1
|
Ma C, Shen B, Chen L, Yang G. Impacts of circadian disruptions on behavioral rhythms in mice. FASEB J 2024; 38:e70183. [PMID: 39570004 DOI: 10.1096/fj.202401536r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/11/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024]
Abstract
Circadian rhythms are fundamental biological processes that recur approximately every 24 h, with the sleep-wake cycle or circadian behavior being a well-known example. In the field of chronobiology, mice serve as valuable model animals for studying mammalian circadian rhythms due to their genetic similarity to humans and the availability of various genetic tools for manipulation. Monitoring locomotor activity in mice provides valuable insights into the impact of various conditions or disturbances on circadian behavior. In this review, we summarized the effects of disturbance of biological rhythms on circadian behavior in mice. External factors, especially light exert a significant impact on circadian behavior. Additionally, feeding timing, food composition, ambient temperature, and physical exercise contribute to variations in the behavior of the mouse. Internal factors, including gender, age, genetic background, and clock gene mutation or deletion, are effective as well. Understanding the effects of circadian disturbances on murine behavior is essential for gaining insights into the underlying mechanisms of circadian regulation and developing potential therapeutic interventions for circadian-related disorders in humans.
Collapse
Affiliation(s)
- Changxiao Ma
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Bingyi Shen
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Lihong Chen
- Health Science Center, East China Normal University, Shanghai, China
| | - Guangrui Yang
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
2
|
Du NH, Sinturel F, Nowak N, Gosselin P, Saini C, Guessous I, Jornayvaz FR, Philippe J, Rey G, Dermitzakis ET, Zenobi R, Dibner C, Brown SA. Multi-omics correlates of insulin resistance and circadian parameters mapped directly from human serum. Eur J Neurosci 2024; 60:5487-5504. [PMID: 39205434 DOI: 10.1111/ejn.16486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/30/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024]
Abstract
While it is generally known that metabolic disorders and circadian dysfunction are intertwined, how the two systems affect each other is not well understood, nor are the genetic factors that might exacerbate this pathological interaction. Blood chemistry is profoundly changed in metabolic disorders, and we have previously shown that serum factors change cellular clock properties. To investigate if circulating factors altered in metabolic disorders have circadian modifying effects, and whether these effects are of genetic origin, we measured circadian rhythms in U2OS cell in the presence of serum collected from diabetic, obese or control subjects. We observed that circadian period lengthening in U2OS cells was associated with serum chemistry that is characteristic of insulin resistance. Characterizing the genetic variants that altered circadian period length by genome-wide association analysis, we found that one of the top variants mapped to the E3 ubiquitin ligase MARCH1 involved in insulin sensitivity. Confirming our data, the serum circadian modifying variants were also enriched in type 2 diabetes and chronotype variants identified in the UK Biobank cohort. Finally, to identify serum factors that might be involved in period lengthening, we performed detailed metabolomics and found that the circadian modifying variants are particularly associated with branched chain amino acids, whose levels are known to correlate with diabetes and insulin resistance. Overall, our multi-omics data showed comprehensively that systemic factors serve as a path through which metabolic disorders influence circadian system, and these can be examined in human populations directly by simple cellular assays in common cultured cells.
Collapse
Affiliation(s)
- Ngoc-Hien Du
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Flore Sinturel
- Department of Surgery, Division of Thoracic and Endocrine Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Nora Nowak
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Pauline Gosselin
- Department of Surgery, Division of Thoracic and Endocrine Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department and Division of Primary Care Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Camille Saini
- Department of Surgery, Division of Thoracic and Endocrine Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department and Division of Primary Care Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Idris Guessous
- Department and Division of Primary Care Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - François R Jornayvaz
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Medicine, Division of Endocrinology, Diabetes, Nutrition, and Therapeutic Education of Patient, University Hospitals of Geneva, Geneva, Switzerland
| | - Jacques Philippe
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Medicine, Division of Endocrinology, Diabetes, Nutrition, and Therapeutic Education of Patient, University Hospitals of Geneva, Geneva, Switzerland
| | - Guillaume Rey
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Emmanouil T Dermitzakis
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Renato Zenobi
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Charna Dibner
- Department of Surgery, Division of Thoracic and Endocrine Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Steven A Brown
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Park J, Nang JH, Cho S, Chung KJ, Kim KH. Chronic Mealtime Shift Disturbs Metabolic and Urinary Functions in Mice: Effects of Daily Antioxidant Supplementation. Int Neurourol J 2024; 28:115-126. [PMID: 38956771 PMCID: PMC11222825 DOI: 10.5213/inj.2448144.072] [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: 03/19/2024] [Accepted: 05/20/2024] [Indexed: 07/04/2024] Open
Abstract
PURPOSE Through their biological clocks, organisms on this rotating planet can coordinate physiological processes according to the time of the day. However, the prevalence of circadian rhythm disorders has increased in modern society with the growing number of shift workers, elevating the risk of various diseases. In this study, we employed a mouse model to investigate the effects of urinary rhythm disturbances resulting from dietary changes commonly experienced by night shift workers. METHODS We established 3 groups based on feeding time and the use of restricted feeding: ad libitum, daytime, and early nighttime feeding. We then examined the urinary rhythm in each group. In addition to the bladder rhythm, we investigated changes in mRNA patterns within the tissues constituting the bladder. Additionally, we assessed the urination rhythm in Per1 and Per2 double-knockout mice and evaluated whether the injection of antioxidants modified the impact of mealtime shift on urination rhythm in wild-type mice. RESULTS Our study revealed that a shift in mealtime significantly impacted the circadian patterns of water intake and urinary excretion. In Per2::Luc knock-in mouse bladders cultured ex vivo, this shift increased the amplitude of Per2 oscillation and delayed its acrophases by several hours. Daily supplementation with antioxidants did not influence the mealtime shift-induced changes in circadian patterns of water intake and urinary excretion, nor did it affect the modified Per2 oscillation patterns in the cultured bladder. However, in aged mice, antioxidants partially restored the urinary rhythm. CONCLUSION A shift in mealtime meaningfully impacted the urination rhythm in mice, regardless of the presence of circadian clock genes.
Collapse
Affiliation(s)
- Jihyun Park
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, Korea
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Korea
| | - Jun-Ho Nang
- Department of Medicine, Graduate School, Kyung Hee University, Seoul, Korea
| | - Sehyung Cho
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, Korea
- Department of Physiology, Kyung Hee University School of Medicine, Seoul, Korea
| | - Kyung Jin Chung
- Department of Urology, Gachon University Gil Medical Center, Gachon Univesity School of Medicine, Incheon, Korea
| | - Khae Hawn Kim
- Department of Urology, Chungnam National University Sejong Hospital, Chungnam National University College of Medicine, Sejong, Korea
| |
Collapse
|
4
|
Caria I, Nunes MJ, Ciraci V, Carvalho AN, Ranito C, Santos SG, Gama MJ, Castro-Caldas M, Rodrigues CMP, Ruas JL, Rodrigues E. NPC1-like phenotype, with intracellular cholesterol accumulation and altered mTORC1 signaling in models of Parkinson's disease. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166980. [PMID: 38061599 DOI: 10.1016/j.bbadis.2023.166980] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/13/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023]
Abstract
Disruption of brain cholesterol homeostasis has been implicated in neurodegeneration. Nevertheless, the role of cholesterol in Parkinson's Disease (PD) remains unclear. We have used N2a mouse neuroblastoma cells and primary cultures of mouse neurons and 1-methyl-4-phenylpyridinium (MPP+), a known mitochondrial complex I inhibitor and the toxic metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), known to trigger a cascade of events associated with PD neuropathological features. Simultaneously, we utilized other mitochondrial toxins, including antimycin A, oligomycin, and carbonyl cyanide chlorophenylhydrazone. MPP+ treatment resulted in elevated levels of total cholesterol and in a Niemann Pick type C1 (NPC1)-like phenotype characterized by accumulation of cholesterol in lysosomes. Interestingly, NPC1 mRNA levels were specifically reduced by MPP+. The decrease in NPC1 levels was also seen in midbrain and striatum from MPTP-treated mice and in primary cultures of neurons treated with MPP+. Together with the MPP+-dependent increase in intracellular cholesterol levels in N2a cells, we observed an increase in 5' adenosine monophosphate-activated protein kinase (AMPK) phosphorylation and a concomitant increase in the phosphorylated levels of mammalian target of rapamycin (mTOR). NPC1 knockout delayed cell death induced by acute mitochondrial damage, suggesting that transient cholesterol accumulation in lysosomes could be a protective mechanism against MPTP/MPP+ insult. Interestingly, we observed a negative correlation between NPC1 protein levels and disease stage, in human PD brain samples. In summary, MPP+ decreases NPC1 levels, elevates lysosomal cholesterol accumulation and alters mTOR signaling, adding to the existing notion that PD may rise from alterations in mitochondrial-lysosomal communication.
Collapse
Affiliation(s)
- Inês Caria
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Maria João Nunes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Viviana Ciraci
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Andreia Neves Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Catarina Ranito
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Susana G Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Maria João Gama
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Margarida Castro-Caldas
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal; UCIBIO, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Jorge L Ruas
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Portugal; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Elsa Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Portugal.
| |
Collapse
|
5
|
Alsayid M, Khan MO, Adnan D, Rasmussen HE, Keshavarzian A, Bishehsari F. Behavioral circadian phenotypes are associated with the risk of elevated body mass index. Eat Weight Disord 2022; 27:1395-1403. [PMID: 34355307 PMCID: PMC8816962 DOI: 10.1007/s40519-021-01276-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Metabolic dysfunction and obesity rates are on the rise. Although the central modes of circadian disruption has been studied in relation to the risk of obesity, the role of eating time has remained unclear. Here, we aimed to assess circadian behavioral phenotypes and their association with the risk of elevated body mass index (BMI). METHODS This was a prospective cross-sectional study of individuals presenting for colorectal cancer screening colonoscopy. Participants completed demographic questionnaires, The Munich ChronoType Questionnaire (MCTQ), and Food Timing Screener (FTS). The primary outcome of the study was the association between circadian phenotypes and elevated BMI. RESULTS A total of 488 individuals completed the survey, with a mean (SD) age of 57.5 (10.8) years. The mean body mass index (BMI) was 28.8 (6.1) kg/m2, with 72.3% of individuals met criteria for elevated BMI. Four circadian behavioral phenotypes were generated: early chronotype with regular food timing (ER) (34.7%), early chronotype with irregular food timing (EI) (11.7%), intermediate/late chronotype with regular food timing (LR) (33.9%), and intermediate/late chronotype with irregular food timing (LI) (19.7%). In a multivariable regression analysis, LI phenotype had 2.9 times higher odds of elevated BMI as compared to ER phenotype (OR 2.9, 95% CI 1.3-6.7, P = 0.01). CONCLUSION The combination of late chronotype and irregular food timing, representative of a behavioral circadian rhythm disruption, is associated with higher rates of elevated BMI. The majority of individuals with this abnormal circadian phenotype were younger than 60 years old. This observation is especially relevant because of the ongoing rise in the obesity rates among young adults. LEVEL III Evidence obtained from well-designed cohort or case-control analytic studies.
Collapse
Affiliation(s)
- Muhammad Alsayid
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, USA
| | - Mohammed Omer Khan
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, USA
| | - Darbaz Adnan
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, USA
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Professional Building, 1725 W. Harrison St. Suite 207, Chicago, IL, 60612, USA
| | - Heather E Rasmussen
- Department of Nutrition and Dietetics, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Ali Keshavarzian
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, USA
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Professional Building, 1725 W. Harrison St. Suite 207, Chicago, IL, 60612, USA
| | - Faraz Bishehsari
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, USA.
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Professional Building, 1725 W. Harrison St. Suite 207, Chicago, IL, 60612, USA.
| |
Collapse
|
6
|
Lacticaseibacillus rhamnosus: A Suitable Candidate for the Construction of Novel Bioengineered Probiotic Strains for Targeted Pathogen Control. Foods 2022; 11:foods11060785. [PMID: 35327208 PMCID: PMC8947445 DOI: 10.3390/foods11060785] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Probiotics, with their associated beneficial effects, have gained popularity for the control of foodborne pathogens. Various sources are explored with the intent to isolate novel robust probiotic strains with a broad range of health benefits due to, among other mechanisms, the production of an array of antimicrobial compounds. One of the shortcomings of these wild-type probiotics is their non-specificity. A pursuit to circumvent this limitation led to the advent of the field of pathobiotechnology. In this discipline, specific pathogen gene(s) are cloned and expressed into a given probiotic to yield a novel pathogen-specific strain. The resultant recombinant probiotic strain will exhibit enhanced species-specific inhibition of the pathogen and its associated infection. Such probiotics are also used as vehicles to deliver therapeutic agents. As fascinating as this approach is, coupled with the availability of numerous probiotics, it brings a challenge with regard to deciding which of the probiotics to use. Nonetheless, it is indisputable that an ideal candidate must fulfil the probiotic selection criteria. This review aims to show how Lacticaseibacillus rhamnosus, a clinically best-studied probiotic, presents as such a candidate. The objective is to spark researchers’ interest to conduct further probiotic-engineering studies using L. rhamnosus, with prospects for the successful development of novel probiotic strains with enhanced beneficial attributes.
Collapse
|
7
|
Park J, Kim J, Yun Y, Han D, Kim K, Hong J, Cho S. Daily injection of melatonin inhibits insulin resistance induced by chronic mealtime shift. Physiol Rep 2022; 10:e15227. [PMID: 35343087 PMCID: PMC8958345 DOI: 10.14814/phy2.15227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 06/14/2023] Open
Abstract
Shift work disorders have become an emerging concern worldwide. Shift disorders encompass a wide range of illnesses that have yet to be identified. The study focused on the relationship between shift work disorders and insulin resistance. Previously, it was reported that advancing the usual mealtime of mice triggered insulin resistance. Here, the hypothesis that chronic mealtime shifts induce oxidative damage leading to chronic diseases such as type 2 diabetes was tested. It was found that mealtime shift causes imbalances between anti-oxidative capacity and reactive oxygen species (ROS) levels, indicating increased oxidative damage during the light/rest phase. This study further demonstrated that daily supplementation of antioxidants at the appropriate time of day inhibited insulin resistance caused by chronic mealtime shifts, suggesting significant and chronic health implications for shift workers. In conclusion, it was confirmed that increased ROS levels caused by mealtime shift induce insulin resistance, which is inhibited by the antioxidant melatonin.
Collapse
Affiliation(s)
- Jihyun Park
- Department of NeuroscienceGraduate SchoolKyung Hee UniversitySeoulSouth Korea
| | - Jichul Kim
- Department of Life & Nanopharmaceutical ScienceGraduate SchoolKyung Hee UniversitySeoulSouth Korea
| | - Yejin Yun
- Department of Biomedical ScienceGraduate SchoolKyung Hee UniversitySeoulSouth Korea
| | - Dong‐Hee Han
- Department of NeuroscienceGraduate SchoolKyung Hee UniversitySeoulSouth Korea
- Present address:
Ildong Pharmaceutical Co. LtdSeocho‐guSeoulRepublic of Korea
| | - Kyungjin Kim
- Department of Brain ScienceDGISTDaeguSouth Korea
| | - Jongki Hong
- College of PharmacyKyung Hee UniversitySeoulSouth Korea
| | - Sehyung Cho
- Department of NeuroscienceGraduate SchoolKyung Hee UniversitySeoulSouth Korea
- Department of PhysiologyKyung Hee University School of MedicineSeoulSouth Korea
| |
Collapse
|
8
|
Chakradeo P, Rasmussen HE, Swanson GR, Swanson B, Fogg LF, Bishehsari F, Burgess HJ, Keshavarzian A. Psychometric Testing of a Food Timing Questionnaire and Food Timing Screener. Curr Dev Nutr 2022; 6:nzab148. [PMID: 35198845 PMCID: PMC8856943 DOI: 10.1093/cdn/nzab148] [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: 08/24/2021] [Revised: 11/29/2021] [Accepted: 12/08/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Circadian rhythms coordinate multiple biological processes, and time of eating is an important entrainer of peripheral circadian clocks, including those in the gastrointestinal tract and liver. Whereas time of eating can be assessed through valid and reliable tools designed to measure nutrient intake (24-h recalls), currently there is no easily administered, valid, and reliable tool designed to specifically assess both time of food intake and sleep. OBJECTIVES The objective of this study was to test the validity and reliability of 2 questionnaires developed to measure food and sleep-wake timing, the Food Timing Questionnaire (FTQ) and Food Timing Screener (FTS), and the agreement between these 2 tools. METHODS The content validity of these tools was assessed by an expert panel of 10 registered dietitian nutritionists. Adult volunteers (n = 61) completed both tools to assess internal consistency and test-retest reliability. Criterion-related validity was determined through the association of FTQ and FTS with 2 valid instruments, the Automated Self-Administered 24-hour recall (ASA24®) Dietary Assessment tool and the Munich Chronotype Questionnaire. Agreement between the FTQ and FTS was tested by calculating the Pearson's correlations for both food and sleep-wake timing. RESULTS The content validity indexes for both tools were >0.80, and internal consistency and test-retest reliability coefficients were >0.50 for all meals and sleep-wake times. Correlation coefficients were >0.40 between both tools and criterion measures of food intake and sleep except for snacks. Correlations between the FTQ and FTS for all eating events and sleep were >0.60 except for snack 1. CONCLUSIONS Both the FTQ and FTS are valid and reliable instruments for meal timing and sleep. However, further psychometric testing in a more expansive and diverse sample will improve the ability of these tools to accurately assess food timing and sleep and their impact on health outcomes.
Collapse
Affiliation(s)
- Prachi Chakradeo
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Heather E Rasmussen
- Department of Clinical Nutrition, Rush University Medical Center, Chicago, IL, USA
| | - Garth R Swanson
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Barbara Swanson
- College of Nursing, Rush University Medical Center, Chicago, IL, USA
| | - Louis F Fogg
- College of Nursing, Rush University Medical Center, Chicago, IL, USA
| | - Faraz Bishehsari
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Helen J Burgess
- Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Ali Keshavarzian
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
9
|
Zhang Y, Song M, Yuan C, Chan AT, Schernhammer ES, Wolpin BM, Stampfer MJ, Meyerhardt JA, Fuchs CS, Roberts SB, Rimm EB, Willett WC, Hu FB, Giovannucci EL, Ng K. Unrestrained eating behavior and risk of mortality: A prospective cohort study. Clin Nutr 2021; 40:5419-5429. [PMID: 34653818 PMCID: PMC8571025 DOI: 10.1016/j.clnu.2021.09.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/30/2021] [Accepted: 09/10/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Unrestrained eating behavior has been thought to be a proxy for diet frequency, timing, and caloric intake. We investigated the association of unrestrained eating with mortality risk in the Nurses' Health Study prospectively. METHODS During follow-up (1994-2016), 21,953 deaths were documented among 63,999 eligible participants in analyses of eating anything at any time, 22,120 deaths were documented among 65,839 participants in analyses of no concern with figure change. Hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated using Cox proportional hazards models. RESULTS Eating anything at any time was associated with an increased mortality from cancer (overall HR, 95%CI: 1.07, 1.00-1.13; driven by gastrointestinal tract cancer: 1.30, 1.10-1.54) and respiratory disease (1.16, 1.05-1.29), and decreased cardiovascular disease-specific mortality (0.92, 0.86-0.99), compared to those without this behavior; however, no association was observed between this behavior and all-cause mortality (1.02, 0.99-1.05). Women who reported having no concern with figure change experienced higher risk of mortality from all-cause (1.08, 1.05-1.11), cancer (1.08, 1.02-1.14), and respiratory disease (1.18, 1.08-1.30), compared to those not reporting this behavior. Their combined effect was associated with a higher all-cause (1.09, 1.04-1.14), cancer-specific (overall: 1.18, 1.09-1.28; gastrointestinal tract cancer: 1.36, 1.08-1.71; lung cancer: 1.09; 1.04-1.14), and respiratory disease-specific (1.30, 1.13-1.50) mortality, and was inversely associated with cardiovascular disease-specific mortality (0.88, 0.80-0.98), compared to those exhibiting the opposite. CONCLUSIONS Unrestrained eating was associated with increased risk of all-cause, cancer-specific (particularly for gastrointestinal tract cancer and lung cancer), and respiratory disease-specific mortality, and decreased risk of cardiovascular disease-specific mortality.
Collapse
Affiliation(s)
- Yin Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| | - Mingyang Song
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Andrew T Chan
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eva S Schernhammer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Center for Public Health, Medical University of Vienna, Vienna, Austria
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Meir J Stampfer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Charles S Fuchs
- Department of Medical Oncology, Smilow Cancer Hospital and Yale Cancer Center, New Haven, CT, USA; Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Susan B Roberts
- USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Eric B Rimm
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Walter C Willett
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Frank B Hu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Edward L Giovannucci
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Zhang Y, Song M, Chan AT, Schernhammer ES, Wolpin BM, Stampfer MJ, Meyerhardt JA, Fuchs CS, Roberts SB, Willett WC, Hu FB, Giovannucci EL, Ng K. Unrestrained eating behavior and risk of digestive system cancers: a prospective cohort study. Am J Clin Nutr 2021; 114:1612-1624. [PMID: 34293086 PMCID: PMC8588850 DOI: 10.1093/ajcn/nqab235] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 06/18/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Unrestrained eating behavior, as a potential proxy for diet frequency, timing, and caloric intake, has been questioned as a plausible risk factor for digestive system cancers, but epidemiological evidence remains sparse. OBJECTIVES We investigated prospectively the associations between unrestrained eating behavior and digestive system cancer risk. METHODS Participants in the Nurses' Health Study who were free of cancer and reported dietary information in 1994 were followed for ≤18 y. Cox models were used to estimate HRs and 95% CIs for unrestrained eating (eating anything at any time, no concern with figure change, or both) and risk of digestive system cancers. RESULTS During follow-up, 2064 digestive system cancer cases were documented among 70,450 eligible participants in analyses of eating anything at any time, In total, 2081 digestive system cancer cases were documented among 72,468 eligible participants in analyses of no concern with figure change. In fully adjusted analyses, women with the behavior of eating anything at any time had a higher risk of overall digestive system cancer (HR: 1.22; 95% CI: 1.10, 1.35), overall gastrointestinal tract cancer ((HR: 1.33; 95% CI: 1.18, 1.50), buccal cavity and pharynx cancer (HR: 1.50; 95% CI: 1.02, 2.21), esophageal cancer (HR: 1.62; 95% CI: 1.01, 2.62), small intestine cancer (HR: 1.92; 95% CI: 1.02,3. 59), and colorectal cancer (HR: 1.20; 95% CI: 1.04, 1.38), and a non-statistically significant increased risk of stomach cancer (HR: 1.54; 95% CI: 0.96,2.48), compared with women without this behavior. No statistically significant association was observed for pancreatic cancer and liver and gallbladder cancer. The combined effect of eating anything at any time and having no concern with figure change was associated with a significantly increased risk of overall digestive system cancer (HR: 1.27; 95% CI: 1.10, 1.46), overall gastrointestinal tract cancer (HR: 1.45; 95% CI: 1.23, 1.71), and colorectal cancer (HR: 1.34; 95% CI: 1.11, 1.63), compared with women exhibiting the opposite. CONCLUSIONS Unrestrained eating behavior was independently associated with increased risk of gastrointestinal tract cancers. The potential importance of unrestrained eating behavior modification in preventing gastrointestinal tract cancers should be noted.
Collapse
Affiliation(s)
- Yin Zhang
- Address correspondence to YZ (emails: and )
| | - Mingyang Song
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Andrew T Chan
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eva S Schernhammer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Center of Public Health, Medical University of Vienna, Vienna, Austria
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Meir J Stampfer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Charles S Fuchs
- Department of Medical Oncology, Smilow Cancer Hospital and Yale Cancer Center, New Haven, CT, USA
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Susan B Roberts
- USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Walter C Willett
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Frank B Hu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Edward L Giovannucci
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Charlot A, Hutt F, Sabatier E, Zoll J. Beneficial Effects of Early Time-Restricted Feeding on Metabolic Diseases: Importance of Aligning Food Habits with the Circadian Clock. Nutrients 2021; 13:nu13051405. [PMID: 33921979 PMCID: PMC8143522 DOI: 10.3390/nu13051405] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/17/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
The importance of metabolic health is a major societal concern due to the increasing prevalence of metabolic diseases such as obesity, diabetes, and various cardiovascular diseases. The circadian clock is clearly implicated in the development of these metabolic diseases. Indeed, it regulates physiological processes by hormone modulation, thus helping the body to perform them at the ideal time of day. Since the industrial revolution, the actions and rhythms of everyday life have been modified and are characterized by changes in sleep pattern, work schedules, and eating habits. These modifications have in turn lead to night shift, social jetlag, late-night eating, and meal skipping, a group of customs that causes circadian rhythm disruption and leads to an increase in metabolic risks. Intermittent fasting, especially the time-restricted eating, proposes a solution: restraining the feeding window from 6 to 10 h per day to match it with the circadian clock. This approach seems to improve metabolic health markers and could be a therapeutic solution to fight against metabolic diseases. This review summarizes the importance of matching life habits with circadian rhythms for metabolic health and assesses the advantages and limits of the application of time-restricted fasting with the objective of treating and preventing metabolic diseases.
Collapse
|
12
|
Bishehsari F, Moossavi S, Engen PA, Liu X, Zhang Y. Abnormal Food Timing Promotes Alcohol-Associated Dysbiosis and Colon Carcinogenesis Pathways. Front Oncol 2020; 10:1029. [PMID: 32850307 PMCID: PMC7396506 DOI: 10.3389/fonc.2020.01029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 05/26/2020] [Indexed: 12/21/2022] Open
Abstract
Background: Alcohol consumption is an established risk factor for colorectal cancer (CRC). Identifying cofactor(s) that modulate the effect of alcohol on colon inflammation and carcinogenesis could help risk stratification for CRC. Disruption of circadian rhythm by light/dark shift promotes alcohol-induced colonic inflammation and cancer. More recently, we found that abnormal food timing causes circadian rhythm disruption and promotes alcohol associated colon carcinogenesis. In this study, we examined the interaction of wrong-time feeding (WTF) and alcohol on CRC-related pathways, in relation to changes in microbial community structure. Methods: Polyposis mice (TS4Cre ×cAPC Δ468) underwent four conditions: alcohol or water and feeding during the light (wrong-time fed/WTF) or during the dark (right-time fed). Colonic cecum mucosal gene expression was analyzed by RNA-seq. Microbiota 16S ribosomal RNA sequencing analysis was used to examine colonic feces. Modeling was used to estimate the extent of the gene expression changes that could be related to the changes in the colonic microbial composition. Results: The circadian rhythm pathway was the most altered pathway by the WTF treatment, indicating that WTF is disruptive to the colonic circadian rhythm. Pathway analysis revealed interaction of WTF with alcohol in dysregulating pathways related to colon carcinogenesis. Similarly, the interaction of alcohol and WTF was detected at multiple parameters of the colonic microbiota including α and β diversity, as well as the community structure. Our modeling revealed that almost a third of total gene alterations induced by our treatments could be related to alterations in the abundance of the microbial taxa. Conclusion: These data support the promoting effect of abnormal food timing alcohol-associated CRC-related pathways in the colon and suggest colon dysbiosis as a targetable mechanism.
Collapse
Affiliation(s)
- Faraz Bishehsari
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL, United States
| | - Shirin Moossavi
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Phillip A. Engen
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL, United States
| | - Xiaohan Liu
- Department of Mathematics, Statistics, and Computer Science, University of Illinois at Chicago, Chicago, IL, United States
| | - Yue Zhang
- Department of Mathematics, Statistics, and Computer Science, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
13
|
Ruddick-Collins LC, Morgan PJ, Johnstone AM. Mealtime: A circadian disruptor and determinant of energy balance? J Neuroendocrinol 2020; 32:e12886. [PMID: 32662577 DOI: 10.1111/jne.12886] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/24/2020] [Accepted: 06/14/2020] [Indexed: 12/21/2022]
Abstract
Circadian rhythms play a critical role in the physiological processes involved in energy metabolism and energy balance (EB). A large array of metabolic processes, including the expression of many energy-regulating endocrine hormones, display temporal rhythms that are driven by both the circadian clock and food intake. Mealtime has been shown to be a compelling zeitgeber in peripheral tissue rhythms. Inconsistent signalling to the periphery, because of mismatched input from the central clock vs time of eating, results in circadian disruption in which central and/or peripheral rhythms are asynchronously time shifted or their amplitudes reduced. A growing body of evidence supports the negative health effects of circadian disruption, with strong evidence in murine models that mealtime-induced circadian disruption results in various metabolic consequences, including energy imbalance and weight gain. Increased weight gain has been reported to occur even without differences in energy intake, indicating an effect of circadian disruption on energy expenditure. However, the translation of these findings to humans is not well established because the ability to undertake rigorously controlled dietary studies that explore the chronic effects on energy regulation is challenging. Establishing the neuroendocrine changes in response to both acute and chronic variations in mealtime, along with observations in populations with routinely abnormal mealtimes, may provide greater insight into underlying mechanisms that influence long-term weight management under different meal patterns. Human studies should explore mechanisms through relevant biomarkers; for example, cortisol, leptin, ghrelin and other energy-regulating neuroendocrine factors. Mistiming between aggregate hormonal signals, or between hormones with their receptors, may cause reduced signalling intensity and hormonal resistance. Understanding how mealtimes may impact on the coordination of endocrine factors is essential for untangling the complex regulation of EB. Here a review is provided on current evidence of the impacts of mealtime on energy metabolism and the underlying neuroendocrine mechanisms, with a specific focus on human research.
Collapse
Affiliation(s)
| | - Peter J Morgan
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
| | | |
Collapse
|
14
|
Androulakis IP. The quest for digital health: From diseases to patients. Comput Chem Eng 2019. [DOI: 10.1016/j.compchemeng.2019.05.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
15
|
Time-restricted feeding improves adaptation to chronically alternating light-dark cycles. Sci Rep 2019; 9:7874. [PMID: 31133707 PMCID: PMC6536683 DOI: 10.1038/s41598-019-44398-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 05/02/2019] [Indexed: 12/19/2022] Open
Abstract
Disturbance of the circadian clock has been associated with increased risk of cardio-metabolic disorders. Previous studies showed that optimal timing of food intake can improve metabolic health. We hypothesized that time-restricted feeding could be a strategy to minimize long term adverse metabolic health effects of shift work and jetlag. In this study, we exposed female FVB mice to weekly alternating light-dark cycles (i.e. 12 h shifts) combined with ad libitum feeding, dark phase feeding or feeding at a fixed clock time, in the original dark phase. In contrast to our expectations, long-term disturbance of the circadian clock had only modest effects on metabolic parameters. Mice fed at a fixed time showed a delayed adaptation compared to ad libitum fed animals, in terms of the similarity in 24 h rhythm of core body temperature, in weeks when food was only available in the light phase. This was accompanied by increased plasma triglyceride levels and decreased energy expenditure, indicating a less favorable metabolic state. On the other hand, dark phase feeding accelerated adaptation of core body temperature and activity rhythms, however, did not improve the metabolic state of animals compared to ad libitum feeding. Taken together, restricting food intake to the active dark phase enhanced adaptation to shifts in the light-dark schedule, without significantly affecting metabolic parameters.
Collapse
|
16
|
Hernández-García J, Navas-Carrillo D, Orenes-Piñero E. Alterations of circadian rhythms and their impact on obesity, metabolic syndrome and cardiovascular diseases. Crit Rev Food Sci Nutr 2019; 60:1038-1047. [PMID: 30633544 DOI: 10.1080/10408398.2018.1556579] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Circadian system is comprised by central circadian pacemaker and several peripheral clocks that receive information from the external environment, synchronizing the circadian clocks. It is widely known that physiology is rhythmic and that the rupture of this rhythmicity can generate serious consequences. Circadian clocks, led by suprachiasmatic nucleus (SCN) in the central nervous system, are the responsible for generating this biological rhythmicity. These clocks are affected by external signals such as light (changes between day and night) and feeding rhythms. In this review, the basic principles of the circadian system and current knowledge of biological clocks are addressed, analyzing the relationship between circadian system, food intake, nutrition, and associated metabolic processes. In addition, the consequences occurring when these systems are not well coordinated with each other, such as the development of cardiovascular and metabolic pathologies, will be thoroughly discussed.
Collapse
Affiliation(s)
| | - Diana Navas-Carrillo
- Department of Surgery, Hospital de la Vega Lorenzo Guirao, University of Murcia, Murcia, Spain
| | - Esteban Orenes-Piñero
- Department of Biochemistry and Molecular Biology-A, University of Murcia, Murcia, Spain
| |
Collapse
|
17
|
Grant AD, Wilsterman K, Smarr BL, Kriegsfeld LJ. Evidence for a Coupled Oscillator Model of Endocrine Ultradian Rhythms. J Biol Rhythms 2018; 33:475-496. [PMID: 30132387 DOI: 10.1177/0748730418791423] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Whereas long-period temporal structures in endocrine dynamics have been well studied, endocrine rhythms on the scale of hours are relatively unexplored. The study of these ultradian rhythms (URs) has remained nascent, in part, because a theoretical framework unifying ultradian patterns across systems has not been established. The present overview proposes a conceptual coupled oscillator network model of URs in which oscillating hormonal outputs, or nodes, are connected by edges representing the strength of node-node coupling. We propose that variable-strength coupling exists both within and across classic hormonal axes. Because coupled oscillators synchronize, such a model implies that changes across hormonal systems could be inferred by surveying accessible nodes in the network. This implication would at once simplify the study of URs and open new avenues of exploration into conditions affecting coupling. In support of this proposed framework, we review mammalian evidence for (1) URs of the gut-brain axis and the hypothalamo-pituitary-thyroid, -adrenal, and -gonadal axes, (2) UR coupling within and across these axes; and (3) the relation of these URs to body temperature. URs across these systems exhibit behavior broadly consistent with a coupled oscillator network, maintaining both consistent URs and coupling within and across axes. This model may aid the exploration of mammalian physiology at high temporal resolution and improve the understanding of endocrine system dynamics within individuals.
Collapse
Affiliation(s)
- Azure D Grant
- The Helen Wills Neuroscience Institute, University of California, Berkeley, California
| | - Kathryn Wilsterman
- Department of Integrative Biology, University of California, Berkeley, California
| | - Benjamin L Smarr
- Department of Psychology, University of California, Berkeley, California
| | - Lance J Kriegsfeld
- The Helen Wills Neuroscience Institute, University of California, Berkeley, California.,Department of Psychology, University of California, Berkeley, California
| |
Collapse
|
18
|
Chakradeo PS, Keshavarzian A, Singh S, Dera AE, Esteban JPG, Lee AA, Burgess HJ, Fogg L, Swanson GR. Chronotype, social jet lag, sleep debt and food timing in inflammatory bowel disease. Sleep Med 2018; 52:188-195. [PMID: 30243610 DOI: 10.1016/j.sleep.2018.08.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/29/2022]
Abstract
The preference of the sleep/wake cycle can be grouped into categories or chronotypes. Inflammatory bowel disease (IBD) has been linked to poor sleep quality which correlates with disease severity. Social jet lag (SJL) is the difference between sleep timing on work and free days and is a marker for circadian misalignment which has been linked to increased inflammation. We investigated whether chronotype, SJL, sleep debt (SD), and food timing were associated with an IBD specific complications and a lower quality of life. Overall, 191 subjects (115 IBD subjects and 76 healthy controls (HC)) completed the Pittsburgh Sleep Quality Index (PSQI), Morningness-Eveningness Questionnaire (MEQ), Munich ChronoType Questionnaire (MCTQ), Short Inflammatory Bowel Disease Questionnaire (SIBDQ), and a structured Food Timing Questionnaire. Later chronotype (by MEQ) was associated with a worse SIBDQ (r = -0.209; P < 0.05). SJL was increased in IBD at 1.32 h ± 1.03 vs. 1.05 h ± 0.97 in HC, P < 0.05, when adjusted for age. SJL (>2 h) was present in 40% of severe/complicated Crohn's patients (fistulizing or structuring Crohn's or history of Crohn's related surgery) compared to only 16% of uncomplicated Crohn's patients (P < 0.05). Sleep debt was increased in IBD subjects compared to HC at 21.90 m ± 25.37 vs. 11.49 m ± 13.58, P < 0.05. IBD subjects with inconsistent breakfast or dinner times had lower SIBDQ scores (4.78 ± 1.28 vs. 5.49 ± 1.02, P < 0.05; 4.95 ± 0.31 vs. 5.42 ± 0.32, P < 0.05 respectively). In summary, later chronotype, and markers of circadian misalignment (social jet lag, sleep debt, and inconsistent meal timing) were associated with IBD disease specific complications and/or lower quality of life.
Collapse
Affiliation(s)
- Prachi S Chakradeo
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Rush University Medical Center, 1725 W. Harrison, Suite 206, Chicago, IL, 60612, United States.
| | - Ali Keshavarzian
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Rush University Medical Center, 1725 W. Harrison, Suite 206, Chicago, IL, 60612, United States.
| | - Shubha Singh
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Rush University Medical Center, 1725 W. Harrison, Suite 206, Chicago, IL, 60612, United States.
| | - Akram E Dera
- Internal Medicine, Greater Baltimore Medical Center, Towson, MD, United States.
| | | | - Alice A Lee
- Rush University Medical Center, Chicago, IL, 60612, United States.
| | - Helen J Burgess
- Biological Rhythms Research Laboratory, Rush University Medical Center, Chicago, IL, 60612, United States.
| | - Louis Fogg
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Rush University Medical Center, 1725 W. Harrison, Suite 206, Chicago, IL, 60612, United States.
| | - Garth R Swanson
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Rush University Medical Center, 1725 W. Harrison, Suite 206, Chicago, IL, 60612, United States.
| |
Collapse
|
19
|
Takeishi K, Kawaguchi H, Akioka K, Noguchi M, Arimura E, Abe M, Ushikai M, Okita S, Tanimoto A, Horiuchi M. Effects of Dietary and Lighting Conditions on Diurnal Locomotor Activity and Body Temperature in Microminipigs. ACTA ACUST UNITED AC 2018; 32:55-62. [PMID: 29275299 DOI: 10.21873/invivo.11204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/25/2017] [Accepted: 11/28/2017] [Indexed: 12/15/2022]
Abstract
The effects of dietary and lighting conditions on diurnal rhythm of locomotor activity (LA) and body temperature (BT) using four adult male microminipigs were investigated. Different feeding times, diet and lighting conditions were applied sequentially for 3 weeks in each phase as follows: Phase I: Morning mealtime, normal diet, 12-h lights on; phase II: mealtime changed to afternoon; phase III: diet changed to high-fat diet; phase IV: lighting changed to 20-h on; and phase V: phase I repeated. LA was measured by an actigraph which was worn on the body of each pig. A BT recording module (Thermochron Type-SL) was implanted in the neck subcutaneously. Phase II increased BT compared with phase I. Phase III increased LA and BT compared with phase II. Phase IV increased LA compared with phase III. LA in phase V was higher compared with phase I. These results can be extrapolated to other diurnal animals such as humans. This study provides an example of the effects of diet and lighting on biological activities in microminipigs under low-invasive procedures measuring LA and BT, leading to low variations in these measures.
Collapse
Affiliation(s)
- Kaichiro Takeishi
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Hiroaki Kawaguchi
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Kohei Akioka
- Laboratory of Veterinary Histopathology, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Michiko Noguchi
- Laboratory of Theriogenology, Azabu University, Kanagawa, Japan
| | - Emi Arimura
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan.,Department of Life and Environmental Science, Kagoshima Prefectural College, Kagoshima, Japan
| | - Masaharu Abe
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Miharu Ushikai
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Shinobu Okita
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Akihide Tanimoto
- Department of Pathology, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Masahisa Horiuchi
- Department of Hygiene and Health Promotion Medicine, Graduate School of Medical and Dental Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
20
|
Wang H, van Spyk E, Liu Q, Geyfman M, Salmans ML, Kumar V, Ihler A, Li N, Takahashi JS, Andersen B. Time-Restricted Feeding Shifts the Skin Circadian Clock and Alters UVB-Induced DNA Damage. Cell Rep 2018; 20:1061-1072. [PMID: 28768192 DOI: 10.1016/j.celrep.2017.07.022] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 06/29/2017] [Accepted: 07/12/2017] [Indexed: 01/25/2023] Open
Abstract
The epidermis is a highly regenerative barrier protecting organisms from environmental insults, including UV radiation, the main cause of skin cancer and skin aging. Here, we show that time-restricted feeding (RF) shifts the phase and alters the amplitude of the skin circadian clock and affects the expression of approximately 10% of the skin transcriptome. Furthermore, a large number of skin-expressed genes are acutely regulated by food intake. Although the circadian clock is required for daily rhythms in DNA synthesis in epidermal progenitor cells, RF-induced shifts in clock phase do not alter the phase of DNA synthesis. However, RF alters both diurnal sensitivity to UVB-induced DNA damage and expression of the key DNA repair gene, Xpa. Together, our findings indicate regulation of skin function by time of feeding and emphasize a link between circadian rhythm, food intake, and skin health.
Collapse
Affiliation(s)
- Hong Wang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA; Department of Cell Biology and Genetics, School of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Elyse van Spyk
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Qiang Liu
- Department of Computer Science, University of California, Irvine, Irvine, CA 92697, USA
| | - Mikhail Geyfman
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Michael L Salmans
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Vivek Kumar
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Alexander Ihler
- Department of Computer Science, University of California, Irvine, Irvine, CA 92697, USA
| | - Ning Li
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Joseph S Takahashi
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bogi Andersen
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA; Department of Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA; Institute for Genomics and Bioinformatics, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
21
|
Abstract
The daily rhythm of mammalian energy metabolism is subject to the circadian clock system, which is made up of the molecular clock machinery residing in nearly all cells throughout the body. The clock genes have been revealed not only to form the molecular clock but also to function as a mediator that regulates both circadian and metabolic functions. While the circadian signals generated by clock genes produce metabolic rhythms, clock gene function is tightly coupled to fundamental metabolic processes such as glucose and lipid metabolism. Therefore, defects in the clock genes not only result in the dysregulation of physiological rhythms but also induce metabolic disorders including diabetes and obesity. Among the clock genes, Dec1 (Bhlhe40/Stra13/Sharp2), Dec2 (Bhlhe41/Sharp1), and Bmal1 (Mop3/Arntl) have been shown to be particularly relevant to the regulation of energy metabolism at the cellular, tissue, and organismal levels. This paper reviews our current knowledge of the roles of Dec1, Dec2, and Bmal1 in coordinating the circadian and metabolic pathways.
Collapse
|
22
|
Shift-work: is time of eating determining metabolic health? Evidence from animal models. Proc Nutr Soc 2018; 77:199-215. [DOI: 10.1017/s0029665117004128] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The circadian disruption in shift-workers is suggested to be a risk factor to develop overweight and metabolic dysfunction. The conflicting time signals given by shifted activity, shifted food intake and exposure to light at night occurring in the shift-worker are proposed to be the cause for the loss of internal synchrony and the consequent adverse effects on body weight and metabolism. Because food elicited signals have proven to be potent entraining signals for peripheral oscillations, here we review the findings from experimental models of shift-work and verify whether they provide evidence about the causal association between shifted feeding schedules, circadian disruption and altered metabolism. We found mainly four experimental models that mimic the conditions of shift-work: protocols of forced sleep deprivation, of forced activity during the normal rest phase, exposure to light at night and shifted food timing. A big variability in the intensity and duration of the protocols was observed, which led to a diversity of effects. A common result was the disruption of temporal patterns of activity; however, not all studies explored the temporal patterns of food intake. According to studies that evaluate time of food intake as an experimental model of shift-work and studies that evaluate shifted food consumption, time of food intake may be a determining factor for the loss of balance at the circadian and metabolic level.
Collapse
|
23
|
Rao RT, Scherholz ML, Hartmanshenn C, Bae SA, Androulakis IP. On the analysis of complex biological supply chains: From Process Systems Engineering to Quantitative Systems Pharmacology. Comput Chem Eng 2017; 107:100-110. [PMID: 29353945 DOI: 10.1016/j.compchemeng.2017.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The use of models in biology has become particularly relevant as it enables investigators to develop a mechanistic framework for understanding the operating principles of living systems as well as in quantitatively predicting their response to both pathological perturbations and pharmacological interventions. This application has resulted in a synergistic convergence of systems biology and pharmacokinetic-pharmacodynamic modeling techniques that has led to the emergence of quantitative systems pharmacology (QSP). In this review, we discuss how the foundational principles of chemical process systems engineering inform the progressive development of more physiologically-based systems biology models.
Collapse
Affiliation(s)
- Rohit T Rao
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854
| | - Megerle L Scherholz
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854
| | - Clara Hartmanshenn
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854
| | - Seul-A Bae
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854
| | - Ioannis P Androulakis
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854.,Department of Biomedical Engineering, Rutgers The State University of New Jersey, 599 Taylor Road, Piscataway, NJ 08854
| |
Collapse
|
24
|
James SM, Honn KA, Gaddameedhi S, Van Dongen HPA. Shift Work: Disrupted Circadian Rhythms and Sleep-Implications for Health and Well-Being. CURRENT SLEEP MEDICINE REPORTS 2017; 3:104-112. [PMID: 29057204 DOI: 10.1007/s40675-017-0071-6] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Our 24/7 society is dependent on shift work, despite mounting evidence for negative health outcomes from sleep displacement due to shift work. This paper reviews short- and long-term health consequences of sleep displacement and circadian misalignment due to shift work. RECENT FINDINGS We focus on four broad health domains: metabolic health; risk of cancer; cardiovascular health; and mental health. Circadian misalignment affects these domains by inducing sleep deficiency, sympathovagal and hormonal imbalance, inflammation, impaired glucose metabolism, and dysregulated cell cycles. This leads to a range of medical conditions, including obesity, metabolic syndrome, type II diabetes, gastrointestinal dysfunction, compromised immune function, cardiovascular disease, excessive sleepiness, mood and social disorders, and increased cancer risk. SUMMARY Interactions of biological disturbances with behavioral and societal factors shape the effects of shift work on health and well-being. Research is needed to better understand the underlying mechanisms and drive the development of countermeasures.
Collapse
Affiliation(s)
- Stephen M James
- Sleep and Performance Research Center, Washington State University, Spokane, WA, USA.,Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Kimberly A Honn
- Sleep and Performance Research Center, Washington State University, Spokane, WA, USA.,Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Shobhan Gaddameedhi
- Sleep and Performance Research Center, Washington State University, Spokane, WA, USA.,College of Pharmacy, Washington State University, Spokane, WA, USA
| | - Hans P A Van Dongen
- Sleep and Performance Research Center, Washington State University, Spokane, WA, USA.,Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| |
Collapse
|
25
|
|
26
|
Opperhuizen AL, Wang D, Foppen E, Jansen R, Boudzovitch-Surovtseva O, de Vries J, Fliers E, Kalsbeek A. Feeding during the resting phase causes profound changes in physiology and desynchronization between liver and muscle rhythms of rats. Eur J Neurosci 2016; 44:2795-2806. [PMID: 27562056 DOI: 10.1111/ejn.13377] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 08/18/2016] [Accepted: 08/22/2016] [Indexed: 12/19/2022]
Abstract
Shiftworkers run an increased risk of developing metabolic disorders, presumably as a result of disturbed circadian physiology. Eating at a time-of-day that is normally dedicated to resting and fasting, may contribute to this association. The hypothalamus is the key brain area that integrates different inputs, including environmental time information from the central biological clock in the suprachiasmatic nuclei, with peripheral information on energy status to maintain energy homeostasis. The orexin system within the lateral hypothalamus is an important output of the suprachiasmatic nuclei involved in the control of sleep/wake behavior and glucose homeostasis, among other functions. In this study, we tested the hypothesis that feeding during the rest period disturbs the orexin system as a possible underlying contributor to metabolic health problems. Male Wistar rats were exposed to an 8-week protocol in which food was available ad libitum for 24-h, for 12-h during the light phase (i.e., unnatural feeding time) or for 12-h during the dark phase (i.e., restricted feeding, but at the natural time-of-day). Animals forced to eat at an unnatural time, i.e., during the light period, showed no changes in orexin and orexin-receptor gene expression in the hypothalamus, but the rhythmic expression of clock genes in the lateral hypothalamus was absent in these animals. Light fed animals did show adverse changes in whole-body physiology and internal desynchronization of muscle and liver clock and metabolic gene expression. Eating at the 'wrong' time-of-day thus causes internal desynchronization at different levels, which in the long run may disrupt body physiology.
Collapse
Affiliation(s)
- Anne-Loes Opperhuizen
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Dawei Wang
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands.,Institute of Plant Protection (IPP), Chinese Academy of Agricultural Science (CAAS), Beijing, China
| | - Ewout Foppen
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Remi Jansen
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Olga Boudzovitch-Surovtseva
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Janneke de Vries
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
27
|
When to eat? The influence of circadian rhythms on metabolic health: are animal studies providing the evidence? Nutr Res Rev 2016; 29:180-193. [PMID: 27364352 DOI: 10.1017/s095442241600010x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
As obesity and metabolic diseases rise, there is need to investigate physiological and behavioural aspects associated with their development. Circadian rhythms have a profound influence on metabolic processes, as they prepare the body to optimise energy use and storage. Moreover, food-related signals confer temporal order to organs involved in metabolic regulation. Therefore food intake should be synchronised with the suprachiasmatic nucleus (SCN) to elaborate efficient responses to environmental challenges. Human studies suggest that a loss of synchrony between mealtime and the SCN promotes obesity and metabolic disturbances. Animal research using different paradigms has been performed to characterise the effects of timing of food intake on metabolic profiles. Therefore the purpose of the present review is to critically examine the evidence of animal studies, to provide a state of the art on metabolic findings and to assess whether the paradigms used in rodent models give the evidence to support a 'best time' for food intake. First we analyse and compare the current findings of studies where mealtime has been shifted out of phase from the light-dark cycle. Then, we analyse studies restricting meal times to different moments within the active period. So far animal studies correlate well with human studies, demonstrating that restricting food intake to the active phase limits metabolic disturbances produced by high-energy diets and that eating during the inactive/sleep phase leads to a worse metabolic outcome. Based on the latter we discuss the missing elements and possible mechanisms leading to the metabolic consequences, as these are still lacking.
Collapse
|
28
|
The acute effects of time-of-day-dependent high fat feeding on whole body metabolic flexibility in mice. Int J Obes (Lond) 2016; 40:1444-51. [PMID: 27133618 PMCID: PMC5022109 DOI: 10.1038/ijo.2016.80] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 02/03/2016] [Accepted: 03/04/2016] [Indexed: 01/24/2023]
Abstract
Background: Both circadian disruption and timing of feeding have important roles in the development of metabolic disease. Despite growing acceptance that the timing of food consumption has long-term impact on metabolic homeostasis, little is known regarding the immediate influence on whole body metabolism, or the mechanisms involved. We aimed to examine the acute effects of time-of-day-dependent high fat feeding on whole body substrate metabolism and metabolic plasticity, and to determine the potential contribution of the adipocyte circadian clock. Methods: Mice were fed a regimen of 4-h meal at the beginning and end of the dark (waking) cycle, separated by 4 h of fasting. Daily experimental conditions consisted of either an early very high fat or high fat (EVHF or EHF, 60 or 45% kcals from fat, respectively) or late (LVHF or LHF) meal, paired with a low fat (LF, 10% kcals from fat) meal. Metabolic parameters, glucose tolerance, body fat composition and weight were assessed. To determine the role of the adipocyte circadian clock, an aP2-CLOCK mutant (ACM) mouse model was used. Results: Mice in the EVHF or EHF groups showed a 13.2 or 8.84 higher percentage of caloric intake from fat and had a 0.013 or 0.026 lower daily average respiratory exchange ratio, respectively, compared with mice eating the opposite feeding regime. Changes in glucose tolerance, body fat composition and weight were not significant at the end of the 9-day restricted feeding period. ACM mice did not exhibit different metabolic responses to the feeding regimes compared with wild-type littermates. Circadian clock disruption did not influence the short-term response to timed feeding. Conclusions: Both the total fat composition of diet and the timing of fat intake may differentially mediate the effect of timed feeding on substrate metabolism, but may not induce acute changes in metabolic flexibility.
Collapse
|
29
|
Abstract
Quantitative Systems Pharmacology (QSP) is receiving increased attention. As the momentum builds and the expectations grow it is important to (re)assess and formalize the basic concepts and approaches. In this short review, I argue that QSP, in addition to enabling the rational integration of data and development of complex models, maybe more importantly, provides the foundations for developing an integrated framework for the assessment of drugs and their impact on disease within a broader context expanding the envelope to account in great detail for physiology, environment and prior history. I articulate some of the critical enablers, major obstacles and exciting opportunities manifesting themselves along the way. Charting such overarching themes will enable practitioners to identify major and defining factors as the field progressively moves towards personalized and precision health care delivery.
Collapse
Affiliation(s)
- Ioannis P Androulakis
- Biomedical Engineering Department, Chemical & Biochemical Engineering Department, Rutgers University, Piscataway, NJ 08854
| |
Collapse
|
30
|
Aktas B, De Wolfe TJ, Tandee K, Safdar N, Darien BJ, Steele JL. The Effect of Lactobacillus casei 32G on the Mouse Cecum Microbiota and Innate Immune Response Is Dose and Time Dependent. PLoS One 2015; 10:e0145784. [PMID: 26714177 PMCID: PMC4705108 DOI: 10.1371/journal.pone.0145784] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 12/08/2015] [Indexed: 01/12/2023] Open
Abstract
Lactobacilli have been associated with a variety of immunomodulatory effects and some of these effects have been related to changes in gastrointestinal microbiota. However, the relationship between probiotic dose, time since probiotic consumption, changes in the microbiota, and immune system requires further investigation. The objective of this study was to determine if the effect of Lactobacillus casei 32G on the murine gastrointestinal microbiota and immune function are dose and time dependent. Mice were fed L. casei 32G at doses of 106, 107, or 108 CFU/day/mouse for seven days and were sacrificed 0.5h, 3.5h, 12h, or 24h after the last administration. The ileum tissue and the cecal content were collected for immune profiling by qPCR and microbiota analysis, respectively. The time required for L. casei 32G to reach the cecum was monitored by qPCR and the 32G bolus reaches the cecum 3.5h after the last administration. L. casei 32G altered the cecal microbiota with the predominance of Lachnospiraceae IS, and Oscillospira decreasing significantly (p < 0.05) in the mice receiving 108 CFU/mouse 32G relative to the control mice, while a significant (p < 0.05) increase was observed in the prevalence of lactobacilli. The lactobacilli that increased were determined to be a commensal lactobacilli. Interestingly, no significant difference in the overall microbiota composition, regardless of 32G doses, was observed at the 12h time point. A likely explanation for this observation is the level of feed derived-nutrients resulting from the 12h light/dark cycle. 32G results in consistent increases in Clec2h expression and reductions in TLR-2, alpha-defensins, and lysozyme. Changes in expression of these components of the innate immune system are one possible explanation for the observed changes in the cecal microbiota. Additionally, 32G administration was observed to alter the expression of cytokines (IL-10rb and TNF-α) in a manner consistent with an anti-inflammatory response.
Collapse
Affiliation(s)
- Busra Aktas
- Department of Food Science, University of Wisconsin, Madison, WI, United States of America
| | - Travis J. De Wolfe
- Department of Food Science, University of Wisconsin, Madison, WI, United States of America
| | - Kanokwan Tandee
- Food Science and Technology, Maejo University, Chiangmai, Thailand
| | - Nasia Safdar
- Infectious Diseases Division, Department of Medicine, University of Wisconsin, Madison, WI, United States of America
- William S. Middleton Veterans Affairs Hospital, Madison, WI, United States of America
| | - Benjamin J. Darien
- Animal Health and Biomedical Sciences, University of Wisconsin, Madison, WI, United States of America
| | - James L. Steele
- Department of Food Science, University of Wisconsin, Madison, WI, United States of America
- * E-mail:
| |
Collapse
|
31
|
Go EH, Lee SH. Effect of Long Term Reverse Feeding on the Reproductive and Non-reproductive Tissues in Male Mice. Dev Reprod 2015; 18:161-6. [PMID: 25949185 PMCID: PMC4282209 DOI: 10.12717/dr.2014.18.3.161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 08/11/2014] [Accepted: 08/18/2014] [Indexed: 01/17/2023]
Abstract
Previously, we demonstrated that the shift and/or restriction of feeding time during relatively short-term period (4 weeks) could alter the pituitary gonadotropin expression and the weights of seminal vesicle and prostate in rats. We also found that the reverse feeding (RF) schedule (up to 8 weeks) might induce an adaptable metabolic stress and cause impairment of androgen-dependent reproductive tissues. In the present study, we extended the RF time regimen up to 12 weeks, and measured the reproductive tissue weights. After 4 and 8 weeks of RF, the weights of epididymis were not significantly different. After 12 weeks, however, epididymis weights of RF animals were significantly different (CON 12W : RF 12W = 48.26±0.62 mg : 44.05±1.57 mg, p<0.05). After 4 and 12 weeks of feeding, seminal vesicle weights of RF animals were significantly decreased (CON 4W : RF 4W = 79.36±8.34 mg : 46.28±2.43 mg, p<0.001; CON 12W : RF 12W = 72.04±3.76 mg : 46.71±2.27 mg, p<0.001, respectively). Prostate weights were not changed by RF. Kidney and spleen weights of RF animals were significantly different on weeks 4 and 12 (Kidney, CON 4W : RF 4W = 249.72±4.20 mg : 228.41±3.03 mg, p<0.001; CON 12W : RF 12W = 309.15±7.49 mg : 250.72±6.13 mg, p<0.001, respectively, Spleen, CON 4W : RF 4W = 111.26±3.76 mg : 96.88±4.69 mg, p<0.05; CON 12W : RF 12W = 123.93±10.72 mg : 94.68±5.65 mg, p<0.05, respectively). Histology analysis of seminal vesicle revealed that the thinner epithelial cell layers, reduced complexities of swollen papilla folding in the exocrine glands on weeks 4 and 12 of RF. There was no histological difference between control and RF group on week 8. The present study indicates that up to 12 weeks RF induced differential changes in tissue weights of male mice. In particular, seminal vesicle, kidney and spleen seemed to temporarily adapted to the RF-induced metabolic stress on week 8 of feeding schedule. These results confirmed the our previous study that the RF might induce an adaptable metabolic stress and cause impairment of androgen-dependent reproductive tissues such as epididymis and seminal vesicle as well as non-reproductive tissues such as kidney and spleen. Further studies will be needed to achieve a better understanding of the how does mealtime shift affect the reproductive function and exact nature of adaptation.
Collapse
Affiliation(s)
- Eun Hye Go
- Department of Life Science, Sangmyung University, Seoul 110-743, Korea
| | - Sung-Ho Lee
- Department of Life Science, Sangmyung University, Seoul 110-743, Korea
| |
Collapse
|
32
|
Opperhuizen AL, van Kerkhof LWM, Proper KI, Rodenburg W, Kalsbeek A. Rodent models to study the metabolic effects of shiftwork in humans. Front Pharmacol 2015; 6:50. [PMID: 25852554 PMCID: PMC4371697 DOI: 10.3389/fphar.2015.00050] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/01/2015] [Indexed: 11/14/2022] Open
Abstract
Our current 24-h society requires an increasing number of employees to work nightshifts with millions of people worldwide working during the evening or night. Clear associations have been found between shiftwork and the risk to develop metabolic health problems, such as obesity. An increasing number of studies suggest that the underlying mechanism includes disruption of the rhythmically organized body physiology. Normally, daily 24-h rhythms in physiological processes are controlled by the central clock in the brain in close collaboration with peripheral clocks present throughout the body. Working schedules of shiftworkers greatly interfere with these normal daily rhythms by exposing the individual to contrasting inputs, i.e., at the one hand (dim)light exposure at night, nightly activity and eating and at the other hand daytime sleep and reduced light exposure. Several different animal models are being used to mimic shiftwork and study the mechanism responsible for the observed correlation between shiftwork and metabolic diseases. In this review we aim to provide an overview of the available animal studies with a focus on the four most relevant models that are being used to mimic human shiftwork: altered timing of (1) food intake, (2) activity, (3) sleep, or (4) light exposure. For all studies we scored whether and how relevant metabolic parameters, such as bodyweight, adiposity and plasma glucose were affected by the manipulation. In the discussion, we focus on differences between shiftwork models and animal species (i.e., rat and mouse). In addition, we comment on the complexity of shiftwork as an exposure and the subsequent difficulties when using animal models to investigate this condition. In view of the added value of animal models over human cohorts to study the effects and mechanisms of shiftwork, we conclude with recommendations to improve future research protocols to study the causality between shiftwork and metabolic health problems using animal models.
Collapse
Affiliation(s)
- Anne-Loes Opperhuizen
- Department of Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Hypothalamic Integration Mechanisms Amsterdam, Netherlands
| | - Linda W M van Kerkhof
- Centre for Health Protection, National Institute for Public Health and the Environment Bilthoven, Netherlands
| | - Karin I Proper
- Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment Bilthoven, Netherlands
| | - Wendy Rodenburg
- Centre for Health Protection, National Institute for Public Health and the Environment Bilthoven, Netherlands
| | - Andries Kalsbeek
- Department of Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Hypothalamic Integration Mechanisms Amsterdam, Netherlands ; Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam Amsterdam, Netherlands
| |
Collapse
|
33
|
Song H, Moon M, Choe HK, Han DH, Jang C, Kim A, Cho S, Kim K, Mook-Jung I. Aβ-induced degradation of BMAL1 and CBP leads to circadian rhythm disruption in Alzheimer's disease. Mol Neurodegener 2015; 10:13. [PMID: 25888034 PMCID: PMC4404698 DOI: 10.1186/s13024-015-0007-x] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 02/26/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Patients with Alzheimer's disease (AD) frequently experience disruption of their circadian rhythms, but whether and how circadian clock molecules are perturbed by AD remains unknown. AD is an age-related neurological disorder and amyloid-β (Aβ) is one of major causative molecules in the pathogenesis of AD. RESULTS In this study, we investigated the role of Aβ in the regulation of clock molecules and circadian rhythm using an AD mouse model. These mice exhibited altered circadian behavior, and altered expression patterns of the circadian clock genes, Bmal1 and Per2. Using cultured cells, we showed that Aβ induces post-translational degradation of the circadian clock regulator CBP, as well as the transcription factor BMAL1, which forms a complex with the master circadian transcription factor CLOCK. Aβ-induced degradation of BMAL1 and CBP correlated with the reduced binding of transcription factors to the Per2 promoter, which in turn resulted in disruptions to PER2 protein expression and the oscillation of Per2 mRNA levels. CONCLUSIONS Our results elucidate the underlying mechanisms for disrupted circadian rhythm in AD.
Collapse
Affiliation(s)
- Hyundong Song
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehak-ro, Seoul, 110-799, Jongno-gu, Korea.
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 302-718, Korea.
| | - Han Kyoung Choe
- Department of Biological Sciences, Seoul National University, Seoul, 151-742, Korea.
| | - Dong-Hee Han
- Department of Neuroscience and Neurodegeneration Control Research Center, Kyung Hee University, Seoul, 130-701, Korea.
| | - Changhwan Jang
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehak-ro, Seoul, 110-799, Jongno-gu, Korea.
| | - Ahbin Kim
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehak-ro, Seoul, 110-799, Jongno-gu, Korea.
| | - Sehyung Cho
- Department of Neuroscience and Neurodegeneration Control Research Center, Kyung Hee University, Seoul, 130-701, Korea.
| | - Kyungjin Kim
- Department of Brain Science, DGIST, Daegu, 711-873, Korea.
| | - Inhee Mook-Jung
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehak-ro, Seoul, 110-799, Jongno-gu, Korea.
| |
Collapse
|
34
|
Ramos-Lobo AM, Buonfiglio DC, Cipolla-Neto J. Streptozotocin-induced diabetes disrupts the body temperature daily rhythm in rats. Diabetol Metab Syndr 2015; 7:39. [PMID: 25960780 PMCID: PMC4424512 DOI: 10.1186/s13098-015-0035-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 04/16/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND In mammals, the temperature rhythm is regulated by the circadian pacemaker located in the suprachiasmatic nuclei, and is considered a "marker rhythm". Melatonin, the pineal gland hormone, is a major regulator of the endogenous rhythms including body temperature. Its production is influenced by many factors, such as type 1 diabetes mellitus. In rats, diabetes leads to hypothermia and reduced melatonin synthesis; insulin treatment reestablishes both. AIM To study the body temperature daily rhythm of diabetic animals and the effects of insulin and/or melatonin treatment on its structure. METHODS We studied the effects of streptozotocin-induced diabetes (60 mg/kg) on the body temperature rhythm of Wistar rats and the possible modifications resulting from early and late treatments with insulin (6U/day) and/or melatonin (daily 0.5 mg/kg). We monitored the daily body temperature rhythm, its rhythmic parameters (MESOR, amplitude and acrophase), glycemia and body weight for 55 days. Data were classified by groups and expressed as mean ± SEM. One-way ANOVA analysis was performed followed by Bonferroni posttest. Statistical significance was set at p < 0.05. RESULTS Diabetes led to complete disruption of the temperature rhythm and hypothermia, which were accentuated over time. All early treatments (insulin or/and melatonin) prevented the temperature rhythm disruption and hypothermia. Insulin plus melatonin restored the body temperature rhythm whereas insulin alone resulted less efficient; melatonin alone did not restore any of the parameters studied; however, when supplemented close to diabetes onset, it maintained the temperature rhythmicity. All these corrective effects of the early treatments were dependent on the continuous maintenance of the treatment. CONCLUSIONS Taken together, our findings show the disruption of the body temperature daily rhythm, a new consequence of insulin-dependent diabetes, as well as the beneficial effect of the complementary action of melatonin and insulin restoring the normal rhythmicity.
Collapse
Affiliation(s)
- Angela M Ramos-Lobo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Lineu Prestes, 1524, São Paulo, SP 05508-000 Brazil
| | - Daniella C Buonfiglio
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Lineu Prestes, 1524, São Paulo, SP 05508-000 Brazil
| | - José Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Lineu Prestes, 1524, São Paulo, SP 05508-000 Brazil
| |
Collapse
|
35
|
Sonnier T, Rood J, Gimble JM, Peterson CM. Glycemic control is impaired in the evening in prediabetes through multiple diurnal rhythms. J Diabetes Complications 2014; 28:836-43. [PMID: 24835190 DOI: 10.1016/j.jdiacomp.2014.04.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 03/11/2014] [Accepted: 04/01/2014] [Indexed: 01/12/2023]
Abstract
AIMS Recent studies suggest that circadian rhythms regulate glucose metabolism, weight loss, and even drug efficacy. Moreover, molecules targeted at the circadian clock show promise in treating metabolic disease. Therefore, this study set out to better characterize interactions among diurnal rhythms in prediabetes. METHODS Ten subjects with prediabetes completed oral glucose tolerance tests at 0700h and 1900h on the same day. Lipids and hormones were also measured. RESULTS Two-hour and three-hour glucose tolerances were worse in the evening by 40±52mg/dl (p=0.02) and 62±46mg/dl (p=0.001), respectively. These impairments were explained by lower insulin sensitivity (OGIS; 5.14±1.02 vs. 4.74±0.77mg/kg/min; p=0.03) and 2-hour AUC insulin levels (87.4±37.6 vs. 69.8±40.2mU∙hr/l; p=0.02) in the evening. Intriguingly, more insulin resistant subjects had weaker rhythms in insulin sensitivity (r=-0.66; p=0.04) but enhanced rhythms in insulin (r=-0.67; p=0.03) and cortisol (r=-0.78; p=0.008) levels. Importantly, the rhythms in cortisol primarily but also insulin sensitivity drove the declines in evening glucose tolerance (r=0.86; p=0.002). CONCLUSIONS Glycemic control is dramatically impaired in the evening in people with prediabetes, particularly when the cortisol rhythm is weak, but is unrelated to the rhythm in insulin levels. Therefore, food intake at dinnertime may need to be curbed in prediabetes.
Collapse
Affiliation(s)
- Tance Sonnier
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | - Jennifer Rood
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | - Jeffrey M Gimble
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA; Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA; LaCell LLC, New Orleans, LA 70112, USA
| | - Courtney M Peterson
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| |
Collapse
|
36
|
Wu T, Guo A, Shu Q, Qi Y, Kong Y, Sun Z, Sun S, Fu Z. L-Carnitine intake prevents irregular feeding-induced obesity and lipid metabolism disorder. Gene 2014; 554:148-54. [PMID: 25445284 DOI: 10.1016/j.gene.2014.10.040] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 10/20/2014] [Accepted: 10/23/2014] [Indexed: 01/07/2023]
Abstract
L-Carnitine supplementation has been used to reduce obesity caused by high-fat diet, which is beneficial for lowering blood and hepatic lipid levels, and for ameliorating fatty liver. However, whether l-carnitine may affect irregular feeding-induced obesity and lipid metabolism disorder is still largely unknown. In the present study, we developed a time-delayed pattern of eating, and investigated the effects of l-carnitine on the irregular eating induced adiposity in mice. After an experimental period of 8 weeks with l-carnitine supplementation, l-carnitine significantly inhibited body weight increase and epididymal fat weight gain induced by the time-delayed feeding. In addition, l-carnitine administration decreased levels of serum alanine aminotransferase (GPT), glutamic oxalacetic transaminase (GOT) and triglyceride (TG), which were significantly elevated by the irregular feeding. Moreover, mice supplemented with l-carnitine did not display glucose intolerance-associated hallmarks, which were found in the irregular feeding-induced obesity. Furthermore, quantitative real-time polymerase chain reaction (qRT-PCR) analysis indicated that l-carnitine counteracted the negative alterations of lipid metabolic gene expression (fatty acid synthase, 3-hydroxy-3-methyl-glutaryl coenzyme A reductase, cholesterol 7α-hydroxylase, carnitine/acylcarnitine translocase) in the liver and fat of mice caused by the irregular feeding. Therefore, our results suggest that the time-delayed pattern of eating can induce adiposity and lipid metabolic disorders, while l-carnitine supplementation might prevent these negative symptoms.
Collapse
Affiliation(s)
- Tao Wu
- College of Biological and Environmental Engineering, Zhejiang University of Technology, China
| | - Anqi Guo
- College of Biological and Environmental Engineering, Zhejiang University of Technology, China
| | - Qingyu Shu
- College of Biological and Environmental Engineering, Zhejiang University of Technology, China
| | - Yangjian Qi
- College of Biological and Environmental Engineering, Zhejiang University of Technology, China
| | - Ying Kong
- College of Biological and Environmental Engineering, Zhejiang University of Technology, China
| | - Zhiping Sun
- College of Biological and Environmental Engineering, Zhejiang University of Technology, China
| | - Shumin Sun
- College of Biological and Environmental Engineering, Zhejiang University of Technology, China
| | - Zhengwei Fu
- College of Biological and Environmental Engineering, Zhejiang University of Technology, China.
| |
Collapse
|
37
|
Arul D, Subramanian P. Daytime food restriction influences the circadian rhythms of circulatory hepatic marker enzymes activity in rats. BIOL RHYTHM RES 2014. [DOI: 10.1080/09291016.2014.884306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
38
|
Ramphal L, Zhang J, Suzuki S. A Cohort Analysis of the Cardiovascular Risk Factors in the Employees of a Pediatric Hospital from 2009 to 2012. Proc (Bayl Univ Med Cent) 2014; 27:96-9. [DOI: 10.1080/08998280.2014.11929067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
39
|
Sunderram J, Sofou S, Kamisoglu K, Karantza V, Androulakis IP. Time-restricted feeding and the realignment of biological rhythms: translational opportunities and challenges. J Transl Med 2014; 12:79. [PMID: 24674294 PMCID: PMC3973614 DOI: 10.1186/1479-5876-12-79] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 03/10/2014] [Indexed: 12/15/2022] Open
Abstract
It has been argued that circadian dysregulation is not only a critical inducer and promoter of adverse health effects, exacerbating symptom burden, but also hampers recovery. Therefore understanding the health-promoting roles of regulating (i.e., restoring) circadian rhythms, thus suppressing harmful effects of circadian dysregulation, would likely improve treatment. At a critical care setting it has been argued that studies are warranted to determine whether there is any use in restoring circadian rhythms in critically ill patients, what therapeutic goals should be targeted, and how these could be achieved. Particularly interesting are interventional approaches aiming at optimizing the time of feeding in relation to individualized day-night cycles for patients receiving enteral nutrition, in an attempt to re-establish circadian patterns of molecular expression. In this short review we wish to explore the idea of transiently imposing (appropriate, but yet to be determined) circadian rhythmicity via regulation of food intake as a means of exploring rhythm-setting properties of metabolic cues in the context of improving immune response. We highlight some of the key elements associated with his complex question particularly as they relate to: a) stress and rhythmic variability; and b) metabolic entrainment of peripheral tissues as a possible intervention strategy through time-restricted feeding. Finally, we discuss the challenges and opportunities for translating these ideas to the bedside.
Collapse
Affiliation(s)
- Jag Sunderram
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Rutgers - Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Stavroula Sofou
- Biomedical Engineering Department, Rutgers University, Piscataway, NJ 08854, USA
- Chemical & Biochemical Engineering Department, Rutgers University, Piscataway, NJ 08854, USA
| | - Kubra Kamisoglu
- Chemical & Biochemical Engineering Department, Rutgers University, Piscataway, NJ 08854, USA
| | - Vassiliki Karantza
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick 08901, USA
| | - Ioannis P Androulakis
- Biomedical Engineering Department, Rutgers University, Piscataway, NJ 08854, USA
- Chemical & Biochemical Engineering Department, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
40
|
Reddy VDK, Jagota A. Effect of restricted feeding on nocturnality and daily leptin rhythms in OVLT in aged male Wistar rats. Biogerontology 2014; 15:245-56. [PMID: 24619733 DOI: 10.1007/s10522-014-9494-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 02/28/2014] [Indexed: 01/08/2023]
Abstract
Circadian system has direct relevance to the problems of modern lifestyle, shift workers, jet lag etc. To understand non-photic regulation of biological clock, the effects of restricted feeding (RF) on locomotor activity and daily leptin immunoreactivity (ir) rhythms in three age groups [3, 12 and 24 months (m)] of male Wistar rats maintained in light:dark (LD) 12:12 h conditions were studied. Leptin-ir was examined in the suprachiasmatic nucleus (SCN), the medial preoptic area (MPOA) and organum vasculosum of the lamina terminalis (OVLT). Reversal of feeding time due to restricted food availability during daytime resulted in switching of the animals from nocturnality to diurnality with significant increase in day time activity and decrease in night time activity. The RF resulted in % diurnality of approximately 32, 29 and 73 from % nocturnality of 82, 92 and 89 in control rats of 3, 12 and 24 m age, respectively. The increase in such switching from nocturnality to diurnality with restricted feeding was found to be robust in 24 m rats. The OVLT region showed daily leptin-ir rhythms with leptin-ir maximum at ZT-0 in all the three age groups. However leptin-ir levels were minimum at ZT-12 in 3 and 12 m though at ZT-18 in 24 m. In addition the mean leptin-ir levels decreased with increase in food intake and body weight significantly in RF aged rats. Thus we report here differential effects of food entrained regulation in switching nocturnality to diurnality and daily leptin-ir rhythms in OVLT in aged rats.
Collapse
Affiliation(s)
- V D K Reddy
- Neurobiology and Molecular Chronobiology Laboratory, Department of Animal Sciences, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Andhra Pradesh, India
| | | |
Collapse
|
41
|
Hwang JW, Sundar IK, Yao H, Sellix MT, Rahman I. Circadian clock function is disrupted by environmental tobacco/cigarette smoke, leading to lung inflammation and injury via a SIRT1-BMAL1 pathway. FASEB J 2014; 28:176-94. [PMID: 24025728 PMCID: PMC3868829 DOI: 10.1096/fj.13-232629] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 08/26/2013] [Indexed: 02/03/2023]
Abstract
Patients with obstructive lung diseases display abnormal circadian rhythms in lung function. We determined the mechanism whereby environmental tobacco/cigarette smoke (CS) modulates expression of the core clock gene BMAL1, through Sirtuin1 (SIRT1) deacetylase during lung inflammatory and injurious responses. Adult C57BL6/J and various mice mutant for SIRT1 and BMAL1 were exposed to both chronic (6 mo) and acute (3 and 10 d) CS, and we measured the rhythmic expression of clock genes, circadian rhythms of locomotor activity, lung function, and inflammatory and emphysematous responses in the lungs. CS exposure (100-300 mg/m(3) particulates) altered clock gene expression and reduced locomotor activity by disrupting the central and peripheral clocks and increased lung inflammation, causing emphysema in mice. BMAL1 was acetylated and degraded in the lungs of mice exposed to CS and in patients with chronic obstructive pulmonary disease (COPD), compared with lungs of the nonsmoking controls, linking it mechanistically to CS-induced reduction of SIRT1. Targeted deletion of Bmal1 in lung epithelium augmented inflammation in response to CS, which was not attenuated by the selective SIRT1 activator SRT1720 (EC50=0.16 μM) in these mice. Thus, the circadian clock, specifically the enhancer BMAL1 in epithelium, plays a pivotal role, mediated by SIRT1-dependent BMAL1, in the regulation of CS-induced lung inflammatory and injurious responses.
Collapse
Affiliation(s)
- Jae-Woong Hwang
- 2Department of Environmental Medicine, University of Rochester Medical Center, Box 850, 601 Elmwood Avenue, Rochester 14642, NY, USA.
| | | | | | | | | |
Collapse
|
42
|
Kim TS, Han DH, Lee YJ, Son GH, Kim K, Kim CJ, Cho S. Adrenal peripheral clock disruption leads to altered circadian behavioral responses to voluntary exercise in middle-aged female mice. Anim Cells Syst (Seoul) 2013. [DOI: 10.1080/19768354.2013.860916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
43
|
Lee YJ, Han DH, Pak YK, Cho SH. Circadian regulation of low density lipoprotein receptor promoter activity by CLOCK/BMAL1, Hes1 and Hes6. Exp Mol Med 2012; 44:642-52. [PMID: 22913986 PMCID: PMC3509181 DOI: 10.3858/emm.2012.44.11.073] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2012] [Indexed: 12/28/2022] Open
Abstract
Low density lipoprotein receptor (LDLR) plays an important role in the cholesterol homeostasis. We examined the possible circadian regulation of LDLR and mechanism(s) underlying it. In mice, blood glucose and plasma triglyceride, total and high density lipoprotein cholesterol varied distinctively throughout a day. In addition, LDLR mRNA oscillated in the liver in a functional clock-dependent manner. Accordingly, analysis of human LDLR promoter sequence revealed three putative E-boxes, raising the possible regulation of LDLR expression by E-box-binding transcription factors. To test this possibility, human LDLR promoter reporter constructs were transfected into HepG2 cells and the effects of CLOCK/BMAL1, Hes1, and Hes6 expression were analyzed. It was found that positive circadian transcription factor complex CLOCK/BMAL1 upregulated human LDLR promoter activity in a serum-independent manner, while Hes family members Hes1 and Hes6 downregulated it only under serum-depleted conditions. Both effects were mapped to proximal promoter region of human LDLR, where mutation or deletion of well-known sterol regulatory element (SRE) abolished only the repressive effect of Hes1. Interestingly, hes6 and hes1 mRNA oscillated in an anti-phasic manner in the wild-type but not in the per1-/-per2 -/- mouse. Comparative analysis of mouse, rat and human hes6 genes revealed that three E-boxes are conserved among three species. Transfection and site-directed mutagenesis studies with hes6 reporter constructs confirmed that the third E-box in the exon IV is functionally induced by CLOCK/BMAL1. Taken together, these results suggest that LDLR expression is under circadian control involving CLOCK/BMAL1 and Hes family members Hes1 and Hes6.
Collapse
Affiliation(s)
- Yeon Ju Lee
- Department of Neuroscience and Neurodegeneration Control Research Center, Kyung Hee University, Seoul 130-701, Korea
| | | | | | | |
Collapse
|