1
|
He Y, Zhao G, Ouyang X, Wang S, Chen Y, Li C, He Y, Gao J, Han S, Zhao J, Wang J, Wang C. Creatine-mediated ferroptosis inhibition is involved in the intestinal radioprotection of daytime-restricted feeding. Gut Microbes 2025; 17:2489072. [PMID: 40205678 PMCID: PMC11988229 DOI: 10.1080/19490976.2025.2489072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025] Open
Abstract
Ionizing radiation-induced intestinal injury (IRIII) is a catastrophic disease lack of sufficient medical countermeasures currently. Regulation of the gut microbiota through dietary adjustments is a potential strategy to mitigate IRIII. Time-restricted feeding (TRF) is an emerging behavioral nutrition intervention with pleiotropic health benefits. Whether this dietary pattern influences the pathogenesis of IRIII remains vague. We evaluated the impact of TRF on intestinal radiosensitivity in this study and discovered that only daytime TRF (DTRF), not nighttime TRF, could ameliorate intestinal damage in mice that received a high dose of IR. Faecal metagenomic and metabolomic studies revealed that the intestinal creatine level was increased by approximate 9 times by DTRF, to which the Bifidobacterium pseudolongum enrichment contribute. Further investigations showed that creatine could activate the energy sensor AMP-activated protein kinase in irradiated enterocytes and induce phosphorylation of acetyl-CoA carboxylase, resulting in reduced production of polyunsaturated fatty acids and reduced ferroptosis after IR. The administration of creatine mitigated IRIII and reduced bacteremia and proinflammatory responses. Blockade of creatine import compromised the ferroptosis inhibition and mitigation of DTRF on IRIII. Our study demonstrates a radioprotective dietary mode that can reshape the gut microbiota and increase intestinal creatine, which can suppress IR-induced ferroptosis, thereby providing effective countermeasures for IRIII prevention.
Collapse
Affiliation(s)
- Yingjuan He
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Gaomei Zhao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Xue Ouyang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Shaobo Wang
- Department of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yin Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Chenwenya Li
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Yongwu He
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Jining Gao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Songling Han
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Jinghong Zhao
- Department of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Cheng Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| |
Collapse
|
2
|
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] [MESH Headings] [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
|
3
|
Panganiban J, Kehar M, Ibrahim SH, Hartmann P, Sood S, Hassan S, Ramirez CM, Kohli R, Censani M, Mauney E, Cuda S, Karjoo S. Metabolic dysfunction-associated steatotic liver disease (MASLD) in children with obesity: An Obesity Medicine Association (OMA) and expert joint perspective 2025. OBESITY PILLARS 2025; 14:100164. [PMID: 40230708 PMCID: PMC11995806 DOI: 10.1016/j.obpill.2025.100164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 04/16/2025]
Abstract
Introduction This Obesity Medicine Association (OMA) Expert Joint Perspective examines steatotic liver disease (SLD), which is composed of metabolic dysfunction-associated steatotic liver disease (MASLD), and metabolic dysfunction-associated steatohepatitis (MASH) in children with obesity. The prevalence of obesity is increasing, rates have tripled since 1963 from 5 % to now 19 % of US children affected in 2018. MASLD, is the most common liver disease seen in children, can be a precursor to the development of Type 2 Diabetes (T2DM) and is the primary reason for liver transplant listing in young adults. We must be vigilant in prevention and treatment of MASLD in childhood to prevent further progression. Methods This joint clinical perspective is based upon scientific evidence, peer and clinical expertise. The medical literature was reviewed via PubMed search and appropriate articles were included in this review. This work was formulated from the collaboration of eight hepatologists/gastroenterologists with MASLD expertise and two physicians from the OMA. Results The authors who are experts in the field, determined sentinel questions often asked by clinicians regarding MASLD in children with obesity. They created a consensus and clinical guideline for clinicians on the screening, diagnosis, and treatment of MASLD associated with obesity in children. Conclusions Obesity and the comorbidity of MASLD is increasing in children, and this is a medical problem that needs to be addressed urgently. It is well known that children with metabolic associated chronic disease often continue to have these chronic diseases as adults, which leads to reduced life expectancy, quality of life, and increasing healthcare needs and financial burden. The authors of this paper recommend healthy weight reduction not only through lifestyle modification but through obesity pharmacotherapy and bariatric surgery. Therefore, this guidance reviews available therapies to achieve healthy weight reduction and reverse MASLD to prevent progressive liver fibrosis, and metabolic disease.
Collapse
Affiliation(s)
| | - Mohit Kehar
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Eastern Ontario, Ottawa, Canada
| | - Samar H. Ibrahim
- Division of Pediatric Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Phillipp Hartmann
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Division of Gastroenterology, Hepatology & Nutrition, Rady Children’s Hospital San Diego, San Diego, CA, USA
| | - Shilpa Sood
- Division of Pediatric Gastroenterology, Boston Children's Health Physicians, New York Medical College, Valhalla, NY, USA
| | - Sara Hassan
- University of Texas Southwestern, Dallas, TX, United States
| | | | - Rohit Kohli
- Children's Hospital Los Angeles, CA, United States
| | - Marisa Censani
- Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, United States
| | - Erin Mauney
- Tufts Medical Center, Boston, MA, United States
| | - Suzanne Cuda
- Alamo City Healthy Kids and Families, San Antonio, TX, United States
| | - Sara Karjoo
- Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
- University of South Florida, Tampa, FL, United States
- Florida State University, Tallahassee, FL, United States
| |
Collapse
|
4
|
Fortin BM, Mahieu AL, Fellows RC, Kang Y, Lewis AN, Ead AS, Lamia KA, Cao Y, Pannunzio NR, Masri S. The diverse roles of the circadian clock in cancer. NATURE CANCER 2025:10.1038/s43018-025-00981-8. [PMID: 40419761 DOI: 10.1038/s43018-025-00981-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 04/03/2025] [Indexed: 05/28/2025]
Abstract
A growing part of the human population is affected by circadian misalignment caused by deregulated sleep, increased nighttime light exposure and erratic eating patterns. Thus, circadian rhythms are a key research area, with compelling links to cancer. Here, we review the circadian regulation of critical cellular processes, including immunity, metabolism, cell cycle control and DNA repair, under physiological homeostasis and in cancer. We discuss the divergent evidence indicating tissue-specific roles of the circadian clock in different cancer types and the potential link between circadian misalignment and early-onset cancers. Finally, we outline how understanding the circadian clock can improve cancer prevention and chronomedicine-based therapies.
Collapse
Affiliation(s)
- Bridget M Fortin
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Alisa L Mahieu
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Rachel C Fellows
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Yi Kang
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Program in Physical Therapy, Washington University School of Medicine, St. Louis, MO, USA
| | - Amber N Lewis
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Aya S Ead
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Katja A Lamia
- Department of Molecular and Cellular Biology, Scripps Research Institute, La Jolla, CA, USA
| | - Yin Cao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicholas R Pannunzio
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA.
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, USA.
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, USA.
| | - Selma Masri
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA.
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
5
|
Zhang Z, Kong APS, Wong VWS, Hui HX. Intermittent fasting and metabolic dysfunction-associated steatotic liver disease: the potential role of the gut-liver axis. Cell Biosci 2025; 15:64. [PMID: 40410852 PMCID: PMC12102857 DOI: 10.1186/s13578-025-01406-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Accepted: 05/05/2025] [Indexed: 05/25/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing public health concern linked to the increasing prevalence of metabolic syndrome, including obesity and type 2 diabetes (T2D). MASLD remains a significant clinical challenge due to the absence of effective therapeutic interventions. Intermittent fasting (IF) has emerged as a promising non-pharmacological strategy for managing MASLD. Although the exact mechanisms underpinning the possible beneficial effects of IF on MASLD are not yet fully elucidated, the gut microbiota and its metabolic byproducts are increasingly recognized as potential mediators of these effects. The gut-liver axis may act as an important conduit through which IF exerts its beneficial influence on hepatic function. This review comprehensively examines the impact of various IF protocols on gut microbiota composition, investigating the resultant alterations in microbial diversity and metabolomic profiles, and their potential implications for liver health and the improvement of MASLD.
Collapse
Affiliation(s)
- Zhaoxi Zhang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Alice Pik-Shan Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Vincent Wai-Sun Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hannah Xiaoyan Hui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
6
|
Cano LC, Navarrete E, Ochoa-Romo JP, Díaz G, Díaz-Hernández V, Montúfar-Chaveznava R, Caldelas I. Chronic Maternal Overnutrition and Nutritional Challenge in Adult Life Disrupt Metabolic Diurnal Rhythmicity and Clock Gene Expression in Central and Peripheral Circadian Oscillators. BIOLOGY 2025; 14:541. [PMID: 40427730 PMCID: PMC12108715 DOI: 10.3390/biology14050541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/07/2024] [Accepted: 11/25/2024] [Indexed: 05/29/2025]
Abstract
In mammals, the core molecular clock genes and the overall circadian system are established during early development; during this critical period of development, maternal metabolic condition plays a major role in programming temporal metabolic regulation. Therefore, this study aimed to evaluate the effects of the chronic maternal intake of a high-fat and high-carbohydrate diet (HFCD) before and during pregnancy, in addition to a challenge with HFCD during adulthood, on offspring diurnal metabolic profile and on clock gene expression in central and peripheral circadian oscillators. The HFCD offspring and/or those exposed to the metabolic challenge exhibited alterations in the temporal profiles of analytes associated with both the carbohydrate and lipid metabolisms, as well as markers associated with liver and kidney damage, ranging from phase changes in rhythmicity or, in some cases, to the complete loss of 24 h variations. At the molecular level, the expression of clock genes (Per1, Cry1, Bmal1, and Clock) in the central and peripheral oscillators showed differential susceptibility to undergoing changes in their abundance. Our data indicate that maternal HFCD during pregnancy, a second exposure in adulthood, or both result in the long-term misalignment of the diurnal rhythm's metabolic and damage markers; these changes are possibly associated with alterations in the core molecular circadian clockwork.
Collapse
Affiliation(s)
- Lucía Carolina Cano
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (L.C.C.); (E.N.); (J.P.O.-R.); (G.D.)
| | - Erika Navarrete
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (L.C.C.); (E.N.); (J.P.O.-R.); (G.D.)
| | - Juan Pablo Ochoa-Romo
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (L.C.C.); (E.N.); (J.P.O.-R.); (G.D.)
| | - Georgina Díaz
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (L.C.C.); (E.N.); (J.P.O.-R.); (G.D.)
| | - Verónica Díaz-Hernández
- Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | | | - Ivette Caldelas
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (L.C.C.); (E.N.); (J.P.O.-R.); (G.D.)
| |
Collapse
|
7
|
Dashti HS, Jansen EC, Zuraikat FM, Dixit S, Brown M, Laposky A, Broussard JL, Butler MP, Creasy SA, Crispim CA, Depner CM, Esser KA, Garaulet M, Hanlon EC, Makarem N, Manoogian ENC, Peterson CM, Scheer FAJL, Wright KP, Goff DC, Pratt CA, Gamble KL, St-Onge MP. Advancing Chrononutrition for Cardiometabolic Health: A 2023 National Heart, Lung, and Blood Institute Workshop Report. J Am Heart Assoc 2025; 14:e039373. [PMID: 40265587 DOI: 10.1161/jaha.124.039373] [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] [Indexed: 04/24/2025]
Abstract
The circadian system maintains optimal biological functions at the appropriate time of day, and the disruption of this organization can contribute to the pathogenesis of cardiometabolic disorders. The timing of eating is a prominent external time cue that influences the circadian system. "Chrononutrition" is an emerging dimension of nutrition and active area of research that examines how timing-related aspects of eating and nutrition impact circadian rhythms, biological processes, and disease pathogenesis. There is evidence to support chrononutrition as a form of chronotherapy, such that optimizing the timing of eating may serve as an actionable strategy to improve cardiometabolic health. This report summarizes key information from the National Heart, Lung, and Blood Institute's virtual workshop entitled "Chrononutrition: Elucidating the Role of Circadian Biology and Meal Timing in Cardiometabolic Health," which convened on May 2 to 3, 2023, to review current literature and identify critical knowledge gaps and research opportunities. The speakers presented evidence highlighting the impact on cardiometabolic health of earlier and shorter eating windows and more consistent day-to-day eating patterns. The multidimensionality of chrononutrition was a common theme, as it encompasses multiple facets of eating along with the timing of other behaviors including sleep and physical activity. Advancing the emerging field of chrononutrition will require: (1) standardization of terminology and metrics; (2) scalable and precise tools for real-world settings; (3) consideration of individual differences that may act as effect modifiers; and (4) deeper understanding of social, behavioral, and cultural influences. Ultimately, there is great potential for circadian-based dietary interventions to improve cardiometabolic health.
Collapse
Affiliation(s)
- Hassan S Dashti
- Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Boston MA USA
- Division of Nutrition Harvard Medical School Boston MA USA
- Division of Sleep Medicine Harvard Medical School Boston MA USA
- Broad Institute Cambridge MA USA
| | - Erica C Jansen
- Department of Nutritional Sciences University of Michigan School of Public Health Ann Arbor MI USA
- Department of Neurology University of Michigan Ann Arbor MI USA
| | - Faris M Zuraikat
- Center of Excellence for Sleep and Circadian Research, Department of Medicine Columbia University Irving Medical Center New York NY USA
- Division of General Medicine, Department of Medicine Columbia University Irving Medical Center New York NY USA
- Institute of Human Nutrition, Columbia University Irving Medical Center New York NY USA
| | - Shilpy Dixit
- National Center on Sleep Disorders Research National Heart, Lung, and Blood Institute, National Institutes of Health Bethesda MD USA
| | - Marishka Brown
- National Center on Sleep Disorders Research National Heart, Lung, and Blood Institute, National Institutes of Health Bethesda MD USA
| | - Aaron Laposky
- National Center on Sleep Disorders Research National Heart, Lung, and Blood Institute, National Institutes of Health Bethesda MD USA
| | - Josiane L Broussard
- Department of Health and Exercise Science Colorado State University Fort Collins CO USA
- Ludeman Family Center for Women's Health Research University of Colorado Anschutz Medical Campus Aurora CO USA
- Division of Endocrinology, Metabolism, and Diabetes University of Colorado Anschutz Medical Campus Aurora CO USA
- Department of Integrative Physiology University of Colorado Boulder Boulder CO USA
| | - Matthew P Butler
- Oregon Institute of Occupational Health Sciences Oregon Health and Sciences University Portland OR USA
- Department of Behavioral Neuroscience, School of Medicine Oregon Health and Sciences University Portland OR USA
| | - Seth A Creasy
- Division of Endocrinology, Metabolism, and Diabetes University of Colorado Anschutz Medical Campus Aurora CO USA
- Anschutz Health and Wellness Center University of Colorado Anschutz Medical Campus Aurora CO USA
| | - Cibele A Crispim
- Chrononutrition Research Group, School of Medicine Federal University of Uberlândia Minas Gerais Brazil
| | | | - Karyn A Esser
- Department of Physiology and Aging, College of Medicine University of Florida Gainesville FL USA
| | - Marta Garaulet
- Department of Physiology, Regional Campus of International Excellence University of Murcia Spain
- Biomedical Research Institute of Murcia, IMIB-Arrixaca-UMU, University Clinical Hospital Murcia Spain
- Division of Sleep and Circadian Disorders, Department of Medicine and Neurology Brigham and Women's Hospital Boston MA USA
| | - Erin C Hanlon
- Section of Adult and Pediatric Endocrinology, Department of Medicine University of Chicago IL USA
| | - Nour Makarem
- Department of Epidemiology, Mailman School of Public Health Columbia University Irving Medical Center New York NY USA
| | - Emily N C Manoogian
- Regulatory Biology Department Salk Institute for Biological Sciences La Jolla CA USA
| | - Courtney M Peterson
- Department of Nutrition Sciences University of Alabama at Birmingham Birmingham AL USA
| | - Frank A J L Scheer
- Division of Nutrition Harvard Medical School Boston MA USA
- Division of Sleep Medicine Harvard Medical School Boston MA USA
- Broad Institute Cambridge MA USA
- Division of Sleep and Circadian Disorders, Department of Medicine and Neurology Brigham and Women's Hospital Boston MA USA
| | - Kenneth P Wright
- Division of Endocrinology, Metabolism, and Diabetes University of Colorado Anschutz Medical Campus Aurora CO USA
- Department of Integrative Physiology University of Colorado Boulder Boulder CO USA
| | - David C Goff
- Division of Cardiovascular Sciences National Heart, Lung, and Blood Institute, National Institutes of Health Bethesda MD USA
| | - Charlotte A Pratt
- Division of Cardiovascular Sciences National Heart, Lung, and Blood Institute, National Institutes of Health Bethesda MD USA
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, School of Medicine University of Alabama at Birmingham Birmingham AL USA
- Nutrition Obesity Research Center University of Alabama at Birmingham Birmingham AL USA
| | - Marie-Pierre St-Onge
- Center of Excellence for Sleep and Circadian Research, Department of Medicine Columbia University Irving Medical Center New York NY USA
- Division of General Medicine, Department of Medicine Columbia University Irving Medical Center New York NY USA
| |
Collapse
|
8
|
Wei C, Xu X, Zhang J, Wang X, Han T, Zhang Y, Pan S, Ming Z, Li R, Lou F, Cheng Y, Xu H, Sun X, Geng G, Pan Y, Liu Q, Qi H, Yan X, Dang K, Zhou J, Sun C, Li Y. Timing of unsaturated fat intake improves insulin sensitivity via the gut microbiota-bile acid axis: a randomized controlled trial. Nat Commun 2025; 16:4211. [PMID: 40328731 PMCID: PMC12056104 DOI: 10.1038/s41467-025-58937-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
The timing of dietary total fat intake influences glucose homeostasis, however, the impact of unsaturated fat (USFA) intake has yet to be explored. This 12-week, double-blind, randomized, controlled, 2 × 2 factorial-designed feeding trial investigated the effects of timing (lunch or dinner) and types of dietary USFA (high monounsaturated fat or polyunsaturated fat diet) intake on glucose metabolism in seventy prediabetes participants (mean age, 57 years). Sixty participants with completed fecal samples were included in the final analysis (n = 15 for each group). Postprandial serum glucose was first primary outcome, postprandial insulin levels and insulin sensitivity indices were co-primary outcomes Secondary outcomes were continuous glucose levels, serum fatty acid profile, gut microbiome (metagenomic sequencing) and fecal metabolites. Results showed no significant differences in postprandial glucose between groups. However, USFA intake at lunch (vs. dinner) improved insulin sensitivity and reduced postprandial insulin and serum free saturated fatty acid (Ptiming < 0.05, Ptype > 0.05, Pinteraction > 0.05), which was associated with alterations in gut microbiome and bile acid metabolism, regardless of USFA type. In summary, these results suggest that advancing timing of USFA intake improves insulin sensitivity through the gut microbiome and bile acid metabolism. Trial registration: ChiCTR2100045645.
Collapse
Affiliation(s)
- Chunbo Wei
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaoqing Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jia Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xuanyang Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Tianshu Han
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yingfeng Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Sijia Pan
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhu Ming
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Ran Li
- Department of Clinical Nutrition, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Fengge Lou
- Public Health Research Office, School of Public Health, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Yu Cheng
- Public Health Research Office, School of Public Health, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Huan Xu
- Department of Clinical Nutrition, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xingyuan Sun
- Department of Neurology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Guannan Geng
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yujun Pan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qianmin Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Haitao Qi
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xuemin Yan
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Keke Dang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiaofeng Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China.
| | - Ying Li
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
9
|
Ashcroft SP, Ehrlich AM, Burek K, Pendergrast LA, Yonamine CY, Treebak JT, Zierath JR. Enhanced metabolic adaptations following late dark phase wheel running in high-fat diet-fed mice. Mol Metab 2025; 95:102116. [PMID: 39993626 PMCID: PMC11930447 DOI: 10.1016/j.molmet.2025.102116] [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: 01/30/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/26/2025] Open
Abstract
Exercise interventions represent an effective strategy to prevent and treat metabolic diseases and the time-of-day-dependent effects of exercise on metabolic outcomes are becoming increasingly apparent. We aimed to study the influence of time-restricted wheel running on whole-body energy and glucose homeostasis. Male, 8-week-old, C57BL/6NTac mice were fed either a 60% high-fat diet (HFD) or a 10% low-fat diet (LFD) for 4 weeks. Following this, mice were given access to a running wheel between zeitgeber time (ZT) 12-16 (early dark phase) or ZT 20-0 (late dark phase). Sedentary mice had access to a permanently locked wheel. Mice were housed under these conditions in metabolic chambers for 4 weeks in which LFD and HFD conditions were maintained. Following the exercise intervention, body composition and glucose tolerance were assessed. Wheel running during either the early or late dark phase resulted in metabolic improvements such as attenuation in body weight gain, enhanced glucose tolerance and reduced ectopic lipid deposition. However, late dark phase exercise resulted in a greater reduction in body weight gain, as well as enhanced metabolic flexibility and insulin sensitivity. Our data suggest that late dark phase versus early dark phase exercise confers greater metabolic adaptations in HFD-fed mice.
Collapse
Affiliation(s)
- Stephen P Ashcroft
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amy M Ehrlich
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Krzysztof Burek
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Logan A Pendergrast
- Integrative Physiology Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Caio Y Yonamine
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Integrative Physiology Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Integrative Physiology Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
10
|
Liu L, Sun P, Lin J, Wu S. Associations of reproductive factors and circadian syndrome in middle-aged and elderly women: A nationwide cross-sectional study from China, the United Kingdom and the United States. Sleep Med 2025; 129:283-291. [PMID: 40068580 DOI: 10.1016/j.sleep.2025.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 04/11/2025]
Abstract
Circadian Syndrome (CircS) was recently recognized as a novel predictor of cardiovascular disease (CVD) risk, with reproductive factors playing an important role in CVD risk. Yet, studies linking reproductive factors to CircS remain sparse. Data on middle-aged and elderly women were extracted from three nationally representative surveys: the China Health and Retirement Longitudinal Study (CHARLS) and the English Longitudinal Study on Ageing (ELSA) provided the training set, and the National Health and Nutrition Examination Survey (NHANES) constituted the validation set. We employed logistic regression to evaluate the association between self-reported reproductive factors and CircS risk, with inverse probability of treatment weighting (IPTW) and subgroup analyses conducted to verify the stability. A total of 11,721 participants were analyzed. CircS prevalence differed significantly across countries, with 51.40 % in China and 20.19 % in the United Kingdom. Early menarche (age <12 years) correlated with increased CircS risk in CHARLS (OR 1.38 [95 % CI 0.99-1.92]; p = 0.061), ELSA (OR 1.64 [95 % CI 1.36-1.98]; p < 0.001), and NHANES (OR1.52 [95 % CI: 1.21-1.89]; p < 0.001). Premature menopause (age <40 years) was associated with a roughly 30 % higher CircS risk. A shorter reproductive lifespan was significantly linked to CircS, with this relationship emerging at a reproductive lifespan of ≥40 years in CHARLS (OR1.39 [95 % CI: 1.04-1.84]; p = 0.024). The aforementioned correlations retained significance following IPTW and subgroup analyses. Early menarche, premature menopause, and abbreviated reproductive lifespans may negatively affect CircS. Public health strategies should incorporate menstrual cycle-related reproductive health into primary CircS prevention.
Collapse
Affiliation(s)
- Linli Liu
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian, China; Department of Gynecology, Fuzhou First General Hospital, Affiliated to Fujian Medical University, No.190, Dadao Road, Taijiang District, Fuzhou, Fujian, China
| | - Pengming Sun
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian, China; Department of Gynecology, Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No. 18 Daoshan Road, Fuzhou, Fujian, China; Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Medical University, No. 18 Daoshan Road, Fuzhou, Fujian, China.
| | - Jun Lin
- Department of Gynecology, Fuzhou First General Hospital, Affiliated to Fujian Medical University, No.190, Dadao Road, Taijiang District, Fuzhou, Fujian, China
| | - Sanshan Wu
- Department of Gynecology, Fuzhou First General Hospital, Affiliated to Fujian Medical University, No.190, Dadao Road, Taijiang District, Fuzhou, Fujian, China
| |
Collapse
|
11
|
Arruda AC, Santos RB, Freitas-Lima LC, Budu A, Perilhão MS, Wasinski F, Arthur GM, Guzmán RR, Gomes G, Pesquero JB, Mecawi AS, Bader M, Keller AC, Donato Junior J, Festuccia WT, Mori MA, Araujo RC. 16/8 intermittent fasting in mice protects from diet-induced obesity by increasing leptin sensitivity and postprandial thermogenesis. Acta Physiol (Oxf) 2025; 241:e70036. [PMID: 40186359 DOI: 10.1111/apha.70036] [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: 07/16/2024] [Revised: 02/20/2025] [Accepted: 03/17/2025] [Indexed: 04/07/2025]
Abstract
AIMS To evaluate the molecular mechanisms involved in intermittent fasting 16/8 (16/8 IF), a widespread dietary practice adopted worldwide that consists of 16 h of fasting and 8 h of feeding. METHODS Obese mice were fasted daily from 6 am to 10 pm. Food intake, body weight, and energy expenditure were measured. Molecular mechanisms were investigated using ELISA, western blot, and qPCR of white and brown adipose tissues. Glucose homeostasis was also evaluated. Ucp1 knockout and ob/ob mice were utilized. RESULTS The 16/8 IF regimen improved glucose homeostasis and reduced body weight, food intake, and overall adiposity. Postprandial VO2, heat production, brown adipose tissue (BAT) temperature, and ketone bodies increased with 16/8 IF. Postprandial thermogenesis induced by 16/8 IF was abolished in mice after BAT denervation or Ucp1 deletion. Serum leptin levels were elevated, and most metabolic effects of 16/8 IF were absent in leptin-deficient ob/ob mice. Additionally, leptin sensitivity increased in mice exposed to 16/8 IF. CONCLUSION The 16/8 IF regimen can improve metabolism, with findings underscoring the role of enhanced leptin action in inhibiting food intake and promoting postprandial thermogenesis during 16/8 IF.
Collapse
Affiliation(s)
- Adriano Cleis Arruda
- Laboratory of Genetics and Exercise Metabolism, Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Raisa Brito Santos
- Laboratory of Genetics and Exercise Metabolism, Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Leandro Ceotto Freitas-Lima
- Laboratory of Genetics and Exercise Metabolism, Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Alexandre Budu
- Laboratory of Genetics and Exercise Metabolism, Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Mauro Sergio Perilhão
- Laboratory of Genetics and Exercise Metabolism, Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Frederick Wasinski
- Department of Neurology and Neurosurgery, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Gabriel Melo Arthur
- Laboratory of Genetics and Exercise Metabolism, Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Roger Rodrigues Guzmán
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Guilherme Gomes
- Department of Science and Innovation, Predikta - Scientific Solutions, São Paulo University, São Paulo, São Paulo, Brazil
| | - Joao Bosco Pesquero
- Departament of Biophysics, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - André Souza Mecawi
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Charité University Medicine, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Alexandre Castro Keller
- Department of Microbiology Immunology and Parasitology, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - José Donato Junior
- Department of Physiology and Biophysics, Institute of Biomedical Science, Sao Paulo University, São Paulo, São Paulo, Brazil
| | - Willian Tadeu Festuccia
- Department of Physiology and Biophysics, Institute of Biomedical Science, Sao Paulo University, São Paulo, São Paulo, Brazil
| | - Marcelo A Mori
- Laboratory of Aging Biology, Department of Biochemistry and Tissue Biology, de Biology Institute, UNICAMP, São Paulo, São Paulo, Brazil
| | - Ronaldo Carvalho Araujo
- Laboratory of Genetics and Exercise Metabolism, Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
12
|
de Morais Araújo NC, Paixão JA, de Oliveira Freitas F, de Araújo Gonçalves DN, de Araujo FWC, da Silva SA, do Nascimento E. Comparative study of time-restricted eating on body composition and metabolic parameters in climacteric women with obesity: analysis of a pre-post intervention. Menopause 2025; 32:453-460. [PMID: 39999466 DOI: 10.1097/gme.0000000000002518] [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: 06/04/2024] [Accepted: 11/12/2024] [Indexed: 02/27/2025]
Abstract
OBJECTIVE The physiological changes inherent to the climacteric period can trigger or aggravate overweight/obesity, among several other health disorders. This study aimed to evaluate the effects of time-restricted eating (TRE) on body composition and cardiometabolic parameters in climacteric women with obesity submitted to caloric restriction (CR) through a hypocaloric diet. METHODS We conducted an analysis of a clinical trial in a pre-post design that included adult climacteric women with obesity. Participants were divided into two groups: hypocaloric diet control group (n = 30) - with hypocaloric diet and free meal times, and hypocaloric diet and time-restricted eating group (HTRE) (n = 27) - with hypocaloric diet and TRE (fasting from 7 pm to 7 am ), for 10 weeks. Anthropometric and biochemical parameters were evaluated before and after the intervention period. A significance level of P < 0.05 was considered for all cases. RESULTS Both groups showed a reduction in all anthropometric parameters, but without significant difference ( P = 0.34) between groups. However, some metabolic parameters were significantly highlighted in the HTRE such as cholesterol level normalization (HTRE, 181.76 ± 34.20 mg/dL) and a decrease in plasma atherogenicity ( P = 0.02), glycated hemoglobin ( P < 0.001), estimated mean glucose ( P = 0.02), and alanine aminotransferase ( P = 0.02), unlike the hypocaloric diet control group, which did not show such significance. CONCLUSIONS In this study, from the perspective of body composition, similar changes were observed between the group subjected to CR alone and the group subjected to CR combined with a 12-hour overnight fast. However, there was an indication of superior improvement in glycemic and lipid parameters in the group subjected to the 12-hour overnight fast. These findings suggest the potential for TRE, as implemented, to have positive effects on reducing cardiovascular risk and other chronic metabolic diseases.
Collapse
Affiliation(s)
- Nathália Cavalcanti de Morais Araújo
- Experimental Nutrition and Metabolism Research Group, Nutrition Department, Health Science Center, Universidade Federal de Pernambuco, Recife, Brazil
| | - Júlia Acioli Paixão
- Experimental Nutrition and Metabolism Research Group, Nutrition Department, Health Science Center, Universidade Federal de Pernambuco, Recife, Brazil
| | - Fabiane de Oliveira Freitas
- Experimental Nutrition and Metabolism Research Group, Nutrition Department, Health Science Center, Universidade Federal de Pernambuco, Recife, Brazil
| | | | - Fernando Wesley Cavalcanti de Araujo
- Experimental Nutrition and Metabolism Research Group, Nutrition Department, Health Science Center, Universidade Federal de Pernambuco, Recife, Brazil
| | - Silvia Alves da Silva
- Nutrition Department, Academic Center of Vitória, Universidade Federal de Pernambuco, Vitória de Santo Antão, Brazil
| | - Elizabeth do Nascimento
- Experimental Nutrition and Metabolism Research Group, Nutrition Department, Health Science Center, Universidade Federal de Pernambuco, Recife, Brazil
| |
Collapse
|
13
|
Ozlu Karahan T, Yilmaz Akyuz E, Yilmaz Karadag D, Yilmaz Y, Eren F. Effects of Intermittent Fasting on Liver Steatosis and Fibrosis, Serum FGF-21 and Autophagy Markers in Metabolic Dysfunction-Associated Fatty Liver Disease: A Randomized Controlled Trial. Life (Basel) 2025; 15:696. [PMID: 40430125 PMCID: PMC12113254 DOI: 10.3390/life15050696] [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: 03/15/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND This randomized controlled study sought to determine the effect of intermittent fasting on anthropometric measurements, fibroblast growth factor (FGF)-21, and autophagy markers, as well as on hepatic steatosis and fibrosis levels in overweight or obese patients with metabolic dysfunction-associated fatty liver disease (MAFLD). METHODS Patients were randomly assigned into two groups: received a dietary treatment involving 22-25 kcal/kg/day of energy for 8 weeks and followed the same dietary intervention and a 16:8 pattern. The extent of hepatic steatosis and fibrosis was determined using transient elastography on a FibroScan® device. The controlled attenuation parameter (CAP) and liver stiffness measurement (LSM), determined by transient elastography, reflect hepatic steatosis and fibrosis, respectively. In duplicate, serum levels of FGF-21, Beclin-1, and ATG-5 were determined using enzyme-linked immunosorbent assay. RESULTS The study included 48 patients with a mean age of 48.2 ± 1.4 years (27 female and 21 male). Improvements in anthropometric measurement and CAP and LSM levels and a decrease in serum FGF-21 levels were found in both groups (p < 0.05). Changes in the CAP and FGF-21 levels were higher in the energy + time-restricted diet group (p < 0.05). Autophagy-related protein (ATG)-5 levels increased only in the energy + time-restricted diet group [(0.74 (0.46-1.29) ng/mL vs. 0.95 (0.73-1.32) ng/mL, p = 0.03]. CONCLUSIONS Intermittent fasting was potentially practical in the management of MAFLD. In particular, changes in FGF-21 and ATG-5 levels indicate the potential of intermittent fasting to regulate metabolic processes and autophagy. However, methodological limitations should be taken into consideration when interpreting the study results.
Collapse
Affiliation(s)
- Tugce Ozlu Karahan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Istanbul Bilgi University, Istanbul 34440, Turkey;
| | - Elvan Yilmaz Akyuz
- Department of Nutrition and Dietetics, Hamidiye Faculty of Health Sciences, University of Health Sciences, Istanbul 34668, Turkey;
| | | | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize 53100, Turkey;
| | - Fatih Eren
- Department of Medical Biology, School of Medicine, Recep Tayyip Erdoğan University, Rize 53100, Turkey
| |
Collapse
|
14
|
Zhang MC, Zhu YZ, Tong YT, Wu HY, Shi CL, Ying Ding, Li W, Liu XF, Yi YY. Postoperative Intermittent Fasting Improves Outcome of Autologous Fat Grafting in Mice. Aesthetic Plast Surg 2025:10.1007/s00266-025-04796-x. [PMID: 40278876 DOI: 10.1007/s00266-025-04796-x] [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: 11/27/2024] [Accepted: 03/05/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Autologous fat grafting confronts challenges of inconsistent retention and complications. Intermittent fasting (IF), an emerging dietary management strategy, shows potential in tissue repair and fat metabolism, and yet to know in fat grafting. OBJECTIVES The aim of this study is to address the impact of 16:8 IF on the outcome of fat grafting in mice. METHODS Male C57BL/6 mice were randomly assigned to postoperative IF regimen group (n = 24) and ad libitum group with unrestricted feeding (n = 24). For postoperative IF group, animals were put on a feeding schedule with 8 hours of unrestricted access to standard diet per day followed by 16-h fasting period after fat grafting. Fat grafts were harvested at 2, 4 and 12 weeks postoperatively. We addressed the mass retention and graft quality through weighting and ultrasound examination. Histological remodeling of fat grafts was evaluated by Masson staining and immunofluorescence staining. RESULTS In comparison with unrestricted feeding, postoperative IF strategies improved the mass retention of fat grafts, and optimized the outcomes characterized by enhanced adipogenesis, accelerated revascularization, facilitated M2 macrophage infiltration as well as reduced fibrosis and oil cyst formation. CONCLUSION Postoperative IF improved the retention and outcomes of fat grafting in mice, and could be suggested as a dietary intervention strategy after fat grafting clinically. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Min-Chen Zhang
- Department of Plastic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Province Key laboratory of Precision Cell Therapy, Jiangxi Medical College, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Yuan-Zheng Zhu
- Department of Plastic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Province Key laboratory of Precision Cell Therapy, Jiangxi Medical College, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Ya-Ting Tong
- Department of Plastic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Province Key laboratory of Precision Cell Therapy, Jiangxi Medical College, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Heng-Yu Wu
- Department of Plastic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Province Key laboratory of Precision Cell Therapy, Jiangxi Medical College, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Chen-Long Shi
- Department of Plastic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Ying Ding
- Department of Plastic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Wei Li
- Department of Plastic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Xue-Fei Liu
- Department of Plastic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Yang-Yan Yi
- Department of Plastic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China.
- Jiangxi Province Key laboratory of Precision Cell Therapy, Jiangxi Medical College, Nanchang, Jiangxi, 330006, People's Republic of China.
| |
Collapse
|
15
|
Sharafifard F, Kazeminasab F, Ghanbari Rad M, Ghaedi K, Rosenkranz SK. The combined effects of high-intensity interval training and time-restricted feeding on the AKT/FOXO1/PEPCK pathway in diabetic rats. Sci Rep 2025; 15:13898. [PMID: 40263494 PMCID: PMC12015413 DOI: 10.1038/s41598-025-96703-2] [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: 12/27/2024] [Accepted: 03/31/2025] [Indexed: 04/24/2025] Open
Abstract
High-intensity interval training (HIIT) and time-restricted feeding (TRF) have shown promise for improving glucose regulation by increasing insulin sensitivity, enhancing glucose uptake, reducing glucose production. Therefore, this study investigates the combined effects of HIIT and TRF on the AKT/FOXO1/PEPCK signaling pathway in the liver tissue of type 2 diabetic rats. 42 male Wistar rats (4-5 weeks of age) were included in the study. The animals were randomly divided into two groups: (1) Standard diet (SD, non-Diabetic (Non-D, n = 7) (2) High-fat diet (HFD, n = 35) for 4 weeks. To induce diabetes, 35 mg/kg of streptozotocin (STZ) was injected intraperitoneally (IP). Animals with blood glucose levels of > 250 mg/dL were considered as diabetic. Diabetic rats were randomly divided into 5 groups (n = 7): (1) Diabetes-exercise (D-EX), (2) Diabetes-TRF (D-TRF), (3) Diabetes-combined TRF and exercise (D-TRF&EX), (4) Diabetes no treatment (D-NT), (5) Diabetes with metformin (D-MET). Interventions (HIIT and TRF) were performed for 10 weeks. Rats in the Non-D group did not exercise and did not receive metformin or TRF. Periodic Acid-Schiff (PAS) staining was used to histologically analyze the liver tissue. Levels of blood glucose, insulin resistance (IR), FOXO1 protein, PEPCK, and area under the curve (AUC) following the IPGTT test, were significantly decreased in treatment groups compared to the D-NT group (p < 0.05). The AKT protein levels (p < 0.01), glycogen content (p < 0.05), and insulin sensitivity (p < 0.001) increased in the treatment groups as compared with the D-NT group. Microscopic examination of the liver tissue in general showed a better tissue arrangement in both treatment groups than in the D-NT group. Combining HIIT and TRF may be effective for improving blood glucose regulation, insulin sensitivity, in type 2 diabetes, as compared to TRF or HIIT interventions alone.
Collapse
Affiliation(s)
- Fatemeh Sharafifard
- Department of Physical Education and Sports Science, Faculty of Humanities, University of Kashan, Kashan, Iran
| | - Fatemeh Kazeminasab
- Department of Physical Education and Sports Science, Faculty of Humanities, University of Kashan, Kashan, Iran.
| | - Mahtab Ghanbari Rad
- Gerash Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Kamran Ghaedi
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Sara K Rosenkranz
- Department of Kinesiology and Nutrition Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| |
Collapse
|
16
|
Mohebinejad M, Kazeminasab F, Ghanbari Rad M, Bagheri R, Razi M, Willoughby D, Dutheil F. The Combined Effect of High-Intensity Interval Training and Time-Restricted Feeding on the AKT-IGF-1-mTOR Signaling Pathway in the Muscle Tissue of Type 2 Diabetic Rats. Nutrients 2025; 17:1404. [PMID: 40362714 PMCID: PMC12073226 DOI: 10.3390/nu17091404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: High-intensity interval training (HIIT) and time-restricted feeding (TRF) have shown potential in enhancing glucose metabolism, increasing insulin sensitivity, and promoting muscle health. This study investigates the combined effects of HIIT and TRF on the AKT-IGF-1-mTOR signaling pathway in the muscle tissue of type 2 diabetic (T2D) rats. Methods: 42 male Wistar rats (4-5 weeks of age) were included in the study. The animals were randomly divided into two groups: 1. Standard diet (SD) non-diabetic (n = 7) and 2. High-fat diet (HFD n = 35) for 4 weeks. T2D was induced by intraperitoneal injection (IP) of streptozotocin (STZ) at 35 mg/kg. Animals with blood glucose levels ≥ 250 mg/dL were considered diabetic. Diabetic rats were randomly divided into five groups (n = 7): 1. Diabetes-HIIT (D-HIIT), 2. Diabetes-TRF (D-T), 3. Diabetes-combined TRF and HIIT (D-T+HIIT), 4. Diabetes-Untreated Control (D), and 5. Diabetes with metformin (D-MET). The HIIT protocol and TRF regimen were followed for 10 weeks. Muscle tissue was collected for histological analysis, and the expression of proteins related to the AKT-IGF-1-mTOR pathway was measured. Results: Blood glucose levels, insulin resistance (IR), and markers of muscle degradation were significantly improved in the D-T+HIIT and D-MET groups compared to the non-diabetes group. Furthermore, the activation of the AKT and mTOR signaling proteins, as well as increased IGF-1 expression, was significantly elevated in the D-T+HIIT group compared to the diabetic control group and other treatment groups, and approached levels observed in the non-diabetes group. Additionally, muscle fiber size and overall tissue structure were improved in the treatment groups, particularly in the D-T+HIIT group. Conclusions: The combination of HIIT and TRF appears to offer superior benefits in improving muscle protein synthesis, and glucose regulation in T2D rats, as compared to either HIIT or TRF alone. These findings highlight the potential of this combined approach for addressing muscle-related complications in T2D.
Collapse
Affiliation(s)
- Motahareh Mohebinejad
- Department of Physical Education and Sports Science, Faculty of Humanities, University of Kashan, Kashan 87317-53153, Iran;
| | - Fatemeh Kazeminasab
- Department of Physical Education and Sports Science, Faculty of Humanities, University of Kashan, Kashan 87317-53153, Iran;
| | - Mahtab Ghanbari Rad
- Gerash Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash 58666-74417, Iran;
| | - Reza Bagheri
- Department of Exercise Physiology, University of Isfahan, Isfahan 81746-73441, Iran;
| | - Mazdak Razi
- Division of Comparative Histology and Embryology, Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia 57561-51818, Iran;
| | - Darryn Willoughby
- Department of Education, Innovation, and Technology, Baylor College of Medicine-School of Medicine, Temple, TX 76513, USA;
| | - Fred Dutheil
- Preventive and Occupational Medicine, Université Clermont Auvergne, CNRS, LaPSCo, Physiological and Psychosocial Stress, CHU Clermont-Ferrand, University Hospital of Clermont-Ferrand, Witty Fit, F-63000 Clermont-Ferrand, France;
| |
Collapse
|
17
|
Gachon F, Bugianesi E, Castelnuovo G, Oster H, Pendergast JS, Montagnese S. Potential bidirectional communication between the liver and the central circadian clock in MASLD. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:15. [PMID: 40225783 PMCID: PMC11981938 DOI: 10.1038/s44324-025-00058-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/25/2025] [Indexed: 04/15/2025]
Abstract
Most aspects of physiology and behaviour fluctuate every 24 h in mammals. These circadian rhythms are orchestrated by an autonomous central clock located in the suprachiasmatic nuclei that coordinates the timing of cellular clocks in tissues throughout the body. The critical role of this circadian system is emphasized by increasing evidence associating disruption of circadian rhythms with diverse pathologies. Accordingly, mounting evidence suggests a bidirectional relationship where disruption of rhythms by circadian misalignment may contribute to liver diseases while liver diseases alter the central clock and circadian rhythms in other tissues. Therefore, liver pathophysiology may broadly impact the circadian system and may provide a mechanistic framework for understanding and targeting metabolic diseases and adjust metabolic setpoints.
Collapse
Affiliation(s)
- Frédéric Gachon
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus, Denmark
| | | | | | - Henrik Oster
- Institute of Neurobiology, Center of Brain, Behavior & Metabolism, University of Lübeck, Lübeck, Germany
| | | | - Sara Montagnese
- Department of Medicine, University of Padova, Padova, Italy
- Chronobiology Section, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
18
|
Feeney SP, McCarthy JM, Petruconis CR, Tudor JC. Sleep loss is a metabolic disorder. Sci Signal 2025; 18:eadp9358. [PMID: 40198749 DOI: 10.1126/scisignal.adp9358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 03/12/2025] [Indexed: 04/10/2025]
Abstract
Sleep loss dysregulates cellular metabolism and energy homeostasis. Highly metabolically active cells, such as neurons, enter a catabolic state during periods of sleep loss, which consequently disrupts physiological functioning. Specific to the central nervous system, sleep loss results in impaired synaptogenesis and long-term memory, effects that are also characteristic of neurodegenerative diseases. In this review, we describe how sleep deprivation increases resting energy expenditure, leading to the development of a negative energy balance-a state with insufficient metabolic resources to support energy expenditure-in highly active cells like neurons. This disruption of energetic homeostasis alters the balance of metabolites, including adenosine, lactate, and lipid peroxides, such that energetically costly processes, such as synapse formation, are attenuated. During sleep loss, metabolically active cells shunt energetic resources away from those processes that are not acutely essential, like memory formation, to support cell survival. Ultimately, these findings characterize sleep loss as a metabolic disorder.
Collapse
Affiliation(s)
- Sierra P Feeney
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Jordan M McCarthy
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Cecilia R Petruconis
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Jennifer C Tudor
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| |
Collapse
|
19
|
Kleckner AS, Clingan CL, Youngblood SM, Kleckner IR, Quick L, Elrod RD, Zhu S, Manoogian ENC, Panda S, Badros AZ, Emadi A. Time-restricted eating to address persistent cancer-related fatigue among cancer survivors: a randomized controlled trial. Support Care Cancer 2025; 33:353. [PMID: 40186671 DOI: 10.1007/s00520-025-09394-w] [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: 11/26/2024] [Accepted: 03/23/2025] [Indexed: 04/07/2025]
Abstract
PURPOSE Time-restricted eating (TRE) helps regulate rest-activity rhythms, blood glucose, and other diurnally regulated energetics processes, which may have implications for persistent fatigue. In a randomized controlled trial, we tested the effects of TRE vs. control on fatigue in cancer survivorship. METHODS Adult cancer survivors were recruited who were 2 months to 2 years post-treatment and reported moderate to severe fatigue. Participants were randomized 1:1, TRE:control, and all received individualized nutrition counseling. The TRE group self-selected a 10-h eating window for 12 weeks. At baseline, week 6, and week 12, participants were asked to log eating instances, complete the Functional Assessment of Chronic Illness Therapy-Fatigue questionnaire (FACIT-F, higher score = less fatigue), and wear an actigraph and continuous glucose monitor. RESULTS Thirty participants completed baseline assessments and were randomized (77% female, 53% Black/African American, 43% White, 7% Hispanic; 54.1 ± 14.7 years old; 87% with blood cancer); 25 completed 12-week assessments. TRE led to a meaningful reduction in fatigue at week 12 controlling for baseline levels (change in FACIT-F fatigue subscale = 0.0 ± 5.4 for control, 4.1 ± 5.7 for TRE, p = 0.11, effect size (ES) = 0.70; clinically meaningful threshold = 3.0 points). Glucose parameters (e.g., average interstitial glucose, average fasting glucose) tended to be lower, and rest-activity rhythms tended to indicate more regularity for those in the TRE vs. control group at weeks 6 and 12, though differences were not statistically significant (p > 0.19). CONCLUSIONS A 12-week, nutritionist-led TRE program led to less fatigue than control. Continued study of TRE patterns are warranted to optimize this eating pattern and address persistent cancer-related fatigue. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05256888.
Collapse
Affiliation(s)
- Amber S Kleckner
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, 655 W. Lombard Ave., 7th Floor, Baltimore, MD, 21201, USA.
- Greenebaum Comprehensive Cancer Center, 22 S. Greene St., Baltimore, MD, 21201, USA.
| | - Carin L Clingan
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, 655 W. Lombard Ave., 7th Floor, Baltimore, MD, 21201, USA
| | - Shari M Youngblood
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, 655 W. Lombard Ave., 7th Floor, Baltimore, MD, 21201, USA
- Department of Integrative and Functional Nutrition, Saybrook University, 55 W. Eureka St., Pasadena, CA, USA
| | - Ian R Kleckner
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, 655 W. Lombard Ave., 7th Floor, Baltimore, MD, 21201, USA
- Greenebaum Comprehensive Cancer Center, 22 S. Greene St., Baltimore, MD, 21201, USA
| | - Lauren Quick
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, 655 W. Lombard Ave., 7th Floor, Baltimore, MD, 21201, USA
- Community College of Baltimore County, 800 S. Rolling Rd., Baltimore, MD, 21228, USA
| | - Rebecca D Elrod
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, 655 W. Lombard Ave., 7th Floor, Baltimore, MD, 21201, USA
| | - Shijun Zhu
- Department of Organizational Systems and Adult Health, University of Maryland School of Nursing, 655 W. Lombard Ave., Room 402H, Baltimore, MD, 21201, USA
| | - Emily N C Manoogian
- Regulatory Biology Laboratory, The Salk Institute for Biological Sciences, La Jolla, CA, 92037, USA
| | - Satchidananda Panda
- Regulatory Biology Laboratory, The Salk Institute for Biological Sciences, La Jolla, CA, 92037, USA
| | - Ashraf Z Badros
- Greenebaum Comprehensive Cancer Center, 22 S. Greene St., Baltimore, MD, 21201, USA
- Department of Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD, 21201, USA
| | - Ashkan Emadi
- Greenebaum Comprehensive Cancer Center, 22 S. Greene St., Baltimore, MD, 21201, USA
- Department of Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD, 21201, USA
- Department of Medical Oncology, West Virginia University Cancer Institute, 1 Medical Center Dr., Morgantown, WV, 26506, USA
| |
Collapse
|
20
|
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
|
21
|
Mattson MP. The cyclic metabolic switching theory of intermittent fasting. Nat Metab 2025; 7:665-678. [PMID: 40087409 DOI: 10.1038/s42255-025-01254-5] [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: 09/25/2024] [Accepted: 02/19/2025] [Indexed: 03/17/2025]
Abstract
Intermittent fasting (IF) and ketogenic diets (KDs) have recently attracted much attention in the scientific literature and in popular culture and follow a longer history of exercise and caloric restriction (CR) research. Whereas IF involves cyclic metabolic switching (CMS) between ketogenic and non-ketogenic states, KDs and CR may not. In this Perspective, I postulate that the beneficial effects of IF result from alternating between activation of adaptive cellular stress response pathways during the fasting period, followed by cell growth and plasticity pathways during the feeding period. Thereby, I establish the cyclic metabolic switching (CMS) theory of IF. The health benefits of IF may go beyond those seen with continuous CR or KDs without CMS owing to the unique interplay between the signalling functions of the ketone β-hydroxybutyrate, mitochondrial adaptations, reciprocal activation of autophagy and mTOR pathways, endocrine and paracrine signalling, gut microbiota, and circadian biology. The CMS theory may have important implications for future basic research, clinical trials, development of pharmacological interventions, and healthy lifestyle practices.
Collapse
Affiliation(s)
- Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
22
|
Steele C, Ostrow A, Wang W, Coleman E, George D, Bing K, Ramanathan S, Gregory A, Gitomer BY, Kline TL, Thomas E, Chonchol M, Nowak KL. Time-restricted eating and autosomal dominant polycystic kidney disease: a pilot, randomized clinical trial. Clin Kidney J 2025; 18:sfaf069. [PMID: 40207097 PMCID: PMC11976525 DOI: 10.1093/ckj/sfaf069] [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: 10/03/2024] [Indexed: 04/11/2025] Open
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) is the most commonly inherited progressive kidney disease. Time-restricted eating (TRE) is a fasting regimen that restricts eating to a particular window (typically 8 hours/day), which could slow cyst growth based on preclinical models. Methods A 12-month, randomized, controlled, behavioral dietary intervention compared TRE with a control group given healthy eating advice without TRE (HE), without caloric restriction. Participants underwent baseline and 12-month measurements, including adherence (percentage of participants adhering to the 8-hour window; primary outcome), and MRI to determine height-adjusted total kidney volume (htTKV) and adiposity. Results Twenty-nine participants (23 females, mean standard ± deviation 48 ± 9 years) with a body mass index of 31.1±5 kg/m2 were randomized to TRE (n = 14) or HE (n = 15). Of the total participants, 71% (n = 10) of TRE and 87% (n = 13) of HE participants completed the intervention. The eating window was 9.6 ± 3.6 hours for TRE (60% achieving the 8-hour window) and 12.0 ± 2.0 for HE groups (P = .07). At month 12, both groups lost modest weight (-2.4 ± 6.4% and -3.6 ± 5.4% in the TRE and HE groups, respectively). Annual change in htTKV was 3.0 ± 8.5% and 4.6 ± 8.8% in the TRE and HE groups, respectively. Both change in weight (r = 0.67, P < .01) and change in visceral adiposity (r = 0.54, P < .01) were positively correlated with change in htTKV. Conclusion Both the TRE and HE group lost modest weight at 12 months. The targeted TRE adherence of ≥75% of participants was not achieved. Weight and adiposity loss may be more important drivers of kidney growth than the timing of eating.
Collapse
Affiliation(s)
- Cortney Steele
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Anna Ostrow
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Wei Wang
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Erin Coleman
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Diana George
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristen Bing
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Berenice Y Gitomer
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Elizabeth Thomas
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michel Chonchol
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristen L Nowak
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
23
|
Hunter AL, Bechtold DA. The metabolic significance of peripheral tissue clocks. Commun Biol 2025; 8:497. [PMID: 40140664 PMCID: PMC11947457 DOI: 10.1038/s42003-025-07932-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
The circadian clock is a transcriptional-translational feedback loop which oscillates in virtually all nucleated cells of the body. In the decades since its discovery, it has become evident that the molecular clockwork is inextricably linked to energy metabolism. Given the frequency with which metabolic dysfunction and clock disruption co-occur, understanding why and how clock and metabolic processes are reciprocally coupled will have important implications for supporting human health and wellbeing. Here, we discuss the relevance of molecular clock function in metabolic tissues and explore its role not only as a driver of day-night variation in gene expression, but as a key mechanism for maintaining metabolic homeostasis in the face of fluctuating energy supply and demand.
Collapse
Affiliation(s)
- A Louise Hunter
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK.
- Diabetes, Endocrinology & Metabolism Centre, Oxford Road Campus, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK.
| | - David A Bechtold
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
24
|
de Melo JML, Blond MB, Jensen VH, Pedersen H, Clemmensen KKB, Jensen MM, Færch K, Quist JS, Størling J. Time-restricted eating in people at high diabetes risk does not affect mitochondrial bioenergetics in peripheral blood mononuclear cells and platelets. Sci Rep 2025; 15:10175. [PMID: 40128559 PMCID: PMC11933372 DOI: 10.1038/s41598-025-94652-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/17/2025] [Indexed: 03/26/2025] Open
Abstract
Overweight and obesity are linked to mitochondrial alterations, impaired glucose tolerance and a high risk of type 2 diabetes. Time-restricted eating (TRE) may aid in facilitating weight loss to prevent diabetes. Here, we investigated if TRE in individuals with overweight and prediabetes or obesity affects mitochondrial bioenergetics of peripheral blood mononuclear cells (PBMCs) and platelets using the Seahorse extracellular flux technology. In a 3-month randomized controlled trial, PBMCs/platelets were analyzed from 52 participants before and after a TRE intervention with a 10-h eating window or habitual living. PBMC and platelet respiratory function was evaluated through sequential addition of substrates, uncouplers, and inhibitors in living cells. After 3 months, there were no statistically significant differences in mitochondrial respiration within or between the TRE and control groups. Association analyses between PBMC/platelet respiration and clinical parameters including body mass index and fat mass showed no significant effects. In conclusion, 3 months of 10-h TRE does not alter the mitochondrial bioenergetics of PBMCs and platelets in individuals with high risk of type 2 diabetes.
Collapse
Affiliation(s)
- Joana Mendes Lopes de Melo
- Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730, Herlev, Denmark
- Novo Nordisk A/S, Måløv, Denmark
| | - Martin Bæk Blond
- Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730, Herlev, Denmark
| | - Verena Hirschberg Jensen
- Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730, Herlev, Denmark
| | - Hanne Pedersen
- Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730, Herlev, Denmark
- Novo Nordisk A/S, Søborg, Denmark
| | - Kim Katrine Bjerring Clemmensen
- Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730, Herlev, Denmark
- Novo Nordisk A/S, Søborg, Denmark
| | - Marie Møller Jensen
- Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730, Herlev, Denmark
| | - Kristine Færch
- Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730, Herlev, Denmark
- Novo Nordisk A/S, Søborg, Denmark
| | - Jonas Salling Quist
- Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730, Herlev, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- School of Psychology, University of Leeds, Leeds, UK
| | - Joachim Størling
- Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730, Herlev, Denmark.
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
25
|
Chen J, Xiang J, Zhou M, Huang R, Zhang J, Cui Y, Jiang X, Li Y, Zhou R, Xin H, Li J, Li L, Lam SM, Zhu J, Chen Y, Yang Q, Xie Z, Shui G, Deng F, Zhang Z, Li MD. Dietary timing enhances exercise by modulating fat-muscle crosstalk via adipocyte AMPKα2 signaling. Cell Metab 2025:S1550-4131(25)00065-8. [PMID: 40088888 DOI: 10.1016/j.cmet.2025.02.007] [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: 01/16/2025] [Accepted: 02/22/2025] [Indexed: 03/17/2025]
Abstract
Feeding rhythms regulate exercise performance and muscle energy metabolism. However, the mechanisms regulating adipocyte functions remain unclear. Here, using multi-omics analyses, involving (phospho-)proteomics and lipidomics, we found that day-restricted feeding (DRF) regulates diurnal rhythms of the mitochondrial proteome, neutral lipidome, and nutrient-sensing pathways in mouse gonadal white adipose tissue (GWAT). Adipocyte-specific knockdown of Prkaa2 (the gene encoding AMPKα2) impairs physical endurance. This defect is associated with altered rhythmicity in acyl-coenzyme A (CoA) metabolism-related genes, a loss of rhythmicity in the GWAT lipidome, and circadian remodeling of serum metabolites-in particular, lactate and succinate. We also found that adipocyte Prkaa2 regulates muscle clock genes during DRF. Notably, oral administration of the AMPK activator C29 increases endurance and muscle functions in a time-of-day manner, which requires intact adipocyte AMPKα2 signaling. Collectively, our work defines adipocyte AMPKα2 signaling as a critical regulator of circadian metabolic coordination between fat and muscle, thereby enhancing exercise performance.
Collapse
Affiliation(s)
- Jianghui Chen
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Jing Xiang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Meiyu Zhou
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Rongfeng Huang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China; Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610072, China
| | - Jianxin Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China; Department of Cardiology, The 960th Hospital of the PLA Joint Service Support Force, Jinan 250000, China
| | - Yuanting Cui
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Xiaoqing Jiang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Yang Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Runchao Zhou
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Haoran Xin
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Jie Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Lihua Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; LipidALL Technologies Company Limited, Changzhou, China
| | - Jianfang Zhu
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Yanxiu Chen
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Qingyuan Yang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Zhifu Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Fang Deng
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing 400038, China; Key Laboratory of High Altitude Medicine, PLA, Chongqing 400038, China
| | - Zhihui Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China.
| | - Min-Dian Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China.
| |
Collapse
|
26
|
Rivera-Alvarez I, Vázquez-Lizárraga R, Mendoza-Viveros L, Sotelo-Rivera I, Viveros-Ruiz TL, Morales-Maza J, Orozco L, Romano MC, Noriega LG, Tovar AR, Aguilar-Arnal L, Cruz-Bautista I, Aguilar-Salinas C, Orozco-Solis R. Transcriptional dynamics in type 2 diabetes progression is linked with circadian, thermogenic, and cellular stress in human adipose tissue. Commun Biol 2025; 8:398. [PMID: 40057615 PMCID: PMC11890630 DOI: 10.1038/s42003-025-07709-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 02/10/2025] [Indexed: 04/03/2025] Open
Abstract
The prevalence of type 2 diabetes (T2D) has increased significantly over the past three decades, with an estimated 30-40% of cases remaining undiagnosed. Brown and beige adipose tissues are known for their remarkable catabolic capacity, and their ability to diminish blood glucose plasma concentration. Beige adipose tissue can be differentiated from adipose-derived stem cells or through transdifferentiation from white adipocytes. However, the impact of T2D progression on beige adipocytes' functional capacity remains unclear. Transcriptomic profiling of subcutaneous adipose tissue biopsies from healthy normal-weight, obese, prediabetic obese, and obese subjects diagnosed with T2D, reveals a progressive alteration in cellular processes associated with catabolic metabolism, circadian rhythms, thermogenesis-related signaling pathways, cellular stress, and inflammation. MAX is a potential transcription factor that links inflammation with the circadian clock and thermogenesis during the progression of T2D. This study unveils an unrecognized transcriptional circuit that increasingly disrupts subcutaneous adipose tissue oxidative capacity during the progression of T2D. These findings could open new research venues for developing chrono-pharmaceutical strategies to treat and prevent T2D.
Collapse
Affiliation(s)
| | - Rosa Vázquez-Lizárraga
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
- Centro de Investigación sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE-CINVESTAV), México City, México
| | - Lucía Mendoza-Viveros
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, San Luis Potosi, S.L.P., México
| | | | - Tannia L Viveros-Ruiz
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Jesús Morales-Maza
- Departamento de Cirugía Endocrina, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Lorena Orozco
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
| | - Marta C Romano
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y Estudios Avanzados (CINVESTAV), México City, México
| | - Lilia G Noriega
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Armando R Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Lorena Aguilar-Arnal
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), México City, México
| | - Ivette Cruz-Bautista
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Carlos Aguilar-Salinas
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, México City, México
| | - Ricardo Orozco-Solis
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México.
- Centro de Investigación sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE-CINVESTAV), México City, México.
| |
Collapse
|
27
|
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
|
28
|
Fleming N. Fasting for weight loss is all the rage: what are the health benefits? Nature 2025; 639:855-857. [PMID: 40133618 DOI: 10.1038/d41586-025-00895-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
|
29
|
Gao Z, Yu Y, Eckel‐Mahan K, Kolonin MG. Caloric Restriction and Telomere Preservation in TERT Knockout Adipocyte Progenitors Does Not Rescue Mice From Metabolic Dysfunction due to a TERT Function in Adipocyte Mitochondria. Aging Cell 2025; 24:e14499. [PMID: 39932851 PMCID: PMC11896407 DOI: 10.1111/acel.14499] [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: 11/04/2024] [Revised: 12/20/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
Inactivation of telomerase (TERT) in adipocyte progenitor cells (APC) expedites telomere attrition, and the onset of diabetes in mice fed high-fat diet (HFD), which promotes APC over-proliferation and replicative senescence. Here, we show that time-restricted feeding or caloric restriction in the postnatal development of mice subsequently subjected to HFD prevents telomere attrition but not glucose intolerance. This metabolic effect of dietary intervention was not observed for mice with TERT KO in endothelial or myeloid cells. To characterize the telomere-independent effects of TERT in the APC lineage, we analyzed mice with TERT knockout in mature adipocytes (AD-TERT-KO), which do not proliferate and avoid telomere attrition. Analysis of adipocytes from AD-TERT-KO mice indicated reliance on glycolysis and decreased mitochondrial oxidative metabolism. We show that AD-TERT-KO mice have reduced cold tolerance and metabolism abnormality indicating a defect in adaptive thermogenesis, characteristic of aging. Conversely, ectopic TERT expression in brown adipocytes-induced mitochondrial oxidation and thermogenic gene expression. We conclude that TERT plays an important non-canonical function in the mitochondria of adipocytes.
Collapse
Affiliation(s)
- Zhanguo Gao
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Yongmei Yu
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Kristin Eckel‐Mahan
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Mikhail G. Kolonin
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| |
Collapse
|
30
|
Tan LJ, Shin S. Impact of eating duration on weight management, sleeping quality, and psychological stress: A pilot study. J Nutr Biochem 2025; 137:109835. [PMID: 39701471 DOI: 10.1016/j.jnutbio.2024.109835] [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: 09/10/2024] [Revised: 12/09/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
The daily eating window significantly impacts weight and metabolic health, yet its ideal duration remains uncertain. Thirty-four healthy middle-aged women were randomly assigned to two intervention groups: 8-h time-restricted eating (TRE) and 14-h time-extended eating (EXE). Each intervention lasted 4 wk, with a 16-d washout period before switching to the other intervention. Clinical biomarkers were collected before and after each intervention, and sleep quality was assessed using the Korean Version of the Pittsburgh Sleep Quality Index (PSQI-K). Additionally, a daily visual analogue scale (VAS) was used to evaluate psychological changes. The TRE group experienced significant weight reduction, lower fasting plasma glucose and total serum cholesterol levels compared to the EXE group, but with an increase in systolic blood pressure. The EXE group showed improved blood pressure. The TRE group reported higher stress levels on the VAS, but the PSQI-K indicated improved sleep quality during the second intervention. An 8-h TRE, without calorie restriction or diet composition changes, proves more beneficial for weight management and plasma glucose control compared to the 14-h EXE among Korean women. Implementation of this approach is recommended to be gradual to mitigate psychological fluctuations and adverse blood pressure changes. The trial was registered with ClinicalTrials.gov (ID: NCT05964179) and Clinical Research Information Service (CRIS, Korea) (ID: KCT0008640).
Collapse
Affiliation(s)
- Li-Juan Tan
- Department of Critical Care Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China; Nutritional Epidemiology Laboratory, Department of Food and Nutrition, Chung-Ang University, Gyeonggi-do, 17546, South Korea.
| | - Sangah Shin
- Nutritional Epidemiology Laboratory, Department of Food and Nutrition, Chung-Ang University, Gyeonggi-do, 17546, South Korea.
| |
Collapse
|
31
|
Wang R, Liao Y, Deng Y, Shuang R. Unraveling the Health Benefits and Mechanisms of Time-Restricted Feeding: Beyond Caloric Restriction. Nutr Rev 2025; 83:e1209-e1224. [PMID: 38954563 DOI: 10.1093/nutrit/nuae074] [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: 07/04/2024] Open
Abstract
Time-restricted feeding (TRF) is a lifestyle intervention that aims to maintain a consistent daily cycle of feeding and fasting to support robust circadian rhythms. Recently, it has gained scientific, medical, and public attention due to its potential to enhance body composition, extend lifespan, and improve overall health, as well as induce autophagy and alleviate symptoms of diseases like cardiovascular diseases, type 2 diabetes, neurodegenerative diseases, cancer, and ischemic injury. However, there is still considerable debate on the primary factors that contribute to the health benefits of TRF. Despite not imposing strict limitations on calorie intake, TRF consistently led to reductions in calorie intake. Therefore, while some studies suggest that the health benefits of TRF are primarily due to caloric restriction (CR), others argue that the key advantages of TRF arise not only from CR but also from factors like the duration of fasting, the timing of the feeding period, and alignment with circadian rhythms. To elucidate the roles and mechanisms of TRF beyond CR, this review incorporates TRF studies that did not use CR, as well as TRF studies with equivalent energy intake to CR, which addresses the previous lack of comprehensive research on TRF without CR and provides a framework for future research directions.
Collapse
Affiliation(s)
- Ruhan Wang
- Department of Nutrition Hygiene and Toxicology, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 43000, China
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 43000, China
| | - Yan Deng
- Department of Nutrition Hygiene and Toxicology, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 43000, China
| | - Rong Shuang
- Department of Nutrition Hygiene and Toxicology, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 43000, China
| |
Collapse
|
32
|
Halder SK, Melkani GC. The Interplay of Genetic Predisposition, Circadian Misalignment, and Metabolic Regulation in Obesity. Curr Obes Rep 2025; 14:21. [PMID: 40024983 PMCID: PMC11872776 DOI: 10.1007/s13679-025-00613-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2025] [Indexed: 03/04/2025]
Abstract
PURPOSE OF REVIEW This review explores the complex interplay between genetic predispositions to obesity, circadian rhythms, metabolic regulation, and sleep. It highlights how genetic factors underlying obesity exacerbate metabolic dysfunction through circadian misalignment and examines promising interventions to mitigate these effects. RECENT FINDINGS Genome-wide association Studies (GWAS) have identified numerous Single Nucleotide Polymorphisms (SNPs) associated with obesity traits, attributing 40-75% heritability to body mass index (BMI). These findings illuminate critical links between genetic obesity, circadian clocks, and metabolic processes. SNPs in clock-related genes influence metabolic pathways, with disruptions in circadian rhythms-driven by poor sleep hygiene or erratic eating patterns-amplifying metabolic dysfunction. Circadian clocks, synchronized with the 24-h light-dark cycle, regulate key metabolic activities, including glucose metabolism, lipid storage, and energy utilization. Genetic mutations or external disruptions, such as irregular sleep or eating habits, can destabilize circadian rhythms, promoting weight gain and metabolic disorders. Circadian misalignment in individuals with genetic predispositions to obesity disrupts the release of key metabolic hormones, such as leptin and insulin, impairing hunger regulation and fat storage. Interventions like time-restricted feeding (TRF) and structured physical activity offer promising strategies to restore circadian harmony, improve metabolic health, and mitigate obesity-related risks.
Collapse
Affiliation(s)
- Sajal Kumar Halder
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Girish C Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- UAB Nathan Shock Center, Birmingham, AL, 35294, USA.
| |
Collapse
|
33
|
Rumanova VS, Foppen E, Okuliarova M, Zeman M, Kalsbeek A. Time-restricted feeding does not improve daily rhythms in locomotion and drinking disrupted by artificial light at night. Physiol Behav 2025; 290:114780. [PMID: 39662693 DOI: 10.1016/j.physbeh.2024.114780] [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: 11/08/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 12/13/2024]
Abstract
Exposure to artificial light at night (ALAN) disrupts natural darkness and desynchronizes daily rhythms in physiological processes and behavior. Previously, in rats, we have shown that dim ALAN disturbed the central circadian control and the temporal organization of behavior, and hormonal and metabolic pathways. The measurements of undisturbed daily behavioral (locomotor activity, feeding and drinking) patterns revealed reduced amplitudes and a transitory activity peak in the middle of the light (i.e. resting) period. Recent studies indicated that time-restricted feeding during the active period (TRFd) can strengthen daily rhythms and improve metabolic health. Therefore, the aim of our study was to prevent the dim ALAN-induced attenuation of daily behavioral rhythms by applying TRFd. Male Wistar rats were kept in a 12/12 light/dark cycle in metabolic cages for one week with free access to food and water. After acclimation, rats were divided into two groups: 1) ad libitum food or 2) time-restricted food during the dark period. After one week, both groups were exposed to dim ALAN for two weeks. Despite the enhanced amplitude of the daily feeding rhythm in TRFd animals, ALAN still suppressed the rhythm of locomotor activity, induced the extra peak during the resting period and reduced the bimodal pattern during the night. Furthermore, TRFd did not prevent the drop in anticipatory thirst caused by ALAN at the end of the active period. In conclusion, TRFd was not able to fully prevent the weakning of daily behavioral rhythms by dim ALAN.
Collapse
Affiliation(s)
- Valentina Sophia Rumanova
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, the Netherlands; Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia; Laboratory of Endocrinology, Amsterdam University Medical Centers, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam, the Netherlands.
| | - Ewout Foppen
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, the Netherlands; Laboratory of Endocrinology, Amsterdam University Medical Centers, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam, the Netherlands.
| | - Monika Okuliarova
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia.
| | - Michal Zeman
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia.
| | - Andries Kalsbeek
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, the Netherlands; Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Laboratory of Endocrinology, Amsterdam University Medical Centers, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam, the Netherlands.
| |
Collapse
|
34
|
Geng F, Zhao N, Ren Q. Circadian rhythm, microglia-mediated neuroinflammation, and Alzheimer's disease. Neurosci Biobehav Rev 2025; 170:106044. [PMID: 39914702 DOI: 10.1016/j.neubiorev.2025.106044] [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: 09/06/2024] [Revised: 10/16/2024] [Accepted: 02/03/2025] [Indexed: 02/09/2025]
Abstract
Microglia, the brain's resident macrophages, are key mediators of neuroinflammation, responding to immune pathogens and toxins. They play a crucial role in clearing cellular debris, regulating synaptic plasticity, and phagocytosing amyloid-β (Aβ) plaques in Alzheimer's disease (AD). Recent studies indicate that microglia not only exhibit intrinsic circadian rhythms but are also regulated by circadian clock genes, influencing specific functions such as phagocytosis and the modulation of neuroinflammation. Disruption of the circadian rhythm is closely associated with AD pathology. In this review, we will provide an overview of how circadian rhythms regulate microglia-mediated neuroinflammation in the progression of AD, focusing on the pathway from the central nervous system (CNS) and the peripheral immune system. We also discuss potential therapeutic targets, including hormone modulation, lifestyle interventions, and anti-inflammatory therapies, aimed at maintaining brain health in AD. This will shed light on the involvement of circadian rhythm in AD and explore new avenues for AD treatment.
Collapse
Affiliation(s)
- Fan Geng
- Department of Neurology, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing 210009, China
| | - Na Zhao
- Department of Neurology, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing 210009, China
| | - Qingguo Ren
- Department of Neurology, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
35
|
Tofani GSS, Clarke G, Cryan JF. I "Gut" Rhythm: the microbiota as a modulator of the stress response and circadian rhythms. FEBS J 2025; 292:1454-1479. [PMID: 39841560 PMCID: PMC11927059 DOI: 10.1111/febs.17400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/20/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025]
Abstract
Modern habits are becoming more and more disruptive to health. As our days are often filled with circadian disruption and stress exposures, we need to understand how our responses to these external stimuli are shaped and how their mediators can be targeted to promote health. A growing body of research demonstrates the role of the gut microbiota in influencing brain function and behavior. The stress response and circadian rhythms, which are essential to maintaining appropriate responses to the environment, are known to be impacted by the gut microbiota. Gut microbes have been shown to alter the host's response to stress and modulate circadian rhythmicity. Although studies demonstrated strong links between the gut microbiota, circadian rhythms and the stress response, such studies were conducted in an independent manner not conducive to understanding the interface between these factors. Due to the interconnected nature of the stress response and circadian rhythms, in this review we explore how the gut microbiota may play a role in regulating the integration of stress and circadian signals in mammals and the consequences for brain health and disease.
Collapse
Affiliation(s)
- Gabriel S. S. Tofani
- APC MicrobiomeUniversity College CorkIreland
- Department of Anatomy & NeuroscienceUniversity College CorkIreland
| | - Gerard Clarke
- APC MicrobiomeUniversity College CorkIreland
- Department of Psychiatry & Neurobehavioural ScienceUniversity College CorkIreland
| | - John F. Cryan
- APC MicrobiomeUniversity College CorkIreland
- Department of Anatomy & NeuroscienceUniversity College CorkIreland
| |
Collapse
|
36
|
Li T, Huang N, Chen H, Yang Y, Zhang J, Xu W, Gong H, Gong C, Yang M, Zhao T, Wang F, Xiao H. Daytime-Restricted Feeding Alleviates D-Galactose-Induced Aging in Mice and Regulates the AMPK and mTORC1 Activities. J Cell Physiol 2025; 240:e70020. [PMID: 40070151 DOI: 10.1002/jcp.70020] [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: 09/01/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/17/2025]
Abstract
Time-restricted feeding (TRF) is a distinct regimen of intermittent fasting advocated for health improving. Although nighttime TRF (NRF) in rodents is analogous to daytime TRF (DRF) in humans and has health benefits, the effects of DRF on rodent's health remain uncertain. The adverse health effects of DRF in rodents are primarily attributed to its implementation-induced temporal shift in the expression of circadian rhythm-related genes. However, studies also demonstrate the health-beneficial effect of restricted feeding itself on metabolic homeostasis, particularly in periphery tissues. Moreover, the direct effects of DRF on aging progression in rodents are underexplored, highlighting a gap in current research. To explore the overall health effects of long-term DRF in rodents, especially its influence on aging progression, we investigated the impact of long-term DRF on mice under a progeric aging condition. Results showed that both 4-h and 8-h DRF regimens exerted positive effects on aging retardation; these effects were manifested as improved physical and memory capacities, enhanced liver and kidney functions, and reduced oxidative damage and inflammatory response. These DRF regimens also lowered the manifestation of aging-related markers in peripheral tissues, with decreased SA-β-gal staining and p16 expression. Mechanistically, DRF regimens, especially DRF8, upregulated AMPK signaling and downregulated mTORC1 signaling. Interestingly, the health benefits of DRF are similar to those of metformin intervention. In conclusion, our study demonstrates for the first time that DRF effectively counteracts oxidative stress-induced aging progression in mice, supporting the viewpoint that TRF as a promising strategy for preventing aging and aging-related disorders.
Collapse
Affiliation(s)
- Tiepeng Li
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ning Huang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Development and Regeneration Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, China
| | - Honghan Chen
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Yang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Zhang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Weitong Xu
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Gong
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chuhui Gong
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Yang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tingting Zhao
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Fangfang Wang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hengyi Xiao
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
37
|
Chen J, Xiang J, Zhou M, Huang R, Zhang J, Cui Y, Jiang X, Li Y, Zhou R, Xin H, Li J, Li L, Lam SM, Zhu J, Chen Y, Yang Q, Xie Z, Shui G, Deng F, Zhang Z, Li MD. Dietary timing enhances exercise by modulating fat-muscle crosstalk via adipocyte AMPKα2 signaling. Cell Metab 2025. [DOI: pmid: 40088888 doi: 10.1016/j.cmet.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
|
38
|
Hou W, Wang W, Sun C. The associations between evening eating and quality of energy and macronutrients and obesity: the National Health and Nutrition Examination Survey (NHANES), 2003-2016. Nutr J 2025; 24:33. [PMID: 40022155 PMCID: PMC11869462 DOI: 10.1186/s12937-025-01094-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/12/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND This study aimed to investigate the associations between evening eating and quality of energy and macronutrients and obesity among U.S. adults. SUBJECTS/METHODS This study adopted the data from the National Health and Nutrition Examination Survey (2003-2016), which involved a total of 27,911 participants. The differences in the ratios of energy and macronutrients with it is subgroups at dinner versus breakfast (ΔRatio) were categorized into quartiles. The differences in the consumption of 17 types of food at dinner versus breakfast (ΔFoods) were considered as continuous variables. Body mass index (BMI) and waist circumference (WC) were used to define general obesity (30.0 ≤ BMI < 40.0), morbid obesity (BMI ≥ 40.0), and abdominal obesity (WC > 102 cm for men or WC > 88 cm for women). Multiple logistic and linear regression models were developed. RESULTS After a variety of covariates were adjusted, participants in the highest quartile (higher energy/macronutrient intake at dinner than breakfast) of the ΔRatio in terms of energy were positively associated with morbid obesity compared with those in the lowest quartile (ORΔRatio of energy 1.27; 95% CI 1.01;1.61) from fat (ORΔRatio of fat 1.27, 95% CI 1.01;1.60); saturated fatty acids(ORΔRatio of SFA 1.27, 95% CI 1.01;1.59) and unsaturated fatty acids (ORΔRatio of USFA 1.28, 95% CI 1.02;1.5). The highest quartile of the ΔRatio of low-quality carbohydrates was associated with increased odds of abdominal obesity (ORΔRatio of low-quality carbohydrates 1.16; 95%CI 1.03-1.31). Moreover, the ΔRatio of low-quality carbohydrates was significantly positively associated with BMI (coefficient: 0.562, 95% CI: 0.217-0.907). ΔFoods, including whole fruits, other starchy vegetables, added sugars, poultry, dairy, and nuts, were positively associated with obesity. CONCLUSIONS In conclusion, with this nationally representative sample of U.S adults, this study demonstrated that excessive intake of energy at dinner than breakfast during a day was associated with a greater risk of obesity, mainly from low-quality carbohydrates, fat, SFAs, and USFA. This study emphasized the importance of diet quality and evening eating in the prevention of obesity.
Collapse
Affiliation(s)
- Wanying Hou
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Weiqi Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
39
|
Zhang KH, He S, Wang QG, Li JJ, Yao CY, Shan CH, Zhang L, Liu ZY, Liu P, Li MY, Guo Y, Wu ZH. Mistimed Feeding Disrupts Metabolic Rhythm and Increases Lipid Accumulation of Growing Rabbits in Winter. Animals (Basel) 2025; 15:692. [PMID: 40075975 PMCID: PMC11899554 DOI: 10.3390/ani15050692] [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: 01/10/2025] [Revised: 02/08/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Maintaining the normal biological rhythms of livestock is of great significance for reflecting the environmental suitability and welfare level of animals. Mistimed feeding can interfere with the circadian rhythms of both humans and animals, resulting in disorders of lipid metabolism, obesity, and metabolic syndrome. Low-temperature environment stimulates increased appetite and decreased physical activity, resulting in higher energy intake than consumption and thus facilitating fat deposition and even obesity. In this study, growing rabbits were randomly allocated to the daytime feeding (DF) group and nighttime restricted feeding (NRF) group. Our research demonstrated that, during winter, the DF regimen disrupted the behavioral rhythms of rabbits and accelerated weight gain without changing overall feed intake. The underlying reason was that DF disturbed the lipid metabolism rhythms, promoted hepatic lipid synthesis regulated by DGAT1 and lipid synthesis of adipose tissues regulated by GPAM, thus triggering fat deposition. In contrast, the NRF regimen enhanced thermogenesis regulated by T3 and elevated body temperature and facilitated ketogenesis mediated by HMGCS2, increasing energy consumption. However, it had no significant impact on the fat content within muscle. This study offers a theoretical foundation for the refinement of feeding management and healthy raising of rabbits.
Collapse
Affiliation(s)
- Ke-Hao Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (K.-H.Z.); (S.H.); (Q.-G.W.); (J.-J.L.); (C.-Y.Y.); (C.-H.S.); (L.Z.); (Z.-Y.L.); (P.L.)
| | - Shuai He
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (K.-H.Z.); (S.H.); (Q.-G.W.); (J.-J.L.); (C.-Y.Y.); (C.-H.S.); (L.Z.); (Z.-Y.L.); (P.L.)
| | - Quan-Gang Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (K.-H.Z.); (S.H.); (Q.-G.W.); (J.-J.L.); (C.-Y.Y.); (C.-H.S.); (L.Z.); (Z.-Y.L.); (P.L.)
| | - Jun-Jiao Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (K.-H.Z.); (S.H.); (Q.-G.W.); (J.-J.L.); (C.-Y.Y.); (C.-H.S.); (L.Z.); (Z.-Y.L.); (P.L.)
| | - Chun-Yan Yao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (K.-H.Z.); (S.H.); (Q.-G.W.); (J.-J.L.); (C.-Y.Y.); (C.-H.S.); (L.Z.); (Z.-Y.L.); (P.L.)
| | - Chun-Hua Shan
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (K.-H.Z.); (S.H.); (Q.-G.W.); (J.-J.L.); (C.-Y.Y.); (C.-H.S.); (L.Z.); (Z.-Y.L.); (P.L.)
| | - Lei Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (K.-H.Z.); (S.H.); (Q.-G.W.); (J.-J.L.); (C.-Y.Y.); (C.-H.S.); (L.Z.); (Z.-Y.L.); (P.L.)
| | - Zhong-Ying Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (K.-H.Z.); (S.H.); (Q.-G.W.); (J.-J.L.); (C.-Y.Y.); (C.-H.S.); (L.Z.); (Z.-Y.L.); (P.L.)
| | - Peng Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (K.-H.Z.); (S.H.); (Q.-G.W.); (J.-J.L.); (C.-Y.Y.); (C.-H.S.); (L.Z.); (Z.-Y.L.); (P.L.)
| | - Ming-Yong Li
- National Rabbit Industry Technology System Qingdao Comprehensive Experimental Station, Qingdao 266431, China;
| | - Yao Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (K.-H.Z.); (S.H.); (Q.-G.W.); (J.-J.L.); (C.-Y.Y.); (C.-H.S.); (L.Z.); (Z.-Y.L.); (P.L.)
| | - Zhong-Hong Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (K.-H.Z.); (S.H.); (Q.-G.W.); (J.-J.L.); (C.-Y.Y.); (C.-H.S.); (L.Z.); (Z.-Y.L.); (P.L.)
| |
Collapse
|
40
|
Nilghaz M, Sadeghi A, Koochakpoor G, Poustchi H, Khodadadi N, Narimani B, Ghods M, Shafiee M, Shahparvari MR, Hekmatdoost A. The efficacy of DASH combined with time-restricted feeding (16/8) on metabolic associated fatty liver disease management: a randomized controlled trial. Sci Rep 2025; 15:7020. [PMID: 40016311 PMCID: PMC11868424 DOI: 10.1038/s41598-025-88393-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 01/28/2025] [Indexed: 03/01/2025] Open
Abstract
Recent studies have utilized time-restricted feeding (16/8) (TRF) and dietary approaches to stop hypertension separately to manage metabolic-associated fatty liver disease (MAFLD); however, the effectiveness of combining these two approaches has not been investigated. The objective of this study was to examine the impact of TRF in conjunction with a DASH diet on various factors related to MAFLD. A 12-week randomized controlled trial was conducted to assess the impact of TRF (16/8), along with a DASH diet, compared with a control diet based on standard meal distribution, in patients with MAFLD. An investigation was conducted to examine alterations in anthropometric indices, as well as liver parameters, serum metabolic indices, and an inflammatory marker. The TRF plus DASH diet reduced body mass index (p = 0.03), abdominal circumference (p = 0.005), controlled attenuation parameter (CAP) (p < 0.001), alanine aminotransferase (p = 0.039), and aspartate aminotransferase (0.047) compared to the control group. The levels of insulin and homeostasis model assessment of insulin resistance reduced in both groups significantly (P < 0.05). In MAFLD patients, TRF (16/8) in combination with a DASH diet is superior to a low-calorie diet in promoting obesity indices, and hepatic steatosis and fibrosis. Further long-term investigations are needed to draw definitive conclusions.
Collapse
Affiliation(s)
- Maryam Nilghaz
- Department of Clinical Nutrition, School of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | - Hossein Poustchi
- Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Navideh Khodadadi
- Department of Clinical Nutrition, School of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnaz Narimani
- Department of Clinical Nutrition, School of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Ghods
- Department of Clinical Nutrition, School of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahshad Shafiee
- Department of Clinical Nutrition, School of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Shahparvari
- Department of Clinical Nutrition, School of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azita Hekmatdoost
- Department of Clinical Nutrition, School of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
41
|
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
|
42
|
Búr Z, Vendl B, Sűdy ÁR, Lumniczky Z, Szántó CG, Mócsai A, Káldi K, Ella K. Time-restricted feeding alleviates arthritis symptoms augmented by high-fat diet. Front Immunol 2025; 16:1512328. [PMID: 40018036 PMCID: PMC11864949 DOI: 10.3389/fimmu.2025.1512328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/10/2025] [Indexed: 03/01/2025] Open
Abstract
Rheumatoid arthritis (RA) affects approximately 1% of the global population. Its hallmark symptoms include severe pain and joint stiffness, which significantly diminish life quality. RA's development is influenced by multiple factors including unhealthy lifestyle habits. Calorie-rich diets, particularly those high in fat and resulting in obesity, are associated with RA and exacerbate its symptoms. Consequently, dietary modifications are recommended as a complementary treatment. However, adherence is often low due to the restrictive changes required in nutrient composition or caloric intake. Our previous findings indicate that time-restricted feeding (TRF) benefits leukocyte rhythm and mitigates autoimmune responses. In this study we explored the impact of TRF on the severity of K/BxN serum-transfer arthritis (STA) in mice subjected to high-fat diet. Three feeding schedules were implemented: a control (Ctrl) with constant access to standard chow, a high-fat diet group (HF) with ad libitum food access, and a high-fat TRF group (HF-TRF) with a 10-hour feeding window during the active phase. After four weeks of conditioning, STA was induced. Although macroscopic markers of inflammation did not differ between the Ctrl and HF groups, histological analysis revealed increased inflammation in HF mice, including expanded edema, pannus formation, bone erosion, elevated synovial neutrophil infiltration and serum leptin levels. Importantly, all these inflammatory markers were significantly reduced in the HF-TRF group, along with synovial IL-1β and monocyte/macrophage counts. Our results indicate that TRF can diminish the impact of a high-fat diet on STA severity, potentially serving as a preventive method and a sustainable therapeutic support for RA management.
Collapse
MESH Headings
- Animals
- Diet, High-Fat/adverse effects
- Mice
- Arthritis, Experimental/etiology
- Arthritis, Experimental/diet therapy
- Arthritis, Experimental/pathology
- Arthritis, Experimental/immunology
- Arthritis, Rheumatoid/etiology
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/diet therapy
- Arthritis, Rheumatoid/pathology
- Male
- Mice, Inbred C57BL
- Disease Models, Animal
- Fasting
Collapse
|
43
|
Paoli A. The Influence of Physical Exercise, Ketogenic Diet, and Time-Restricted Eating on De Novo Lipogenesis: A Narrative Review. Nutrients 2025; 17:663. [PMID: 40004991 PMCID: PMC11858292 DOI: 10.3390/nu17040663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
De novo lipogenesis (DNL) is a metabolic pathway that converts carbohydrates into fatty acids, primarily occurring in the liver and, to a lesser extent, in adipose tissue. While hepatic DNL is highly responsive to dietary carbohydrate intake and regulated by insulin via transcription factors like SREBP-1c, adipose DNL is more modest and less sensitive to dietary overfeeding. Dysregulated DNL contributes to metabolic disorders, including metabolic dysfunction-associated steatotic liver disease (MASLD). Lifestyle interventions, such as physical exercise, ketogenic diets, and time-restricted eating (TRE) offer promising strategies to regulate DNL and improve metabolic health. Physical exercise enhances glucose uptake in muscles, reduces insulin levels, and promotes lipid oxidation, thereby suppressing hepatic DNL. Endurance and resistance training also improve mitochondrial function, further mitigating hepatic triglyceride accumulation. Ketogenic diets shift energy metabolism toward fatty acid oxidation and ketogenesis, lower insulin, and directly downregulate lipogenic enzyme activity in the liver. TRE aligns feeding with circadian rhythms by optimizing AMP-activated protein kinase (AMPK) activation during fasting periods, which suppresses DNL and enhances lipid metabolism. The combined effects of these interventions demonstrate significant potential for improving lipid profiles, reducing hepatic triglycerides, and preventing lipotoxicity. By addressing the distinct roles of the liver and adipose DNL, these strategies target systemic and localized lipid metabolism dysregulation. Although further research is needed to fully understand their long-term impact, these findings highlight the transformative potential of integrating these approaches into clinical practice to manage metabolic disorders and their associated complications.
Collapse
Affiliation(s)
- Antonio Paoli
- Department of Biomedical Sciences, University of Padua, 35100 Padua, Italy;
- Research Center for High Performance Sport, UCAM Catholic University of Murcia, 30107 Murcia, Spain
| |
Collapse
|
44
|
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
|
45
|
Parrotta ME, Colangeli L, Scipione V, Vitale C, Sbraccia P, Guglielmi V. Time Restricted Eating: A Valuable Alternative to Calorie Restriction for Addressing Obesity? Curr Obes Rep 2025; 14:17. [PMID: 39899119 PMCID: PMC11790783 DOI: 10.1007/s13679-025-00609-z] [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: 01/20/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE OF REVIEW In this review, we summarize the molecular effects of time-restricted eating (TRE) and its possible role in appetite regulation. We also discuss the potential clinical benefits of TRE in obesity. RECENT FINDINGS TRE is an emerging dietary approach consisting in limiting food intake to a specific window of time each day. The rationale behind this strategy is to restore the circadian misalignment, commonly seen in obesity. Preclinical studies have shown that restricting food intake only during the active phase of the day can positively influence several cellular functions including senescence, mitochondrial activity, inflammation, autophagy and nutrients' sensing pathways. Furthermore, TRE may play a role by modulating appetite and satiety hormones, though further research is needed to clarify its exact mechanisms. Clinical trials involving patients with obesity or type 2 diabetes suggest that TRE can be effective for weight loss, but its broader effects on improving other clinical outcomes, such as cardiovascular risk factors, remain less certain. The epidemic proportions of obesity cause urgency to find dietary, pharmacological and surgical interventions that can be effective in the medium and long term. According to its molecular effects, TRE can be an interesting alternative to caloric restriction in the treatment of obesity, but the considerable variability across clinical trials regarding population, intervention, and follow-up duration makes it difficult to reach definitive conclusions.
Collapse
Affiliation(s)
| | - Luca Colangeli
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Valeria Scipione
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carolina Vitale
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Valeria Guglielmi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy.
| |
Collapse
|
46
|
Hou T, Su W, Chacon AN, Lin AH, Guo Z, Gong MC. Feeding- and Light-Cycle Synergistically Regulate Mouse Blood Pressure Daily Rhythm via Bmal1-Dependent and Independent Mechanisms. J Biol Rhythms 2025; 40:76-90. [PMID: 39772880 PMCID: PMC11835536 DOI: 10.1177/07487304241302510] [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
Cardiovascular health requires the orchestration of the daily rhythm of blood pressure (BP), which responds to changes in light exposure and dietary patterns. Whether rhythmic light and feeding can modulate daily BP rhythm directly or via modulating intrinsic core clock gene Bmal1 is unknown. Using inducible global Bmal1 knockout mice (iBmal1KO), we explored the impact of rhythmic light, rhythmic feeding, or their combination on various physiological parameters. Daily rhythms of BP, heart rate, and locomotor activity were monitored via radiotelemetry, while food intake patterns were tracked using the BioDAQ system. Respiratory exchange ratio (RER) and energy expenditure (EE) were assessed through indirect calorimetry. In addition, spectrum analysis was employed to analyze spontaneous baroreflex sensitivity and heart rate variability, and urinary norepinephrine excretion was quantified using high-performance liquid chromatography (HPLC). Neither rhythmic feeding nor rhythmic light alone was sufficient to reinstate the daily BP rhythm in arrhythmic iBmal1KO mice. However, combining the light and feeding cues in synchrony partially restored the daily BP rhythm. Interestingly, rhythmic feeding alone robustly reinstated RER and EE rhythms, even without rhythmic light. Similar to BP, the partial reinstatement of the daily rhythms in heart rate and locomotor activity was observed only when rhythmic light and feeding were applied in tandem. Rhythmic light by itself did not restore the light-dark phase difference in baroreflex sensitivity, urinary norepinephrine excretion, or the daily rhythm of heart rate variability. However, rhythmic feeding, alone or in combination with rhythmic light, successfully reinstated the light-dark phase differences in these parameters. In the absence of Bmal1, the synergy between rhythmic light and feeding can partially restore daily BP rhythm.
Collapse
Affiliation(s)
- Tianfei Hou
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Wen Su
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Aaron N. Chacon
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - An-Hsuan Lin
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Zhenheng Guo
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, United States
- Research and Development, Lexington Veterans Affairs Medical Center, Lexington, KY, United States
| | - Ming C. Gong
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
47
|
Yang M, Singh A, de Araujo A, McDougle M, Ellis H, Décarie-Spain L, Kanoski SE, de Lartigue G. Separate orexigenic hippocampal ensembles shape dietary choice by enhancing contextual memory and motivation. Nat Metab 2025; 7:276-296. [PMID: 39815079 PMCID: PMC11860247 DOI: 10.1038/s42255-024-01194-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/28/2024] [Indexed: 01/18/2025]
Abstract
The hippocampus (HPC) has emerged as a critical player in the control of food intake, beyond its well-known role in memory. While previous studies have primarily associated the HPC with food intake inhibition, recent research suggests a role in appetitive processes. Here we identified spatially distinct neuronal populations within the dorsal HPC (dHPC) that respond to either fats or sugars, potent natural reinforcers that contribute to obesity development. Using activity-dependent genetic capture of nutrient-responsive dHPC neurons, we demonstrate a causal role of both populations in promoting nutrient-specific intake through different mechanisms. Sugar-responsive neurons encoded spatial memory for sugar location, whereas fat-responsive neurons selectively enhanced the preference and motivation for fat intake. Importantly, stimulation of either nutrient-responsive dHPC neurons increased food intake, while ablation differentially impacted obesogenic diet consumption and prevented diet-induced weight gain. Collectively, these findings uncover previously unknown orexigenic circuits underlying macronutrient-specific consumption and provide a foundation for developing potential obesity treatments.
Collapse
Affiliation(s)
- Mingxin Yang
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arashdeep Singh
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alan de Araujo
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Molly McDougle
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hillary Ellis
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Léa Décarie-Spain
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Scott E Kanoski
- Human & Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Guillaume de Lartigue
- Monell Chemical Senses Center, Philadelphia, PA, USA.
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
48
|
Livelo C, Guo Y, Madhanagopal J, Morrow C, Melkani GC. Time-restricted feeding mediated modulation of microbiota leads to changes in muscle physiology in Drosophila obesity models. Aging Cell 2025; 24:e14382. [PMID: 39446089 PMCID: PMC11822661 DOI: 10.1111/acel.14382] [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: 05/16/2024] [Revised: 09/04/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024] Open
Abstract
Recent research has highlighted the essential role of the microbiome in maintaining skeletal muscle physiology. The microbiota influences muscle health by regulating lipid metabolism, protein synthesis, and insulin sensitivity. However, metabolic disturbances such as obesity can lead to dysbiosis, impairing muscle function. Time-restricted feeding (TRF) has been shown to mitigate obesity-related muscle dysfunction, but its effects on restoring healthy microbiomes remain poorly understood. This study utilizes 16S microbiome analysis and bacterial supplementation to investigate the bacterial communities influenced by TRF that may benefit skeletal muscle physiology. In wild-type and obese Drosophila models (axenic models devoid of natural microbial communities), the absence of microbiota influence muscle performance and metabolism differently. Specifically, axenic wild-type Drosophila exhibited reduced muscle performance, higher glucose levels, insulin resistance, ectopic lipid accumulation, and decreased ATP levels. Interestingly, in obese Drosophila (induced by a high-fat diet or predisposed obesity mutant Sk2), the absence of microbiota improved muscle performance, lowered glucose levels, reduced insulin resistance, and increased ATP levels. TRF was found to modulate microbiota composition, notably increasing Acetobacter pasteurianus (AP) and decreasing Staphylococcus aureus (SA) in both obesity models. Supplementation with AP improved muscle performance and reduced glucose and insulin resistance, while SA supplementation had the opposite effect. This study provides novel insights into the complex interactions between TRF, microbiota, and skeletal muscle physiology in different Drosophila models.
Collapse
Affiliation(s)
- Christopher Livelo
- Division of Molecular and Cellular Pathology, Department of Pathology, Heersink School of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Yiming Guo
- Division of Molecular and Cellular Pathology, Department of Pathology, Heersink School of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Jagathnarayan Madhanagopal
- Division of Molecular and Cellular Pathology, Department of Pathology, Heersink School of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Casey Morrow
- Department of Cell, Developmental, and Integrative BiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Girish C. Melkani
- Division of Molecular and Cellular Pathology, Department of Pathology, Heersink School of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- UAB Nathan Shock Center1300 University Boulevard BirminghamBirminghamAlabamaUSA
| |
Collapse
|
49
|
Buesing D, Fourman S, Ulrich-Lai YM. Time-limited access to palatable food reveals differential effects of psychological stress on homeostatic vs. hedonic feeding behavior in male rats. Appetite 2025; 206:107791. [PMID: 39608722 PMCID: PMC11789920 DOI: 10.1016/j.appet.2024.107791] [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: 06/28/2024] [Revised: 11/14/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
Psychological stress has complex effects on eating behavior, appearing to reduce homeostatically regulated feeding, while increasing hedonically motivated feeding. The present work tests this idea using two feeding paradigms that offer a highly palatable food on a time-limited basis, together with continual access to a low palatability food. This approach provides a natural separation between periods of eating that are primarily homeostatic vs. hedonically regulated. First, the impact of acute stress exposure on feeding behavior was tested using an acute "meal-dessert" paradigm. When fasted adult male rats were given a recent stressor of moderate intensity (restraint), refeeding with a chow-meal was reduced, without affecting chocolate-dessert intake, thereby increasing the proportion of calories derived from chocolate. Next, the effect of chronic moderate stress was tested using a "binge" eating paradigm. Chow-fed rats were given unexpected (3d per week) vs. expected (7d per week) brief access to a highly palatable high-fat diet (HFD), and feeding behavior was compared to control groups that were maintained with continuous access to only chow or only HFD. Chronic stress reduced total caloric intake in all groups, including binge-like HFD intake. Binge-like HFD intake caused metabolic dysfunction (increased adiposity and impaired glucose homeostasis) to an extent beyond that predicted by total caloric intake or body weight gain. Finally, binge-like HFD intake shifted stress coping behavior from an active to a passive phenotype, particularly in rats receiving concurrent chronic stress, suggesting the possibility of increased risk for stress-related disorders, like depression, in individuals who binge eat during stress.
Collapse
Affiliation(s)
- Dana Buesing
- Department of Pharmacology, Physiology, and Neurobiology, College of Medicine, University of Cincinnati, OH, USA
| | - Sarah Fourman
- Department of Pharmacology, Physiology, and Neurobiology, College of Medicine, University of Cincinnati, OH, USA
| | - Yvonne M Ulrich-Lai
- Department of Pharmacology, Physiology, and Neurobiology, College of Medicine, University of Cincinnati, OH, USA.
| |
Collapse
|
50
|
Godos J, Currenti W, Ferri R, Lanza G, Caraci F, Frias-Toral E, Guglielmetti M, Ferraris C, Lipari V, Carvajal Altamiranda S, Galvano F, Castellano S, Grosso G. Chronotype and Cancer: Emerging Relation Between Chrononutrition and Oncology from Human Studies. Nutrients 2025; 17:529. [PMID: 39940387 PMCID: PMC11819666 DOI: 10.3390/nu17030529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
Fasting-feeding timing is a crucial pattern implicated in the regulation of daily circadian rhythms. The interplay between sleep and meal timing underscores the importance of maintaining circadian alignment in order to avoid creating a metabolic environment conducive to carcinogenesis following the molecular and systemic disruption of metabolic performance and immune function. The chronicity of such a condition may support the initiation and progression of cancer through a variety of mechanisms, including increased oxidative stress, immune suppression, and the activation of proliferative signaling pathways. This review aims to summarize current evidence from human studies and provide an overview of the potential mechanisms underscoring the role of chrononutrition (including time-restricted eating) on cancer risk. Current evidence shows that the morning chronotype, suggesting an alignment between physiological circadian rhythms and eating timing, is associated with a lower risk of cancer. Also, early time-restricted eating and prolonged nighttime fasting were also associated with a lower risk of cancer. The current evidence suggests that the chronotype influences cancer risk through cell cycle regulation, the modulation of metabolic pathways and inflammation, and gut microbiota fluctuations. In conclusion, although there are no clear guidelines on this matter, emerging evidence supports the hypothesis that the role of time-related eating (i.e., time/calorie-restricted feeding and intermittent/periodic fasting) could potentially lead to a reduced risk of cancer.
Collapse
Affiliation(s)
- Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (J.G.)
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, 95123 Catania, Italy
| | - Walter Currenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (J.G.)
| | | | - Giuseppe Lanza
- Oasi Research Institute-IRCCS, 94018 Troina, Italy
- Department of Surgery and Medical-Surgical Specialties, University of Catania, 95125 Catania, Italy
| | - Filippo Caraci
- Oasi Research Institute-IRCCS, 94018 Troina, Italy
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
| | - Evelyn Frias-Toral
- School of Medicine, Universidad Espíritu Santo, Samborondón 0901952, Ecuador
| | - Monica Guglielmetti
- Human Nutrition and Eating Disorder Research Center, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
- Laboratory of Food Education and Sport Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
| | - Cinzia Ferraris
- Human Nutrition and Eating Disorder Research Center, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
- Laboratory of Food Education and Sport Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
| | - Vivian Lipari
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain
- Universidad de La Romana, La Romana 22000, Dominican Republic
- Universidad Internacional Iberoamericana, Campeche 24560, Mexico
| | - Stefanía Carvajal Altamiranda
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain
- Universidade Internacional do Cuanza, Cuito EN250, Angola
- Fundación Universitaria Internacional de Colombia, Bogotá 111321, Colombia
| | - Fabio Galvano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (J.G.)
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, 95123 Catania, Italy
| | - Sabrina Castellano
- Department of Educational Sciences, University of Catania, 95124 Catania, Italy
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (J.G.)
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, 95123 Catania, Italy
| |
Collapse
|