., Bridgewater, MA 02325, USA.
| |
Collapse
|
32
|
Ma C, Shen B, Chen L, Yang G. Impacts of circadian disruptions on behavioral rhythms in mice. FASEB J 2024; 38:e70183. [PMID: 39570004 DOI: 10.1096/fj.202401536r] [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: 07/05/2024] [Revised: 10/11/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024]
Abstract
Circadian rhythms are fundamental biological processes that recur approximately every 24 h, with the sleep-wake cycle or circadian behavior being a well-known example. In the field of chronobiology, mice serve as valuable model animals for studying mammalian circadian rhythms due to their genetic similarity to humans and the availability of various genetic tools for manipulation. Monitoring locomotor activity in mice provides valuable insights into the impact of various conditions or disturbances on circadian behavior. In this review, we summarized the effects of disturbance of biological rhythms on circadian behavior in mice. External factors, especially light exert a significant impact on circadian behavior. Additionally, feeding timing, food composition, ambient temperature, and physical exercise contribute to variations in the behavior of the mouse. Internal factors, including gender, age, genetic background, and clock gene mutation or deletion, are effective as well. Understanding the effects of circadian disturbances on murine behavior is essential for gaining insights into the underlying mechanisms of circadian regulation and developing potential therapeutic interventions for circadian-related disorders in humans.
Collapse
Affiliation(s)
- Changxiao Ma
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Bingyi Shen
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Lihong Chen
- Health Science Center, East China Normal University, Shanghai, China
| | - Guangrui Yang
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
33
|
Yang Z, Zarbl H, Kong B, Taylor R, Black K, Kipen H, Basaly V, Fang M, Guo GL. Liver-gut axis signaling regulates circadian energy metabolism in shift workers. FASEB J 2024; 38:e70203. [PMID: 39588921 PMCID: PMC11590413 DOI: 10.1096/fj.202402102r] [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: 09/06/2024] [Revised: 11/03/2024] [Accepted: 11/13/2024] [Indexed: 11/27/2024]
Abstract
Circadian rhythm is critical to maintaining the whole-body metabolic homeostasis of an organism. Chronic disruption of circadian rhythm by shift work is an important risk factor for metabolic diseases. Fibroblast growth factor 15/19 (FGF15/19), a key component in the liver-gut axis, potently suppresses bile acid (BA) synthesis and improves insulin sensitivity. FGF15/19 emerges as a novel pharmaceutical target for prevention and treatment of metabolic diseases. The nicotinamide adenine dinucleotide (NAD+)-dependent sirtuin 1 (SIRT1) deacetylase plays an important role in the maintenance of hepatic homeostasis by linking hepatic metabolism to circadian rhythm. Here, our clinical study identified that circadian rhythmicity and levels of plasma FGF19 and BA profiling, and cellular NAD+-dependent SIRT1 signaling were disturbed in night shift (NS, n = 10) compared to day shift (DS, n = 12) nurses. Our in vitro data showed that recombinant FGF19 protein rescued cellular circadian rhythm disrupted by SIRT1 inhibitors. Furthermore, we determined the effect of FGF15 on circadian rhythm and hepatic metabolism in wild-type (WT), Fgf15 knockout (KO), and Fgf15 transgenic (TG) mice. The expressions of circadian-controlled genes (CCGs) involved in SIRT1 signaling, BA and lipid metabolism, and inflammation were disrupted in Fgf15 KO compared to WT and/or Fgf15 TG mice. Moreover, systemic FGF15 deficiency led to the circadian disturbance of NAD+-dependent SIRT1 signaling and significant reduction during nighttime in mice. These findings suggest that FGF15/19 regulates the circadian energy metabolism, which warrants further studies as a putative prognostic biomarker and pharmaceutical target for preventing against metabolic diseases associated with chronic shift work.
Collapse
Affiliation(s)
- Zhenning Yang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- Environmental and Occupational Health Sciences Institute, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Helmut Zarbl
- Environmental and Occupational Health Sciences Institute, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- Department of Environmental and Occupational Health and Justice, School of Public Health, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Bo Kong
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Rulaiha Taylor
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Kathleen Black
- Environmental and Occupational Health Sciences Institute, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Howard Kipen
- Environmental and Occupational Health Sciences Institute, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- Department of Environmental and Occupational Health and Justice, School of Public Health, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Veronia Basaly
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Mingzhu Fang
- Environmental and Occupational Health Sciences Institute, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- Department of Environmental and Occupational Health and Justice, School of Public Health, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Grace L. Guo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- Environmental and Occupational Health Sciences Institute, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- VA New Jersey Health Care SystemVeterans Administration Medical CenterEast OrangeNew JerseyUSA
| |
Collapse
|
34
|
Jakubowicz D, Matz Y, Landau Z, Rosenblum RC, Twito O, Wainstein J, Tsameret S. Interaction Between Early Meals (Big-Breakfast Diet), Clock Gene mRNA Expression, and Gut Microbiome to Regulate Weight Loss and Glucose Metabolism in Obesity and Type 2 Diabetes. Int J Mol Sci 2024; 25:12355. [PMID: 39596418 PMCID: PMC11594859 DOI: 10.3390/ijms252212355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/09/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
The circadian clock gene system plays a pivotal role in coordinating the daily rhythms of most metabolic processes. It is synchronized with the light-dark cycle and the eating-fasting schedule. Notably, the interaction between meal timing and circadian clock genes (CGs) allows for optimizing metabolic processes at specific times of the day. Breakfast has a powerful resetting effect on the CG network. A misaligned meal pattern, such as skipping breakfast, can lead to a discordance between meal timing and the endogenous CGs, and is associated with obesity and T2D. Conversely, concentrating most calories and carbohydrates (CH) in the early hours of the day upregulates metabolic CG expression, thus promoting improved weight loss and glycemic control. Recently, it was revealed that microorganisms in the gastrointestinal tract, known as the gut microbiome (GM), and its derived metabolites display daily oscillation, and play a critical role in energy and glucose metabolism. The timing of meal intake coordinates the oscillation of GM and GM-derived metabolites, which in turn influences CG expression, playing a crucial role in the metabolic response to food intake. An imbalance in the gut microbiota (dysbiosis) can also reciprocally disrupt CG rhythms. Evidence suggests that misaligned meal timing may cause such disruptions and can lead to obesity and hyperglycemia. This manuscript focuses on the reciprocal interaction between meal timing, GM oscillation, and circadian CG rhythms. It will also review studies demonstrating how aligning meal timing with the circadian clock can reset and synchronize CG rhythms and GM oscillations. This synchronization can facilitate weight loss and improve glycemic control in obesity and those with T2D.
Collapse
Affiliation(s)
- Daniela Jakubowicz
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Yael Matz
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Zohar Landau
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Rachel Chava Rosenblum
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Orit Twito
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Julio Wainstein
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Shani Tsameret
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| |
Collapse
|
35
|
Martinez-Sanchez N, Ray D. Rhythmic liver drives feeding behavior. Science 2024; 386:622-623. [PMID: 39509522 DOI: 10.1126/science.adt0743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
The hepatic vagal nerve mediates the impact of circadian disruption on food intake in mice.
Collapse
Affiliation(s)
- Noelia Martinez-Sanchez
- National Institute for Health and Care Research Oxford Health Biomedical Research Centre and John Radcliffe Hospital, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism and Oxford Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - David Ray
- National Institute for Health and Care Research Oxford Health Biomedical Research Centre and John Radcliffe Hospital, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism and Oxford Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| |
Collapse
|
36
|
Woodie LN, Melink LC, Midha M, de Araújo AM, Geisler CE, Alberto AJ, Krusen BM, Zundell DM, de Lartigue G, Hayes MR, Lazar MA. Hepatic vagal afferents convey clock-dependent signals to regulate circadian food intake. Science 2024; 386:673-677. [PMID: 39509517 PMCID: PMC11629121 DOI: 10.1126/science.adn2786] [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: 12/01/2023] [Accepted: 08/30/2024] [Indexed: 11/15/2024]
Abstract
Circadian desynchrony induced by shiftwork or jet lag is detrimental to metabolic health, but how synchronous or desynchronous signals are transmitted among tissues is unknown. We report that liver molecular clock dysfunction is signaled to the brain through the hepatic vagal afferent nerve (HVAN), leading to altered food intake patterns that are corrected by ablation of the HVAN. Hepatic branch vagotomy also prevents food intake disruptions induced by high-fat diet feeding and reduces body weight gain. Our findings reveal a homeostatic feedback signal that relies on communication between the liver and the brain to control circadian food intake patterns. This identifies the hepatic vagus nerve as a potential therapeutic target for obesity in the setting of chronodisruption.
Collapse
Affiliation(s)
- Lauren N. Woodie
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lily C. Melink
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mohit Midha
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Caroline E. Geisler
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ahren J. Alberto
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brianna M. Krusen
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Delaine M. Zundell
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guillaume de Lartigue
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Matthew R. Hayes
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mitchell A. Lazar
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
37
|
Balbo SL, Soares GM, Morari J, Felisberto AM, Vettorazzi JF, Bronczek GA, Bonfleur ML, Carneiro EM, Boschero AC, Velloso LA. Impact of Sleeve Gastrectomy on Body Weight and Food Intake Regulation in Diet-Induced Obese Mice. Curr Issues Mol Biol 2024; 46:12633-12640. [PMID: 39590343 PMCID: PMC11592983 DOI: 10.3390/cimb46110749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
The epidemic of obesity has increased worldwide and is associated with comorbidities such as diabetes and cardiovascular disease. In this context, strategies that modulate body weight and improve glycemic metabolism have increased, and bariatric surgeries such as Sleeve Gastrectomy (SG) have been highlighted in obesity treatment. However, the mechanism by which SG reduces body weight and improves glycemic control remains unknown. Thus, in this study, we aimed to evaluate food intake and the expression of hypothalamic genes involved with the regulation of this process in diet-induced obese mice submitted to SG. For this, we used C57BL/6 mice submitted to a 10-week high-fat diet protocol and submitted to SG. Food intake, fed and fasted glycemia, as well as hypothalamic anorexigenic and orexigenic gene expression were evaluated 4 weeks after the surgical procedure. First, we observed that SG reduces body weight (44.19 ± 0.47 HFD, 43.51 ± 0.71 HFD-SHAM, and 38.22 ± 1.31 HFD-SG), fasting glycemia (115.0 ± 4.60 HFD, 122.4 ± 3.48 HFD-SHAM, and 93.43 ± 4.67 HFD-SG), insulinemia (1.77 ± 0.15 HFD, 1.92 ± 0.27 HFD-SHAM, and 0.93 ± 0.05 HFD-SG), and leptinemia (5.86 ± 1.38 HFD, 6.44 ± 1.51 HFD-SHAM, and 1.43 ± 0.35 HFD-SG) in obese mice. Additionally, SG reduces food (5.15 ± 0.18 HFD, 5.49 ± 0.32, HFD-SHAM, and 3.28 ± 0.26 HFD-SG) and total (16.88 ± 0.88 HFD, 17.05 ± 0.42, HFD-SHAM, and 14.30 ± 0.73 HFD-SG) calorie intake without alterations in anorexigenic and orexigenic gene expression. In conclusion, these data indicate that SG improves obesity-associated alterations at least in part by a reduction in food intake. This effect is not associated with the canonical food intake pathway in the hypothalamus, indicating the involvement of non-canonical pathways in this process.
Collapse
Affiliation(s)
- Sandra Lucinei Balbo
- Laboratory of Endocrine Physiology and Metabolism, Biological Sciences and Health Center, Western Parana State University, Cascavel 85819210, PR, Brazil; (A.M.F.J.); (M.L.B.)
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083864, SP, Brazil; (G.M.S.); (J.M.); (G.A.B.); (E.M.C.); (A.C.B.); (L.A.V.)
| | - Gabriela Moreira Soares
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083864, SP, Brazil; (G.M.S.); (J.M.); (G.A.B.); (E.M.C.); (A.C.B.); (L.A.V.)
| | - Joseane Morari
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083864, SP, Brazil; (G.M.S.); (J.M.); (G.A.B.); (E.M.C.); (A.C.B.); (L.A.V.)
| | - Antonio Machado Felisberto
- Laboratory of Endocrine Physiology and Metabolism, Biological Sciences and Health Center, Western Parana State University, Cascavel 85819210, PR, Brazil; (A.M.F.J.); (M.L.B.)
- Laboratory of Medical Sciences, Latin-American Institute of Life and Natural Sciences, Federal University of Latin-American Integration (UNILA), Foz do Iguassu 85867970, PR, Brazil
| | - Jean Franciesco Vettorazzi
- Laboratory of Medical Sciences, Latin-American Institute of Life and Natural Sciences, Federal University of Latin-American Integration (UNILA), Foz do Iguassu 85867970, PR, Brazil
| | - Gabriela Alves Bronczek
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083864, SP, Brazil; (G.M.S.); (J.M.); (G.A.B.); (E.M.C.); (A.C.B.); (L.A.V.)
| | - Maria Lúcia Bonfleur
- Laboratory of Endocrine Physiology and Metabolism, Biological Sciences and Health Center, Western Parana State University, Cascavel 85819210, PR, Brazil; (A.M.F.J.); (M.L.B.)
| | - Everardo Magalhães Carneiro
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083864, SP, Brazil; (G.M.S.); (J.M.); (G.A.B.); (E.M.C.); (A.C.B.); (L.A.V.)
| | - Antonio Carlos Boschero
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083864, SP, Brazil; (G.M.S.); (J.M.); (G.A.B.); (E.M.C.); (A.C.B.); (L.A.V.)
| | - Lício Augusto Velloso
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083864, SP, Brazil; (G.M.S.); (J.M.); (G.A.B.); (E.M.C.); (A.C.B.); (L.A.V.)
| |
Collapse
|
38
|
Helbling JC, Ginieis R, Mortessagne P, Ruiz-Gayo M, Bakoyiannis I, Ducourneau EG, Ciocca D, Bouleté IM, Favereaux A, Ces A, Montalban E, Capuron L, Jeanneteau F, Ferreira G, Challet E, Moisan MP. Time-restricted feeding prevents memory impairments induced by obesogenic diet consumption, via hippocampal thyroid hormone signaling. Mol Metab 2024; 90:102061. [PMID: 39515608 DOI: 10.1016/j.molmet.2024.102061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE The early consumption of calorie-rich diet disrupts circadian rhythms and has adverse effects on memory, yet the effects of time-restricted feeding (TRF) and the underlying molecular mechanisms are unknown. Here, we set out to identify the behavioral and molecular circadian rhythms disruptions generated by juvenile obesogenic diet consumption and their restoration by TRF in male mice. METHODS Metabolic rhythms were measured by indirect calorimetry and memory performances by behavioral tasks. Hippocampal translatome (pS6_TRAP), enrichment and co-regulated gene network analyses were conducted to identify the molecular pathways involved in memory impairments and their restoration by TRF. Differential exon usage analyses, mass spectrometry and pharmacological intervention were used to confirm thyroid hormone signaling involvement. RESULTS We show that four weeks of TRF restore the rhythmicity of metabolic parameters and prevents memory impairments in mice fed a high fat-high sucrose (HFS) diet since weaning, independently of body fat levels. Hippocampal translatome and differential exon usage analyses indicate that impaired memory of mice under ad libitum HFS diet is accompanied by reduced thyroid hormone signaling and altered expression of astrocytic genes regulating glutamate neurotransmission. TRF restored the diurnal expression variation of part of these genes and intra-hippocampal infusion of T3, the active form of thyroid hormone, rescues memory performances and astrocytic gene expression of ad libitum HFS diet-fed mice. CONCLUSIONS Thus, thyroid hormones contribute to the TRF positive effects on both metabolism and memory in mice fed an obesogenic diet, highlighting this nutritional approach as a powerful tool in addressing obesity brain comorbidities and paving the way for further mechanistic studies on hippocampal thyroid signaling.
Collapse
Affiliation(s)
- Jean-Christophe Helbling
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Rachel Ginieis
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Pierre Mortessagne
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Mariano Ruiz-Gayo
- Department of Health and Pharmaceutical Sciences, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Ioannis Bakoyiannis
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Eva-Gunnel Ducourneau
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Dominique Ciocca
- Chronobiotron, Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, France
| | - Illona-Marie Bouleté
- Chronobiotron, Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, France
| | - Alexandre Favereaux
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Aurélia Ces
- Institute of Cellular and Integrative Neurosciences, CNRS, University of Strasbourg, France
| | - Enrica Montalban
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Lucile Capuron
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Freddy Jeanneteau
- Institut de Génomique Fonctionnelle, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Guillaume Ferreira
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France
| | - Etienne Challet
- Institute of Cellular and Integrative Neurosciences, CNRS, University of Strasbourg, France
| | - Marie-Pierre Moisan
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Teams NutriPsy & FoodCircus, Bordeaux, France.
| |
Collapse
|
39
|
Jin H, Yao L, Chen W, Hou T, Li J, Li B. Konjac glucomannan Inhibits Appetite of Obese Mice by Suppressing Hypothalamic Inflammatory Response and Agrp/ Npy Neuron Expression. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24489-24503. [PMID: 39465542 DOI: 10.1021/acs.jafc.4c05901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Konjac glucomannan (KGM) is used for appetite management. However, KGM's regulation of appetite through hypothalamic neurons and gut microbiota, particularly in nonobese populations, is required to be investigated. This study investigated the differential effects of KGM on appetite and energy metabolism in obese and nonobese mice. In obese mice, KGM inhibited food intake, hypothalamic inflammation, and increased energy expenditure. Conversely, in nonobese mice, KGM maintained food intake and energy expenditure but increased hypothalamic inflammation. KGM downregulated hypothalamic Agrp, Npy, and Orx expression and upregulated Cart in obese mice, while it had no effect on orexigenic genes and downregulated Cart in nonobese mice. Additionally, KGM reshaped gut microbiota and increased Short-chain fatty acids (SCFAs) formation of obese mice, where Alistipes, Bifidobacterium, and Lactobacillus, as well as SCFAs, correlated with suppressed appetite. In nonobese mice, KGM has no significant effect on SCFAs but microbes such as Blautia, Alistipes, and Flavonifractor levels were negatively correlated with hypothalamic inflammation. KGM maintains appetite and was linked to liver-derived phosphatidylcholine, countering increased hypothalamic inflammation. The differential regulation of appetite by KGM between obese and nonobese mice is associated with hypothalamic inflammatory, neuronal, and KGM-induced personalized reshaping of gut microbiota. KGM may regulate energy intake and expenditure through the microbiota-gut-brain axis.
Collapse
Affiliation(s)
- Hong Jin
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan 430070, China
| | - Lanlan Yao
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan 430070, China
| | - Wenjing Chen
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan 430070, China
| | - Tao Hou
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan 430070, China
| | - Jing Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan 430070, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan 430070, China
| |
Collapse
|
40
|
Nguyen HH, Talbot J, Li D, Raghavan V, Littman DR. Modulating intestinal neuroimmune VIPergic signaling attenuates the reduction in ILC3-derived IL-22 and hepatic steatosis in MASLD. Hepatol Commun 2024; 8:e0528. [PMID: 39761015 PMCID: PMC11495769 DOI: 10.1097/hc9.0000000000000528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/18/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD, formerly known as NAFLD) is a major driver of cirrhosis and liver-related mortality. However, therapeutic options for MASLD, including prevention of liver steatosis, are limited. We previously described that vasoactive intestinal peptide-producing neurons (VIP-neurons) regulate the efficiency of intestinal dietary fat absorption and IL-22 production by type 3 innate lymphoid cells (ILC3) in the intestine. Given the described hepatoprotective role of IL-22, we hypothesize that modulation of this neuroimmune circuit could potentially be an innovative approach for the control of liver steatosis. METHODS We used a model of diet-induced MASLD by exposing mice to a high-fat diet (HFD) for 16 weeks, when the development of liver steatosis was first observed in our animals. We characterized IL-22 production by intestinal ILC3 at this dietary endpoint. We then evaluated whether communication between VIP-neurons and ILC3 affected IL-22 production and MASLD development by exposing mice with a conditional genetic deletion of Vipr2 in ILC3 (Rorc(t)CreVipr2fl/fl) to the HFD. We also performed intermittent global inhibition of VIP-neurons using a chemogenetic inhibitory approach (VipIres-CrehM4DiLSL) in HFD-fed mice. RESULTS Production of IL-22 by intestinal ILC3 is reduced in steatotic mice that were exposed to an HFD for 16 weeks. Targeted deletion of VIP receptor 2 in ILC3 resulted in higher production of IL-22 in ILC3 and was associated with a significant reduction in liver steatosis in mice under HFD. Global inhibition of VIP-producing neurons also resulted in a significant reduction in liver steatosis. CONCLUSIONS Modulating VIPergic neuroimmune signaling can ameliorate the development of hepatic steatosis induced by a surplus of fat ingestion in the diet. This neuroimmune pathway should be further investigated as a potential therapeutic avenue in MASLD.
Collapse
Affiliation(s)
- Henry H. Nguyen
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
- Department of Medicine and Department of Microbiology, Immunology, and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jhimmy Talbot
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Dayi Li
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
| | - Varsha Raghavan
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
| | - Dan R. Littman
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
- Howard Hughes Medical Institute, New York, New York, USA
| |
Collapse
|
41
|
Sun J, Zhang Y, Adams JA, Higgins CB, Kelly SC, Zhang H, Cho KY, Johnson UG, Swarts BM, Wada SI, Patti GJ, Shriver LP, Finck BN, Herzog ED, DeBosch BJ. Hepatocyte Period 1 dictates oxidative substrate selection independent of the core circadian clock. Cell Rep 2024; 43:114865. [PMID: 39412985 PMCID: PMC11601098 DOI: 10.1016/j.celrep.2024.114865] [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: 04/08/2024] [Revised: 09/09/2024] [Accepted: 09/26/2024] [Indexed: 10/18/2024] Open
Abstract
Organisms integrate circadian and metabolic signals to optimize substrate selection to survive starvation, yet precisely how this occurs is unclear. Here, we show that hepatocyte Period 1 (Per1) is selectively induced during fasting, and mice lacking hepatocyte Per1 fail to initiate autophagic flux, ketogenesis, and lipid accumulation. Transcriptomic analyses show failed induction of the fasting hepatokine Fgf21 in Per1-deficient mice, and single-nucleus multiome sequencing defines a putative responding hepatocyte subpopulation that fails to induce the chromatin accessibility near the Fgf21 locus. In vivo isotopic tracing and indirect calorimetry demonstrate that hepatocyte Per1-deficient mice fail to transit from oxidation of glucose to fat, which is completely reversible by exogenous FGF21 or by inhibiting pyruvate dehydrogenase. Strikingly, disturbing other core circadian genes does not perturb Per1 induction during fasting. We thus describe Per1 as an important mechanism by which hepatocytes integrate internal circadian rhythm and external nutrition signals to facilitate proper fuel utilization.
Collapse
Affiliation(s)
- Jiameng Sun
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yiming Zhang
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua A Adams
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cassandra B Higgins
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shannon C Kelly
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hao Zhang
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA; Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kevin Y Cho
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA; Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ulysses G Johnson
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI 48859, USA; Biochemistry, Cellular, and Molecular Biology Program, Central Michigan University, Mount Pleasant, MI, USA
| | - Benjamin M Swarts
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI 48859, USA; Biochemistry, Cellular, and Molecular Biology Program, Central Michigan University, Mount Pleasant, MI, USA
| | - Shun-Ichi Wada
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Gary J Patti
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA; Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Leah P Shriver
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA; Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brian N Finck
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Erik D Herzog
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Brian J DeBosch
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
42
|
Klingbeil EA, Schade R, Lee SH, Kirkland R, de La Serre CB. Manipulation of feeding patterns in high fat diet fed rats improves microbiota composition dynamics, inflammation and gut-brain signaling. Physiol Behav 2024; 285:114643. [PMID: 39059597 DOI: 10.1016/j.physbeh.2024.114643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/29/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024]
Abstract
Chronic consumption of high fat (HF) diets has been shown to increase meal size and meal frequency in rodents, resulting in overeating. Reducing meal frequency and establishing periods of fasting, independently of caloric intake, may improve obesity-associated metabolic disorders. Additionally, diet-driven changes in microbiota composition have been shown to play a critical role in the development and maintenance of metabolic disorders. In this study, we used a pair-feeding paradigm to reduce meal frequency and snacking episodes while maintaining overall intake and body weight in HF fed rats. We hypothesized that manipulation of feeding patterns would improve microbiota composition and metabolic outcomes. Male Wistar rats were placed in three groups consuming either a HF, low fat diet (LF, matched for sugar), or pair-fed HF diet for 7 weeks (n = 11-12/group). Pair-fed animals received the same amount of food consumed by the HF fed group once daily before dark onset (HF-PF). Rats underwent oral glucose tolerance and gut peptide cholecystokinin sensitivity tests. Bacterial DNA was extracted from the feces collected during both dark and light cycles and sequenced via Illumina MiSeq sequencing of the 16S V4 region. Our pair-feeding paradigm reduced meal numbers, especially small meals in the inactive phase, without changing total caloric intake. This shift in feeding patterns reduced relative abundances of obesity-associated bacteria and maintained circadian fluctuations in microbial abundances. These changes were associated with improved gastrointestinal (GI) function, reduced inflammation, and improved glucose tolerance and gut to brain signaling. We concluded from these data that targeting snacking may help improve metabolic outcomes, independently of energy content of the diet and hyperphagia.
Collapse
Affiliation(s)
- E A Klingbeil
- Department of Nutritional Sciences, The University of Texas at Austin, United States
| | - R Schade
- Department of Microbiology and Immunology, Stanford University School of Medicine, United States
| | - S H Lee
- Department of Food Sciences, Sun Moon University, South Korea
| | - R Kirkland
- Office of Research, University of Georgia, United States
| | - C B de La Serre
- Department of Nutritional Sciences, University of Georgia, United States; Department of Biomedical Sciences, Colorado State University, United States.
| |
Collapse
|
43
|
Du NH, Sinturel F, Nowak N, Gosselin P, Saini C, Guessous I, Jornayvaz FR, Philippe J, Rey G, Dermitzakis ET, Zenobi R, Dibner C, Brown SA. Multi-omics correlates of insulin resistance and circadian parameters mapped directly from human serum. Eur J Neurosci 2024; 60:5487-5504. [PMID: 39205434 DOI: 10.1111/ejn.16486] [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: 03/27/2024] [Revised: 06/30/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024]
Abstract
While it is generally known that metabolic disorders and circadian dysfunction are intertwined, how the two systems affect each other is not well understood, nor are the genetic factors that might exacerbate this pathological interaction. Blood chemistry is profoundly changed in metabolic disorders, and we have previously shown that serum factors change cellular clock properties. To investigate if circulating factors altered in metabolic disorders have circadian modifying effects, and whether these effects are of genetic origin, we measured circadian rhythms in U2OS cell in the presence of serum collected from diabetic, obese or control subjects. We observed that circadian period lengthening in U2OS cells was associated with serum chemistry that is characteristic of insulin resistance. Characterizing the genetic variants that altered circadian period length by genome-wide association analysis, we found that one of the top variants mapped to the E3 ubiquitin ligase MARCH1 involved in insulin sensitivity. Confirming our data, the serum circadian modifying variants were also enriched in type 2 diabetes and chronotype variants identified in the UK Biobank cohort. Finally, to identify serum factors that might be involved in period lengthening, we performed detailed metabolomics and found that the circadian modifying variants are particularly associated with branched chain amino acids, whose levels are known to correlate with diabetes and insulin resistance. Overall, our multi-omics data showed comprehensively that systemic factors serve as a path through which metabolic disorders influence circadian system, and these can be examined in human populations directly by simple cellular assays in common cultured cells.
Collapse
Affiliation(s)
- Ngoc-Hien Du
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Flore Sinturel
- Department of Surgery, Division of Thoracic and Endocrine Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Nora Nowak
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Pauline Gosselin
- Department of Surgery, Division of Thoracic and Endocrine Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department and Division of Primary Care Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Camille Saini
- Department of Surgery, Division of Thoracic and Endocrine Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department and Division of Primary Care Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Idris Guessous
- Department and Division of Primary Care Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - François R Jornayvaz
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Medicine, Division of Endocrinology, Diabetes, Nutrition, and Therapeutic Education of Patient, University Hospitals of Geneva, Geneva, Switzerland
| | - Jacques Philippe
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Medicine, Division of Endocrinology, Diabetes, Nutrition, and Therapeutic Education of Patient, University Hospitals of Geneva, Geneva, Switzerland
| | - Guillaume Rey
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Emmanouil T Dermitzakis
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Renato Zenobi
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Charna Dibner
- Department of Surgery, Division of Thoracic and Endocrine Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Steven A Brown
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
44
|
la Fleur SE, Blancas-Velazquez AS, Stenvers DJ, Kalsbeek A. Circadian influences on feeding behavior. Neuropharmacology 2024; 256:110007. [PMID: 38795953 DOI: 10.1016/j.neuropharm.2024.110007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/15/2024] [Accepted: 05/19/2024] [Indexed: 05/28/2024]
Abstract
Feeding, like many other biological functions, displays a daily rhythm. This daily rhythmicity is controlled by the circadian timing system of which the central master clock is located in the hypothalamic suprachiasmatic nucleus (SCN). Other brain areas and tissues throughout the body also display rhythmic functions and contain the molecular clock mechanism known as peripheral oscillators. To generate the daily feeding rhythm, the SCN signals to different hypothalamic areas with the lateral hypothalamus, paraventricular nucleus and arcuate nucleus being the most prominent. With respect to the rewarding aspects of feeding behavior, the dopaminergic system is also under circadian influence. However the SCN projects only indirectly to the different reward regions, such as the ventral tegmental area where dopamine neurons are located. In addition, high palatable, high caloric diets have the potential to disturb the normal daily rhythms of physiology and have been shown to alter for example meal patterns. Around a meal several hormones and peptides are released that are also under circadian influence. For example, the release of postprandial insulin and glucagon-like peptide following a meal depend on the time of the day. Finally, we review the effect of deletion of different clock genes on feeding behavior. The most prominent effect on feeding behavior has been observed in Clock mutants, whereas deletion of Bmal1 and Per1/2 only disrupts the day-night rhythm, but not overall intake. Data presented here focus on the rodent literature as only limited data are available on the mechanisms underlying daily rhythms in human eating behavior.
Collapse
Affiliation(s)
- Susanne E la Fleur
- Amsterdam UMC, University of Amsterdam, Laboratory of Endocrinology, Department of Laboratory Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Neuroscience, Cellular and Molecular Mechanisms, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, the Netherlands.
| | - Aurea S Blancas-Velazquez
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dirk Jan Stenvers
- Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - Andries Kalsbeek
- Amsterdam UMC, University of Amsterdam, Laboratory of Endocrinology, Department of Laboratory Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Neuroscience, Cellular and Molecular Mechanisms, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism, Meibergdreef 9, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience (NIN), an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA, Amsterdam, the Netherlands
| |
Collapse
|
45
|
Chaix A, Lin T, Ramms B, Cutler RG, Le T, Lopez C, Miu P, Pinto AFM, Saghatelian A, Playford MP, Mehta NN, Mattson MP, Gordts P, Witztum JL, Panda S. Time-Restricted Feeding Reduces Atherosclerosis in LDLR KO Mice but Not in ApoE Knockout Mice. Arterioscler Thromb Vasc Biol 2024; 44:2069-2087. [PMID: 39087348 PMCID: PMC11409897 DOI: 10.1161/atvbaha.124.320998] [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: 03/23/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Dyslipidemia increases cardiovascular disease risk, the leading cause of death worldwide. Under time-restricted feeding (TRF), wherein food intake is restricted to a consistent window of <12 hours, weight gain, glucose intolerance, inflammation, dyslipidemia, and hypercholesterolemia are all reduced in mice fed an obesogenic diet. LDLR (low-density lipoprotein receptor) mutations are a major cause of familial hypercholesterolemia and early-onset cardiovascular disease. METHODS We subjected benchmark preclinical models, mice lacking LDLR-knockout or ApoE knockout to ad libitum feeding of an isocaloric atherogenic diet either ad libitum or 9 hours TRF for up to 13 weeks and assessed disease development, mechanism, and global changes in hepatic gene expression and plasma lipids. In a regression model, a subset of LDLR-knockout mice were ad libitum fed and then subject to TRF. RESULTS TRF could significantly attenuate weight gain, hypercholesterolemia, and atherosclerosis in mice lacking the LDLR-knockout mice under experimental conditions of both prevention and regression. In LDLR-knockout mice, increased hepatic expression of genes mediating β-oxidation during fasting is associated with reduced VLDL (very-low-density lipoprotein) secretion and lipid accumulation. Additionally, increased sterol catabolism coupled with fecal loss of cholesterol and bile acids contributes to the atheroprotective effect of TRF. Finally, TRF alone or combined with a cholesterol-free diet can reduce atherosclerosis in LDLR-knockout mice. However, mice lacking ApoE, which is an important protein for hepatic lipoprotein reuptake do not respond to TRF. CONCLUSIONS In a preclinical animal model, TRF is effective in both the prevention and regression of atherosclerosis in LDLR knockout mice. The results suggest TRF alone or in combination with a low-cholesterol diet can be a lifestyle intervention for reducing cardiovascular disease risk in humans.
Collapse
Affiliation(s)
- Amandine Chaix
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Terry Lin
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Bastian Ramms
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA. 92093
| | - Roy G. Cutler
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD, USA. 21224
| | - Tiffani Le
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Catherine Lopez
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Phuong Miu
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA. 92093
| | - Antonio F. M. Pinto
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Martin P. Playford
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Nehal N. Mehta
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD, USA. 21224
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States. 21205
| | - Philip Gordts
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA. 92093
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA
| | - Joseph L. Witztum
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA. 92093
| | - Satchidananda Panda
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Lead contact
| |
Collapse
|
46
|
Qi D, Huang D, Ba M, Xuan S, Si H, Lu D, Pei X, Zhang W, Huang S, Li Z. Long-term high fructose intake reprograms the circadian transcriptome and disrupts homeostasis in mouse extra-orbital lacrimal glands. Exp Eye Res 2024; 246:110008. [PMID: 39025460 DOI: 10.1016/j.exer.2024.110008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
This study aims to explore the effects of long-term high fructose intake (LHFI) on the structure, functionality, and physiological homeostasis of mouse extra-orbital lacrimal glands (ELGs), a critical component of ocular health. Our findings reveal significant reprogramming of the circadian transcriptome in ELGs following LHFI, alongside the activation of specific inflammatory pathways, as well as metabolic and neural pathways. Notably, LHFI resulted in increased inflammatory infiltration, enhanced lipid deposition, and reduced nerve fiber density in ELGs compared to controls. Functional assessments indicated a marked reduction in lacrimal secretion following cholinergic stimulation in LHFI-treated mice, suggesting impaired gland function. Overall, our results suggest that LHFI disrupts lacrimal gland homeostasis, potentially leading to dry eye disease by altering its structure and secretory function. These insights underscore the profound impact of dietary choices on ocular health and highlight the need for strategies to mitigate these risks.
Collapse
Affiliation(s)
- Di Qi
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Duliurui Huang
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Mengru Ba
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Shuting Xuan
- Department of Ophthalmology, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Hongli Si
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Wenxiao Zhang
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Zhijie Li
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China.
| |
Collapse
|
47
|
Romero MDM, Martín-González MZ, Aragonès G, Muguerza B, Remesar X, Arola-Arnal A, Fernández-López JA. Time-of-Day Adrenal Modulation of Corticosterone Synthesis is Affected by Sex and Diet but Not by Proanthocyanidins in Rat. Mol Nutr Food Res 2024; 68:e2400323. [PMID: 39148153 DOI: 10.1002/mnfr.202400323] [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: 05/02/2024] [Revised: 07/22/2024] [Indexed: 08/17/2024]
Abstract
SCOPE The aim of this study is to investigate the effect of time-of-day on serum hormones and gene expression in adrenal glands, studying the impact of sex, obesogenic diet, and timing of proanthocyanidins administration, with a focus on glucocorticoids synthesis by this gland. METHODS AND RESULTS Female and male rats, assigned to a standard chow or a cafeteria diet-fed group, receive a daily oral dose of a grape seed proanthocyanidin extract (GSPE), or a vehicle (when light is turned on, or when light is turned off). Corticosterone, estradiol, and testosterone serum levels, and the expression analysis of clock genes and genes related to corticosterone synthesis pathway, are assessed. Serum hormone levels exhibited a marked time-of-day effect also see in the expression of scavenger receptor class B member 1 (Scarb1) and cyp11b genes. The correlation between these two genes and period circadian regulator 2 (Per2) is also extended to other clock genes, although to a lesser extent: cryptochrome (Cry) and nuclear receptor subfamily 1 group D member 1 (Rev-erba). CONCLUSION The strong correlations found suggest an important role of local Per2 (but also of Cry and Rev-erbA) in regulating the expression of the enzymes involved in the corticosterone synthesis pathway. The expression of clock genes in adrenals is influenced by sex and diet but not by GSPE.
Collapse
Affiliation(s)
- Maria-Del-Mar Romero
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, 08028, Spain
- CIBER Obesity and Nutrition, Institute of Health Carlos III, Av. Diagonal 643, Barcelona, 08028, Spain
| | - Miguel Z Martín-González
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, 43007, Spain
| | - Gerard Aragonès
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, 43007, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), Universitat Rovira i Virgili, Tarragona, 43007, Spain
| | - Begoña Muguerza
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, 43007, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), Universitat Rovira i Virgili, Tarragona, 43007, Spain
| | - Xavier Remesar
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, 08028, Spain
- CIBER Obesity and Nutrition, Institute of Health Carlos III, Av. Diagonal 643, Barcelona, 08028, Spain
| | - Anna Arola-Arnal
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, 08028, Spain
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, 43007, Spain
- Center of Environmental, Food and Toxicological Technology (TecnATox), Universitat Rovira i Virgili, Tarragona, 43007, Spain
| | - José-Antonio Fernández-López
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, 08028, Spain
- CIBER Obesity and Nutrition, Institute of Health Carlos III, Av. Diagonal 643, Barcelona, 08028, Spain
| |
Collapse
|
48
|
Rajan PK, Udoh UAS, Finley R, Pierre SV, Sanabria J. The Biological Clock of Liver Metabolism in Metabolic Dysfunction-Associated Steatohepatitis Progression to Hepatocellular Carcinoma. Biomedicines 2024; 12:1961. [PMID: 39335475 PMCID: PMC11428469 DOI: 10.3390/biomedicines12091961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024] Open
Abstract
Circadian rhythms are endogenous behavioral or physiological cycles that are driven by a daily biological clock that persists in the absence of geophysical or environmental temporal cues. Circadian rhythm-related genes code for clock proteins that rise and fall in rhythmic patterns driving biochemical signals of biological processes from metabolism to physiology and behavior. Clock proteins have a pivotal role in liver metabolism and homeostasis, and their disturbances are implicated in various liver disease processes. Encoded genes play critical roles in the initiation and progression of metabolic dysfunction-associated steatohepatitis (MASH) to hepatocellular carcinoma (HCC) and their proteins may become diagnostic markers as well as therapeutic targets. Understanding molecular and metabolic mechanisms underlying circadian rhythms will aid in therapeutic interventions and may have broader clinical applications. The present review provides an overview of the role of the liver's circadian rhythm in metabolic processes in health and disease, emphasizing MASH progression and the oncogenic associations that lead to HCC.
Collapse
Affiliation(s)
- Pradeep Kumar Rajan
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Utibe-Abasi S Udoh
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Robert Finley
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
| | - Juan Sanabria
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
- Department of Nutrition and Metabolomic Core Facility, School of Medicine, Case Western Reserve University, Cleveland, OH 44100, USA
| |
Collapse
|
49
|
Wildes MP, Fernando DG, Grobe CC, Reho JJ, Grobe JL, Kidambi S, Kindel TL, Kwitek AE, Segar JL, Williams JS, Morselli LL. Long-term Metabolic Dysfunction Programming in Female Mice by Serial Moderate Restriction of a High-fat High-sucrose Diet. Endocrinology 2024; 165:bqae117. [PMID: 39236000 PMCID: PMC11408931 DOI: 10.1210/endocr/bqae117] [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: 12/12/2023] [Revised: 05/22/2024] [Accepted: 09/04/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND While intermittent fasting leads to weight loss and improved glucose metabolism, food insecurity, the insufficient access to food for a healthy life, is associated with obesity and adverse cardiometabolic health, especially in women. We aimed to characterize the effects of intermittently restricted feeding on energy balance and glucose tolerance in female mice. METHODS Female C57BL/6J mice were fed a high-fat, high-sucrose diet and intermittently food restricted to 60% of control littermates' ad libitum intake, starting at weaning and until week 19. Restricted mice were subsequently allowed ad libitum access to the same diet. Body composition and energy balance were measured at weeks 18.5, 19, 30, and 40. At week 42, mice underwent an intraperitoneal glucose tolerance test and plasma appetitive hormones measurements after nutrient gavage. RESULTS During the food restriction phase, restricted mice accrued lower weight and fat mass than controls despite periodic ad libitum food access. Reintroduction of continuous ad libitum food caused increased food intake during the light phase and increased body mass in restricted mice. Minor differences in body composition-adjusted energy expenditure between groups were observed at week 40. At week 42, glucose tolerance was impaired in restricted mice compared to controls, and trends toward lower levels of postprandial anorexigenic hormones glucagon-like peptide-1 and pancreatic polypeptide were observed. CONCLUSION Our findings suggest that repeated intermittent food restriction leads to changes in eating behavior that predispose to glucose intolerance when food is freely available. Future studies are needed to elucidate the specific mechanisms underlying these changes.
Collapse
Affiliation(s)
- Micah P Wildes
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Medical Student Summer Research Program, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | - Connie C Grobe
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - John J Reho
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Srividya Kidambi
- Department of Medicine, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Tammy L Kindel
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Anne E Kwitek
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jeffrey L Segar
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Joni S Williams
- Department of Medicine, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Center for Advancing Population Science, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Lisa L Morselli
- Department of Medicine, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
50
|
Acosta-Rodríguez VA, Rijo-Ferreira F, van Rosmalen L, Izumo M, Park N, Joseph C, Hepler C, Thorne AK, Stubblefield J, Bass J, Green CB, Takahashi JS. Misaligned feeding uncouples daily rhythms within brown adipose tissue and between peripheral clocks. Cell Rep 2024; 43:114523. [PMID: 39046875 DOI: 10.1016/j.celrep.2024.114523] [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: 05/22/2023] [Revised: 04/24/2024] [Accepted: 07/05/2024] [Indexed: 07/27/2024] Open
Abstract
Extended food consumption during the rest period perturbs the phase relationship between circadian clocks in the periphery and the brain, leading to adverse health effects. Beyond the liver, how metabolic organs respond to a timed hypocaloric diet is largely unexplored. We investigated how feeding schedules impacted circadian gene expression in epididymal white and brown adipose tissue (eWAT and BAT) compared to the liver and hypothalamus. We restricted food to either daytime or nighttime in C57BL/6J male mice, with or without caloric restriction. Unlike the liver and eWAT, rhythmic clock genes in the BAT remained insensitive to feeding time, similar to the hypothalamus. We uncovered an internal split within the BAT in response to conflicting environmental cues, displaying inverted oscillations on a subset of metabolic genes without modifying its local core circadian machinery. Integrating tissue-specific responses on circadian transcriptional networks with metabolic outcomes may help elucidate the mechanism underlying the health burden of eating at unusual times.
Collapse
Affiliation(s)
- Victoria A Acosta-Rodríguez
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| | - Filipa Rijo-Ferreira
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; Berkeley Public Health, Molecular Cell Biology Department, University of California, Berkeley, Berkeley, CA, USA
| | - Laura van Rosmalen
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Mariko Izumo
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Noheon Park
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Chryshanthi Joseph
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Chelsea Hepler
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Anneke K Thorne
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jeremy Stubblefield
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; Benedictine College, Atchison, KS, USA
| | - Joseph Bass
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Carla B Green
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| |
Collapse
|