1
|
Mortimer T, Smith JG, Muñoz-Cánoves P, Benitah SA. Circadian clock communication during homeostasis and ageing. Nat Rev Mol Cell Biol 2025; 26:314-331. [PMID: 39753699 DOI: 10.1038/s41580-024-00802-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 03/28/2025]
Abstract
Maintaining homeostasis is essential for continued health, and the progressive decay of homeostatic processes is a hallmark of ageing. Daily environmental rhythms threaten homeostasis, and circadian clocks have evolved to execute physiological processes in a manner that anticipates, and thus mitigates, their effects on the organism. Clocks are active in almost all cell types; their rhythmicity and functional output are determined by a combination of tissue-intrinsic and systemic inputs. Numerous inputs for a specific tissue are produced by the activity of circadian clocks of other tissues or cell types, generating a form of crosstalk known as clock communication. In mammals, the central clock in the hypothalamus integrates signals from external light-dark cycles to align peripheral clocks elsewhere in the body. This regulation is complemented by a tissue-specific milieu of external, systemic and niche inputs that modulate and cooperate with the cellular circadian clock machinery of a tissue to tailor its functional output. These mechanisms of clock communication decay during ageing, and growing evidence suggests that this decline might drive ageing-related morbidities. Dietary, behavioural and pharmacological interventions may offer the possibility to overcome these changes and in turn improve healthspan.
Collapse
Affiliation(s)
- Thomas Mortimer
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| | - Jacob G Smith
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain.
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain.
| | - Pura Muñoz-Cánoves
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
- Altos Labs Inc., San Diego Institute of Science, San Diego, CA, USA.
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| |
Collapse
|
2
|
Yao N, Kinouchi K, Katoh M, Ashtiani KC, Abdelkarim S, Morimoto H, Torimitsu T, Kozuma T, Iwahara A, Kosugi S, Komuro J, Kato K, Tonomura S, Nakamura T, Itoh A, Yamaguchi S, Yoshino J, Irie J, Hashimoto H, Yuasa S, Satoh A, Mikami Y, Uchida S, Ueki T, Nomura S, Baldi P, Hayashi K, Itoh H. Maternal circadian rhythms during pregnancy dictate metabolic plasticity in offspring. Cell Metab 2025; 37:395-412.e6. [PMID: 39814018 PMCID: PMC11872692 DOI: 10.1016/j.cmet.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 04/29/2024] [Accepted: 12/04/2024] [Indexed: 01/18/2025]
Abstract
Tissue-level oscillation is achieved by tissue-intrinsic clocks along with network-dependent signals originating from distal organs and organismal behavior. Yet, it remains unexplored whether maternal circadian rhythms during pregnancy influence fetal rhythms and impact long-term susceptibility to dietary challenges in offspring. Here, we demonstrate that circadian disruption during pregnancy decreased placental and neonatal weight yet retained transcriptional and structural maturation. Intriguingly, diet-induced obesity was exacerbated in parallel with arrhythmic feeding behavior, hypothalamic leptin resistance, and hepatic circadian reprogramming in offspring of chronodisrupted mothers. In utero circadian desynchrony altered the phase-relationship between the mother and fetus and impacted placental efficiency. Temporal feeding restriction in offspring failed to fully prevent obesity, whereas the circadian alignment of caloric restriction with the onset of the active phase virtually ameliorated the phenotype. Thus, maternal circadian rhythms during pregnancy confer adaptive properties to metabolic functions in offspring and provide insights into the developmental origins of health and disease.
Collapse
Affiliation(s)
- Na Yao
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kenichiro Kinouchi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Manami Katoh
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Frontier Cardiovascular Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Sherif Abdelkarim
- Department of Computer Science, University of California, Irvine, Irvine, CA 92697, USA
| | - Hiroyuki Morimoto
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takuto Torimitsu
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takahide Kozuma
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akihide Iwahara
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shotaro Kosugi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; Health Center, Keio University, Yokohama, Japan
| | - Jin Komuro
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Kyosuke Kato
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shun Tonomura
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Toshifumi Nakamura
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Arata Itoh
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shintaro Yamaguchi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Jun Yoshino
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo, Japan; The Center for Integrated Kidney Research and Advance (IKRA), Faculty of Medicine, Shimane University, Izumo, Japan
| | - Junichiro Irie
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hisayuki Hashimoto
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Shinsuke Yuasa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan; Department of Cardiovascular Medicine, Academic Field, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Akiko Satoh
- Department of Integrative Physiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan; Department of Integrative Physiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shusaku Uchida
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takatoshi Ueki
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Frontier Cardiovascular Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Pierre Baldi
- Department of Computer Science, University of California, Irvine, Irvine, CA 92697, USA
| | - Kaori Hayashi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Itoh
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; Center for Preventive Medicine, Keio University, Tokyo, Japan.
| |
Collapse
|
3
|
Parrotta ME, Colangeli L, Scipione V, Vitale C, Sbraccia P, Guglielmi V. Time Restricted Eating: A Valuable Alternative to Calorie Restriction for Addressing Obesity? Curr Obes Rep 2025; 14:17. [PMID: 39899119 PMCID: PMC11790783 DOI: 10.1007/s13679-025-00609-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE OF REVIEW In this review, we summarize the molecular effects of time-restricted eating (TRE) and its possible role in appetite regulation. We also discuss the potential clinical benefits of TRE in obesity. RECENT FINDINGS TRE is an emerging dietary approach consisting in limiting food intake to a specific window of time each day. The rationale behind this strategy is to restore the circadian misalignment, commonly seen in obesity. Preclinical studies have shown that restricting food intake only during the active phase of the day can positively influence several cellular functions including senescence, mitochondrial activity, inflammation, autophagy and nutrients' sensing pathways. Furthermore, TRE may play a role by modulating appetite and satiety hormones, though further research is needed to clarify its exact mechanisms. Clinical trials involving patients with obesity or type 2 diabetes suggest that TRE can be effective for weight loss, but its broader effects on improving other clinical outcomes, such as cardiovascular risk factors, remain less certain. The epidemic proportions of obesity cause urgency to find dietary, pharmacological and surgical interventions that can be effective in the medium and long term. According to its molecular effects, TRE can be an interesting alternative to caloric restriction in the treatment of obesity, but the considerable variability across clinical trials regarding population, intervention, and follow-up duration makes it difficult to reach definitive conclusions.
Collapse
Affiliation(s)
| | - Luca Colangeli
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Valeria Scipione
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carolina Vitale
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Valeria Guglielmi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
- Internal Medicine Unit - Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy.
| |
Collapse
|
4
|
Peng L, Xiang S, Wang T, Yang M, Duan Y, Ma X, Li S, Yu C, Zhang X, Hu H, Liu Z, Sun J, Sun C, Wang C, Liu B, Wang Z, Qian M. The hepatic clock synergizes with HIF-1α to regulate nucleotide availability during liver damage repair. Nat Metab 2025; 7:148-165. [PMID: 39775529 DOI: 10.1038/s42255-024-01184-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 11/18/2024] [Indexed: 01/11/2025]
Abstract
Nucleotide availability is crucial for DNA replication and repair; however, the coordinating mechanisms in vivo remain unclear. Here, we show that the circadian clock in the liver controls the activity of the pentose phosphate pathway (PPP) to support de novo nucleotide biosynthesis for DNA synthesis demands. We demonstrate that disrupting the hepatic clock by genetic manipulation or mistimed feeding impairs PPP activity in male mice, leading to nucleotide imbalance. Such defects not only elicit DNA replication stress to limit liver regeneration after resection but also allow genotoxin-induced hepatocyte senescence and STING signalling-dependent inflammation. Mechanistically, the molecular clock activator BMAL1 synergizes with hypoxia-inducible factor-1α (HIF-1α) to regulate the transcription of the PPP rate-limiting enzyme glucose-6-phosphate dehydrogenase (G6PD), which is enhanced during liver regeneration. Overexpressing G6PD restores the compromised regenerative capacity of the BMAL1- or HIF-1α-deficient liver. Moreover, boosting G6PD expression genetically or through preoperative intermittent fasting potently facilitates liver repair in normal mice. Hence, our findings highlight the physiological importance of the hepatic clock and suggest a promising pro-regenerative strategy.
Collapse
Affiliation(s)
- Linyuan Peng
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Siliang Xiang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Center for Anti-aging and Regenerative Medicine, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Tianzhi Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Mei Yang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yajun Duan
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiaoyu Ma
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Su Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Cong Yu
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Center for Anti-aging and Regenerative Medicine, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xin Zhang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Haiyang Hu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zuojun Liu
- School of Life Sciences, Hainan University, Haikou, China
| | - Jie Sun
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Center for Anti-aging and Regenerative Medicine, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Chunmeng Sun
- State Key Laboratory of Natural Medicines, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Baohua Liu
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Center for Anti-aging and Regenerative Medicine, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, China.
| | - Zhongyuan Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| | - Minxian Qian
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
5
|
Fu M, Lu S, Gong L, Zhou Y, Wei F, Duan Z, Xiang R, Gonzalez FJ, Li G. Intermittent fasting shifts the diurnal transcriptome atlas of transcription factors. Mol Cell Biochem 2025; 480:491-504. [PMID: 38528297 DOI: 10.1007/s11010-024-04928-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/05/2024] [Indexed: 03/27/2024]
Abstract
Intermittent fasting remains a safe and effective strategy to ameliorate various age-related diseases, but its specific mechanisms are not fully understood. Considering that transcription factors (TFs) determine the response to environmental signals, here, we profiled the diurnal expression of 600 samples across four metabolic tissues sampled every 4 over 24 h from mice placed on five different feeding regimens to provide an atlas of TFs in biological space, time, and feeding regimen. Results showed that 1218 TFs exhibited tissue-specific and temporal expression profiles in ad libitum mice, of which 974 displayed significant oscillations at least in one tissue. Intermittent fasting triggered more than 90% (1161 in 1234) of TFs to oscillate somewhere in the body and repartitioned their tissue-specific expression. A single round of fasting generally promoted TF expression, especially in skeletal muscle and adipose tissues, while intermittent fasting mainly suppressed TF expression. Intermittent fasting down-regulated aging pathway and upregulated the pathway responsible for the inhibition of mammalian target of rapamycin (mTOR). Intermittent fasting shifts the diurnal transcriptome atlas of TFs, and mTOR inhibition may orchestrate intermittent fasting-induced health improvements. This atlas offers a reference and resource to understand how TFs and intermittent fasting may contribute to diurnal rhythm oscillation and bring about specific health benefits.
Collapse
Affiliation(s)
- Min Fu
- Department of Neurology, The Fourth Hospital of Changsha, Affiliated Changsha Hospital of Hunan Normal University, Changsha, 410006, Hunan, China
| | - Siyu Lu
- Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Lijun Gong
- Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Yiming Zhou
- Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Fang Wei
- Department of Neurology, The Fourth Hospital of Changsha, Affiliated Changsha Hospital of Hunan Normal University, Changsha, 410006, Hunan, China.
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
| | - Zhigui Duan
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Rong Xiang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 41001, Hunan, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Guolin Li
- Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China.
- Center for Aging Biomedicine, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
| |
Collapse
|
6
|
Lenihan‐Geels G, Garcia Carrizo F, Leer M, Gohlke S, Oster M, Pöhle‐Kronawitter S, Ott C, Chadt A, Reinisch IN, Galhuber M, Li C, Jonas W, Jähnert M, Klaus S, Al‐Hasani H, Grune T, Schürmann A, Madl T, Prokesch A, Schupp M, Schulz TJ. Skeletal muscle p53-depletion uncovers a mechanism of fuel usage suppression that enables efficient energy conservation. J Cachexia Sarcopenia Muscle 2024; 15:1772-1784. [PMID: 39010299 PMCID: PMC11446685 DOI: 10.1002/jcsm.13529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 03/25/2024] [Accepted: 06/03/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND The ability of skeletal muscle to respond adequately to changes in nutrient availability, known as metabolic flexibility, is essential for the maintenance of metabolic health and loss of flexibility contributes to the development of diabetes and obesity. The tumour suppressor protein, p53, has been linked to the control of energy metabolism. We assessed its role in the acute control of nutrient allocation in skeletal muscle in the context of limited nutrient availability. METHODS A mouse model with inducible deletion of the p53-encoding gene, Trp53, in skeletal muscle was generated using the Cre-loxP-system. A detailed analysis of nutrient metabolism in mice with control and knockout genotypes was performed under ad libitum fed and fasting conditions and in exercised mice. RESULTS Acute deletion of p53 in myofibres of mice activated catabolic nutrient usage pathways even under ad libitum fed conditions, resulting in significantly increased overall energy expenditure (+10.6%; P = 0.0385) and a severe nutrient deficit in muscle characterized by depleted intramuscular glucose and glycogen levels (-62,0%; P < 0.0001 and -52.7%; P < 0.0001, respectively). This was accompanied by changes in marker gene expression patterns of circadian rhythmicity and hyperactivity (+57.4%; P = 0.0068). These metabolic changes occurred acutely, within 2-3 days after deletion of Trp53 was initiated, suggesting a rapid adaptive response to loss of p53, which resulted in a transient increase in lactate release to the circulation (+46.6%; P = 0.0115) from non-exercised muscle as a result of elevated carbohydrate mobilization. Conversely, an impairment of proteostasis and amino acid metabolism was observed in knockout mice during fasting. During endurance exercise testing, mice with acute, muscle-specific Trp53 inactivation displayed an early exhaustion phenotype with a premature shift in fuel usage and reductions in multiple performance parameters, including a significantly reduced running time and distance (-13.8%; P = 0.049 and -22.2%; P = 0.0384, respectively). CONCLUSIONS These findings suggest that efficient nutrient conservation is a key element of normal metabolic homeostasis that is sustained by p53. The homeostatic state in metabolic tissues is actively maintained to coordinate efficient energy conservation and metabolic flexibility towards nutrient stress. The acute deletion of Trp53 unlocks mechanisms that suppress the activity of nutrient catabolic pathways, causing substantial loss of intramuscular energy stores, which contributes to a fasting-like state in muscle tissue. Altogether, these findings uncover a novel function of p53 in the short-term regulation of nutrient metabolism in skeletal muscle and show that p53 serves to maintain metabolic homeostasis and efficient energy conservation.
Collapse
Affiliation(s)
- Georgia Lenihan‐Geels
- Department of Adipocyte Development and NutritionGerman Institute of Human Nutrition Potsdam‐Rehbrücke (DIfE)NuthetalGermany
| | - Francisco Garcia Carrizo
- Department of Adipocyte Development and NutritionGerman Institute of Human Nutrition Potsdam‐Rehbrücke (DIfE)NuthetalGermany
- German Center for Diabetes Research (DZD)München‐NeuherbergGermany
| | - Marina Leer
- Department of Adipocyte Development and NutritionGerman Institute of Human Nutrition Potsdam‐Rehbrücke (DIfE)NuthetalGermany
- German Center for Diabetes Research (DZD)München‐NeuherbergGermany
| | - Sabrina Gohlke
- Department of Adipocyte Development and NutritionGerman Institute of Human Nutrition Potsdam‐Rehbrücke (DIfE)NuthetalGermany
| | - Moritz Oster
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular‐Metabolic‐Renal ResearchBerlinGermany
| | - Sophie Pöhle‐Kronawitter
- Department of Molecular ToxicologyGerman Institute of Human Nutrition Potsdam‐RehbrückeNuthetalGermany
| | - Christiane Ott
- Department of Molecular ToxicologyGerman Institute of Human Nutrition Potsdam‐RehbrückeNuthetalGermany
| | - Alexandra Chadt
- German Center for Diabetes Research (DZD)München‐NeuherbergGermany
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ)Leibniz Center for Diabetes Research at Heinrich Heine UniversityDüsseldorfGermany
| | - Isabel N. Reinisch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and EmbryologyMedical University of GrazGrazAustria
| | - Markus Galhuber
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and EmbryologyMedical University of GrazGrazAustria
| | - Chen Li
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular‐Metabolic‐Renal ResearchBerlinGermany
| | - Wenke Jonas
- German Center for Diabetes Research (DZD)München‐NeuherbergGermany
- Department of Experimental DiabetologyGerman Institute of Human Nutrition Potsdam‐RehbrückeNuthetalGermany
| | - Markus Jähnert
- German Center for Diabetes Research (DZD)München‐NeuherbergGermany
- Department of Experimental DiabetologyGerman Institute of Human Nutrition Potsdam‐RehbrückeNuthetalGermany
| | - Susanne Klaus
- Department Physiology of Energy MetabolismGerman Institute of Human Nutrition Potsdam‐RehbrückeNuthetalGermany
| | - Hadi Al‐Hasani
- German Center for Diabetes Research (DZD)München‐NeuherbergGermany
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ)Leibniz Center for Diabetes Research at Heinrich Heine UniversityDüsseldorfGermany
| | - Tilman Grune
- German Center for Diabetes Research (DZD)München‐NeuherbergGermany
- Department of Molecular ToxicologyGerman Institute of Human Nutrition Potsdam‐RehbrückeNuthetalGermany
| | - Annette Schürmann
- German Center for Diabetes Research (DZD)München‐NeuherbergGermany
- Department of Experimental DiabetologyGerman Institute of Human Nutrition Potsdam‐RehbrückeNuthetalGermany
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Molecular Biology and BiochemistryMedical University of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
| | - Andreas Prokesch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and EmbryologyMedical University of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
| | - Michael Schupp
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinInstitute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular‐Metabolic‐Renal ResearchBerlinGermany
| | - Tim J. Schulz
- Department of Adipocyte Development and NutritionGerman Institute of Human Nutrition Potsdam‐Rehbrücke (DIfE)NuthetalGermany
- German Center for Diabetes Research (DZD)München‐NeuherbergGermany
- Institute of Nutritional ScienceUniversity of Potsdam, Potsdam‐RehbrückeNuthetalGermany
| |
Collapse
|
7
|
Chesters RA, Zhu J, Coull BM, Baidoe-Ansah D, Baumer L, Palm L, Klinghammer N, Chen S, Hahm A, Yagoub S, Cantacorps L, Bernardi D, Ritter K, Lippert RN. Fasting-induced activity changes in MC3R neurons of the paraventricular nucleus of the thalamus. Life Sci Alliance 2024; 7:e202402754. [PMID: 39107065 PMCID: PMC11303869 DOI: 10.26508/lsa.202402754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/09/2024] Open
Abstract
The brain controls energy homeostasis by regulating food intake through signaling within the melanocortin system. Whilst we understand the role of the hypothalamus within this system, how extra-hypothalamic brain regions are involved in controlling energy balance remains unclear. Here we show that the melanocortin 3 receptor (MC3R) is expressed in the paraventricular nucleus of the thalamus (PVT). We tested whether fasting would change the activity of MC3R neurons in this region by assessing the levels of c-Fos and pCREB as neuronal activity markers. We determined that overnight fasting causes a significant reduction in pCREB levels within PVT-MC3R neurons. We then questioned whether perturbation of MC3R signaling, during fasting, would result in altered refeeding. Using chemogenetic approaches, we show that modulation of MC3R activity, during the fasting period, does not impact body weight regain or total food intake in the refeeding period. However, we did observe significant differences in the pattern of feeding-related behavior. These findings suggest that the PVT is a region where MC3R neurons respond to energy deprivation and modulate refeeding behavior.
Collapse
Affiliation(s)
- Robert A Chesters
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Jiajie Zhu
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin, Berlin, Germany
| | - Bethany M Coull
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin, Berlin, Germany
| | - David Baidoe-Ansah
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin, Berlin, Germany
| | - Lea Baumer
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Lydia Palm
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Niklas Klinghammer
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Seve Chen
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Anneke Hahm
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Selma Yagoub
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition, Nuthetal, Germany
| | - Lídia Cantacorps
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Daniel Bernardi
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition Nuthetal, Germany
| | - Katrin Ritter
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition Nuthetal, Germany
| | - Rachel N Lippert
- Department of Neurocircuit Development and Function, German Institute of Human Nutrition Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
8
|
Qi D, Huang D, Ba M, Xuan S, Si H, Lu D, Pei X, Zhang W, Huang S, Li Z. Long-term high fructose intake reprograms the circadian transcriptome and disrupts homeostasis in mouse extra-orbital lacrimal glands. Exp Eye Res 2024; 246:110008. [PMID: 39025460 DOI: 10.1016/j.exer.2024.110008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
This study aims to explore the effects of long-term high fructose intake (LHFI) on the structure, functionality, and physiological homeostasis of mouse extra-orbital lacrimal glands (ELGs), a critical component of ocular health. Our findings reveal significant reprogramming of the circadian transcriptome in ELGs following LHFI, alongside the activation of specific inflammatory pathways, as well as metabolic and neural pathways. Notably, LHFI resulted in increased inflammatory infiltration, enhanced lipid deposition, and reduced nerve fiber density in ELGs compared to controls. Functional assessments indicated a marked reduction in lacrimal secretion following cholinergic stimulation in LHFI-treated mice, suggesting impaired gland function. Overall, our results suggest that LHFI disrupts lacrimal gland homeostasis, potentially leading to dry eye disease by altering its structure and secretory function. These insights underscore the profound impact of dietary choices on ocular health and highlight the need for strategies to mitigate these risks.
Collapse
Affiliation(s)
- Di Qi
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Duliurui Huang
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Mengru Ba
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Shuting Xuan
- Department of Ophthalmology, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Hongli Si
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Wenxiao Zhang
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Zhijie Li
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China.
| |
Collapse
|
9
|
Savvidis C, Kallistrou E, Kouroglou E, Dionysopoulou S, Gavriiloglou G, Ragia D, Tsiama V, Proikaki S, Belis K, Ilias I. Circadian rhythm disruption and endocrine-related tumors. World J Clin Oncol 2024; 15:818-834. [PMID: 39071458 PMCID: PMC11271730 DOI: 10.5306/wjco.v15.i7.818] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
This review delved into the intricate relationship between circadian clocks and physiological processes, emphasizing their critical role in maintaining homeostasis. Orchestrated by interlocked clock genes, the circadian timekeeping system regulates fundamental processes like the sleep-wake cycle, energy metabolism, immune function, and cell proliferation. The central oscillator in the hypothalamic suprachiasmatic nucleus synchronizes with light-dark cycles, while peripheral tissue clocks are influenced by cues such as feeding times. Circadian disruption, linked to modern lifestyle factors like night shift work, correlates with adverse health outcomes, including metabolic syndrome, cardiovascular diseases, infections, and cancer. We explored the molecular mechanisms of circadian clock genes and their impact on metabolic disorders and cancer pathogenesis. Specific associations between circadian disruption and endocrine tumors, spanning breast, ovarian, testicular, prostate, thyroid, pituitary, and adrenal gland cancers, are highlighted. Shift work is associated with increased breast cancer risk, with PER genes influencing tumor progression and drug resistance. CLOCK gene expression correlates with cisplatin resistance in ovarian cancer, while factors like aging and intermittent fasting affect prostate cancer. Our review underscored the intricate interplay between circadian rhythms and cancer, involving the regulation of the cell cycle, DNA repair, metabolism, immune function, and the tumor microenvironment. We advocated for integrating biological timing into clinical considerations for personalized healthcare, proposing that understanding these connections could lead to novel therapeutic approaches. Evidence supports circadian rhythm-focused therapies, particularly chronotherapy, for treating endocrine tumors. Our review called for further research to uncover detailed connections between circadian clocks and cancer, providing essential insights for targeted treatments. We emphasized the importance of public health interventions to mitigate lifestyle-related circadian disruptions and underscored the critical role of circadian rhythms in disease mechanisms and therapeutic interventions.
Collapse
Affiliation(s)
- Christos Savvidis
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Efthymia Kallistrou
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Eleni Kouroglou
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Sofia Dionysopoulou
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | | | - Dimitra Ragia
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Vasiliki Tsiama
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Stella Proikaki
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Konstantinos Belis
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| | - Ioannis Ilias
- Department of Endocrinology, Hippocration General Hospital, Athens GR-11527, Greece
| |
Collapse
|
10
|
Han Y, Shon J, Kwon SY, Park YJ. Effects of Dietary Protein Intake Levels on Peripheral Circadian Rhythm in Mice. Int J Mol Sci 2024; 25:7373. [PMID: 39000480 PMCID: PMC11242084 DOI: 10.3390/ijms25137373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
The regulation of the circadian clock plays an important role in influencing physiological conditions. While it is reported that the timing and quantity of energy intake impact circadian regulation, the underlying mechanisms remain unclear. This study investigated the impact of dietary protein intake on peripheral clocks. Firstly, transcriptomic analysis was conducted to investigate molecular targets of low-protein intake. Secondly, mPer2::Luc knock-in mice, fed with either a low-protein, normal, or high-protein diet for 6 weeks, were analyzed for the oscillation of PER2 expression in peripheral tissues and for the expression profiles of circadian and metabolic genes. Lastly, the candidate pathway identified by the in vivo analysis was validated using AML12 cells. As a result, using transcriptomic analysis, we found that the low-protein diet hardly altered the circadian rhythm in the central clock. In animal experiments, expression levels and period lengths of PER2 were different in peripheral tissues depending on dietary protein intake; moreover, mRNA levels of clock-controlled genes and endoplasmic reticulum (ER) stress genes were affected by dietary protein intake. Induction of ER stress in AML12 cells caused an increased amplitude of Clock and Bmal1 and an advanced peak phase of Per2. This result shows that the intake of different dietary protein ratios causes an alteration of the circadian rhythm, especially in the peripheral clock of mice. Dietary protein intake modifies the oscillation of ER stress genes, which may play key roles in the regulation of the circadian clock.
Collapse
Affiliation(s)
- Yerim Han
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jinyoung Shon
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| | - So Young Kwon
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yoon Jung Park
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Republic of Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
11
|
Usha Satheesan S, Chowdhury S, Kolthur-Seetharam U. Metabolic and circadian inputs encode anticipatory biogenesis of hepatic fed microRNAs. Life Sci Alliance 2024; 7:e202302180. [PMID: 38408795 PMCID: PMC10897495 DOI: 10.26508/lsa.202302180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 02/13/2024] [Accepted: 02/13/2024] [Indexed: 02/28/2024] Open
Abstract
Starvation and refeeding are mostly unanticipated in the wild in terms of duration, frequency, and nutritional value of the refed state. Notwithstanding this, organisms mount efficient and reproducible responses to restore metabolic homeostasis. Hence, it is intuitive to invoke expectant molecular mechanisms that build anticipatory responses to enable physiological toggling during fed-fast cycles. In this regard, we report anticipatory biogenesis of oscillatory hepatic microRNAs that peak during a fed state and inhibit starvation-responsive genes. Our results clearly demonstrate that the levels of primary and precursor microRNA transcripts increase during a fasting state, in anticipation of a fed response. We delineate the importance of both metabolic and circadian cues in orchestrating hepatic fed microRNA homeostasis in a physiological setting. Besides illustrating metabo-endocrine control, our findings provide a mechanistic basis for the overarching influence of starvation on anticipatory biogenesis. Importantly, by using pharmacological agents that are widely used in clinics, we point out the high potential of interventions to restore homeostasis of hepatic microRNAs, whose deregulated expression is otherwise well established to cause metabolic diseases.
Collapse
Affiliation(s)
- Sandra Usha Satheesan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Shreyam Chowdhury
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Ullas Kolthur-Seetharam
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
- Tata Institute of Fundamental Research- Hyderabad (TIFR-H), Hyderabad, India
| |
Collapse
|
12
|
Bass J. Interorgan rhythmicity as a feature of healthful metabolism. Cell Metab 2024; 36:655-669. [PMID: 38335957 PMCID: PMC10990795 DOI: 10.1016/j.cmet.2024.01.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 02/12/2024]
Abstract
The finding that animals with circadian gene mutations exhibit diet-induced obesity and metabolic syndrome with hypoinsulinemia revealed a distinct role for the clock in the brain and peripheral tissues. Obesogenic diets disrupt rhythmic sleep/wake patterns, feeding behavior, and transcriptional networks, showing that metabolic signals reciprocally control the clock. Providing access to high-fat diet only during the sleep phase (light period) in mice accelerates weight gain, whereas isocaloric time-restricted feeding during the active period enhances energy expenditure due to circadian induction of adipose thermogenesis. This perspective focuses on advances and unanswered questions in understanding the interorgan circadian control of healthful metabolism.
Collapse
Affiliation(s)
- Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
13
|
Vlachou D, Veretennikova M, Usselmann L, Vasilyev V, Ott S, Bjarnason GA, Dallmann R, Levi F, Rand DA. TimeTeller: A tool to probe the circadian clock as a multigene dynamical system. PLoS Comput Biol 2024; 20:e1011779. [PMID: 38422117 DOI: 10.1371/journal.pcbi.1011779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 03/12/2024] [Accepted: 12/21/2023] [Indexed: 03/02/2024] Open
Abstract
Recent studies have established that the circadian clock influences onset, progression and therapeutic outcomes in a number of diseases including cancer and heart diseases. Therefore, there is a need for tools to measure the functional state of the molecular circadian clock and its downstream targets in patients. Moreover, the clock is a multi-dimensional stochastic oscillator and there are few tools for analysing it as a noisy multigene dynamical system. In this paper we consider the methodology behind TimeTeller, a machine learning tool that analyses the clock as a noisy multigene dynamical system and aims to estimate circadian clock function from a single transcriptome by modelling the multi-dimensional state of the clock. We demonstrate its potential for clock systems assessment by applying it to mouse, baboon and human microarray and RNA-seq data and show how to visualise and quantify the global structure of the clock, quantitatively stratify individual transcriptomic samples by clock dysfunction and globally compare clocks across individuals, conditions and tissues thus highlighting its potential relevance for advancing circadian medicine.
Collapse
Affiliation(s)
- Denise Vlachou
- Mathematics Institute & Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, University of Warwick, Coventry, United Kingdom
| | - Maria Veretennikova
- Mathematics Institute & Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, University of Warwick, Coventry, United Kingdom
| | - Laura Usselmann
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Vadim Vasilyev
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Sascha Ott
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Georg A Bjarnason
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Robert Dallmann
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Francis Levi
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Department of Statistics, University of Warwick, Coventry, United Kingdom
- UPR "Chronotherapy, Cancer and Transplantation", Medical School, Paris-Saclay University, Medical Oncology Department, Paul Brousse Hospital, Villejuif, France
| | - David A Rand
- Mathematics Institute & Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
14
|
Brooks TG, Manjrekar A, Mrcˇela A, Grant GR. Meta-analysis of Diurnal Transcriptomics in Mouse Liver Reveals Low Repeatability of Rhythm Analyses. J Biol Rhythms 2023; 38:556-570. [PMID: 37382061 PMCID: PMC10615793 DOI: 10.1177/07487304231179600] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
To assess the consistency of biological rhythms across studies, 57 public mouse liver tissue timeseries totaling 1096 RNA-seq samples were obtained and analyzed. Only the control groups of each study were included, to create comparable data. Technical factors in RNA-seq library preparation were the largest contributors to transcriptome-level differences, beyond biological or experiment-specific factors such as lighting conditions. Core clock genes were remarkably consistent in phase across all studies. Overlap of genes identified as rhythmic across studies was generally low, with no pair of studies having over 60% overlap. Distributions of phases of significant genes were remarkably inconsistent across studies, but the genes that consistently identified as rhythmic had acrophase clustering near ZT0 and ZT12. Despite the discrepancies between single-study analyses, cross-study analyses found substantial consistency. Running compareRhythms on each pair of studies identified a median of only 11% of the identified rhythmic genes as rhythmic in only 1 of the 2 studies. Data were integrated across studies in a joint and individual variance estimate (JIVE) analysis, which showed that the top 2 components of joint within-study variation are determined by time of day. A shape-invariant model with random effects was fit to the genes to identify the underlying shape of the rhythms, consistent across all studies, including identifying 72 genes with consistently multiple peaks.
Collapse
Affiliation(s)
- Thomas G. Brooks
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Aditi Manjrekar
- Department of Neuroscience, The University of Texas at Dallas, Richardson, Texas
| | - Antonijo Mrcˇela
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gregory R. Grant
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
15
|
Nomura S, Hosono T, Ono M, Daikoku T, Michihiro M, Kagami K, Iizuka T, Chen Y, Shi Y, Morishige JI, Fujiwara T, Fujiwara H, Ando H. Desynchronization between Food Intake and Light Stimulations Induces Uterine Clock Quiescence in Female Mice. J Nutr 2023; 153:2283-2290. [PMID: 37336322 DOI: 10.1016/j.tjnut.2023.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/15/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Dysmenorrhea is associated with breakfast skipping in young women, suggesting that fasting in the early active phase disrupts uterine functions. OBJECTIVES To investigate the possible involvement of the uterine clock system in fasting-induced uterine dysfunction, we examined core clock gene expressions in the uterus using a 28-h interval-fed mouse model. METHODS Young female mice (8 wk of age) were divided into 3 groups: group I (ad libitum feeding), group II (time-restricted feeding, initial 4 h of the active period every day), and group III (time-restricted feeding for 8 h with a 28-h cycle). Groups II and III have the same fasting interval of 20 h. After analyzing feeding and wheel running behaviors during 2 wk of dietary restriction, mice were sacrificed at 4-h intervals, and the expression profiles of clock genes in the uterus and liver were examined by qPCR. RESULTS The mice in group I took food mainly during the dark phase and those in group II during the initial 4 h of the dark phase, whereas those in group III delayed feeding time by 4 h per cycle. In all groups, spontaneous wheel running was observed during the dark phase. There was no difference in the quantity of feeding and the amount of running exercise among the 3 groups during the second week. The mRNA expressions of peripheral clock genes, Bmal1, Clock, Per1, Per2, Cry1, Nr1d1, and Dbp and a clock-controlled gene, Fabp1, in the uterus showed rhythmic oscillations with normal sequential expression cascade in groups I and II, whereas their expressions decreased and circadian cycles disappeared in group III. In contrast, liver core clock genes in group III showed clear circadian cycles. CONCLUSIONS Fluctuations in the timing of the first food intake impair the uterine clock oscillator system to reduce clock gene expressions and abolish their circadian rhythms.
Collapse
Affiliation(s)
- Satoshi Nomura
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Takashi Hosono
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, Japan.
| | - Takiko Daikoku
- Division of Animal Disease Model, Research Center for Experimental Modeling of Human Disease, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Mieda Michihiro
- Department of Integrative Neurophysiology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Kyosuke Kagami
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Takashi Iizuka
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Yuchen Chen
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Yifan Shi
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Jun-Ichi Morishige
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Tomoko Fujiwara
- Department of Social Work and Life Design, Kyoto Notre Dame University, Kyoto, Japan
| | - Hiroshi Fujiwara
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan.
| | - Hitoshi Ando
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
16
|
Alkhoury C, Henneman NF, Petrenko V, Shibayama Y, Segaloni A, Gadault A, Nemazanyy I, Le Guillou E, Wolide AD, Antoniadou K, Tong X, Tamaru T, Ozawa T, Girard M, Hnia K, Lutter D, Dibner C, Panasyuk G. Class 3 PI3K coactivates the circadian clock to promote rhythmic de novo purine synthesis. Nat Cell Biol 2023; 25:975-988. [PMID: 37414850 PMCID: PMC10344785 DOI: 10.1038/s41556-023-01171-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 05/22/2023] [Indexed: 07/08/2023]
Abstract
Metabolic demands fluctuate rhythmically and rely on coordination between the circadian clock and nutrient-sensing signalling pathways, yet mechanisms of their interaction remain not fully understood. Surprisingly, we find that class 3 phosphatidylinositol-3-kinase (PI3K), known best for its essential role as a lipid kinase in endocytosis and lysosomal degradation by autophagy, has an overlooked nuclear function in gene transcription as a coactivator of the heterodimeric transcription factor and circadian driver Bmal1-Clock. Canonical pro-catabolic functions of class 3 PI3K in trafficking rely on the indispensable complex between the lipid kinase Vps34 and regulatory subunit Vps15. We demonstrate that although both subunits of class 3 PI3K interact with RNA polymerase II and co-localize with active transcription sites, exclusive loss of Vps15 in cells blunts the transcriptional activity of Bmal1-Clock. Thus, we establish non-redundancy between nuclear Vps34 and Vps15, reflected by the persistent nuclear pool of Vps15 in Vps34-depleted cells and the ability of Vps15 to coactivate Bmal1-Clock independently of its complex with Vps34. In physiology we find that Vps15 is required for metabolic rhythmicity in liver and, unexpectedly, it promotes pro-anabolic de novo purine nucleotide synthesis. We show that Vps15 activates the transcription of Ppat, a key enzyme for the production of inosine monophosphate, a central metabolic intermediate for purine synthesis. Finally, we demonstrate that in fasting, which represses clock transcriptional activity, Vps15 levels are decreased on the promoters of Bmal1 targets, Nr1d1 and Ppat. Our findings open avenues for establishing the complexity for nuclear class 3 PI3K signalling for temporal regulation of energy homeostasis.
Collapse
Affiliation(s)
- Chantal Alkhoury
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Nathaniel F Henneman
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Volodymyr Petrenko
- The Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| | - Yui Shibayama
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Arianna Segaloni
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Alexis Gadault
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS, UAR 3633, Paris, France
| | - Edouard Le Guillou
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Amare Desalegn Wolide
- Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München (TUM), Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Konstantina Antoniadou
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Xin Tong
- Department of Molecular and Integrative Physiology, Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Teruya Tamaru
- Department of Physiology, Toho University School of Medicine, Tokyo, Japan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo, Japan
| | - Muriel Girard
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Karim Hnia
- Institute of Cardiovascular and Metabolic Diseases (I2MC), INSERM-UMR 1297, University Paul Sabatier, Toulouse, France
| | - Dominik Lutter
- Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Charna Dibner
- The Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| | - Ganna Panasyuk
- Institut Necker-Enfants Malades (INEM), Paris, France.
- INSERM U1151/CNRS UMR 8253, Paris, France.
- Université Paris Cité, Paris, France.
| |
Collapse
|
17
|
Sato S, Hishida T, Kinouchi K, Hatanaka F, Li Y, Nguyen Q, Chen Y, Wang PH, Kessenbrock K, Li W, Izpisua Belmonte JC, Sassone-Corsi P. The circadian clock CRY1 regulates pluripotent stem cell identity and somatic cell reprogramming. Cell Rep 2023; 42:112590. [PMID: 37261952 DOI: 10.1016/j.celrep.2023.112590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 03/28/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Distinct metabolic conditions rewire circadian-clock-controlled signaling pathways leading to the de novo construction of signal transduction networks. However, it remains unclear whether metabolic hallmarks unique to pluripotent stem cells (PSCs) are connected to clock functions. Reprogramming somatic cells to a pluripotent state, here we highlighted non-canonical functions of the circadian repressor CRY1 specific to PSCs. Metabolic reprogramming, including AMPK inactivation and SREBP1 activation, was coupled with the accumulation of CRY1 in PSCs. Functional assays verified that CRY1 is required for the maintenance of self-renewal capacity, colony organization, and metabolic signatures. Genome-wide occupancy of CRY1 identified CRY1-regulatory genes enriched in development and differentiation in PSCs, albeit not somatic cells. Last, cells lacking CRY1 exhibit differential gene expression profiles during induced PSC (iPSC) reprogramming, resulting in impaired iPSC reprogramming efficiency. Collectively, these results suggest the functional implication of CRY1 in pluripotent reprogramming and ontogenesis, thereby dictating PSC identity.
Collapse
Affiliation(s)
- Shogo Sato
- Center for Epigenetics and Metabolism, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA; Center for Biological Clocks Research, Department of Biology, Texas A&M University, College Station, TX, USA.
| | - Tomoaki Hishida
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA; Laboratory of Biological Chemistry, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
| | - Kenichiro Kinouchi
- Center for Epigenetics and Metabolism, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Fumiaki Hatanaka
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA; Altos Labs, San Diego, CA, USA
| | - Yumei Li
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Quy Nguyen
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Yumay Chen
- UC Irvine Diabetes Center, Sue and Bill Gross Stem Cell Research Center, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Ping H Wang
- UC Irvine Diabetes Center, Sue and Bill Gross Stem Cell Research Center, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Wei Li
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA; Altos Labs, San Diego, CA, USA.
| | - Paolo Sassone-Corsi
- Center for Epigenetics and Metabolism, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
18
|
Jakubowicz D, Rosenblum RC, Wainstein J, Twito O. Influence of Fasting until Noon (Extended Postabsorptive State) on Clock Gene mRNA Expression and Regulation of Body Weight and Glucose Metabolism. Int J Mol Sci 2023; 24:ijms24087154. [PMID: 37108316 PMCID: PMC10138720 DOI: 10.3390/ijms24087154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
The trend of fasting until noon (omission or delayed breakfast) is increasingly prevalent in modern society. This eating pattern triggers discordance between endogenous circadian clock rhythms and the feeding/fasting cycle and is associated with an increased incidence of obesity and T2D. Although the underlying mechanism of this association is not well understood, growing evidence suggests that fasting until noon, also known as an "extended postabsorptive state", has the potential to cause a deleterious effect on clock gene expression and to disrupt regulation of body weight, postprandial and overall glycemia, skeletal muscle protein synthesis, and appetite, and may also lead to lower energy expenditure. This manuscript overviews the clock gene-controlled glucose metabolism during the active and resting phases and the consequences of postponing until noon the transition from postabsorptive to fed state on glucose metabolism, weight control, and energy expenditure. Finally, we will discuss the metabolic advantages of shifting more energy, carbohydrates (CH), and proteins to the early hours of the day.
Collapse
Affiliation(s)
- Daniela Jakubowicz
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Rachel Chava Rosenblum
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Julio Wainstein
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Orit Twito
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| |
Collapse
|
19
|
Malhan D, Yalçin M, Schoenrock B, Blottner D, Relógio A. Skeletal muscle gene expression dysregulation in long-term spaceflights and aging is clock-dependent. NPJ Microgravity 2023; 9:30. [PMID: 37012297 PMCID: PMC10070655 DOI: 10.1038/s41526-023-00273-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 03/13/2023] [Indexed: 04/05/2023] Open
Abstract
The circadian clock regulates cellular and molecular processes in mammals across all tissues including skeletal muscle, one of the largest organs in the human body. Dysregulated circadian rhythms are characteristic of aging and crewed spaceflight, associated with, for example, musculoskeletal atrophy. Molecular insights into spaceflight-related alterations of circadian regulation in skeletal muscle are still missing. Here, we investigated potential functional consequences of clock disruptions on skeletal muscle using published omics datasets obtained from spaceflights and other clock-altering, external (fasting and exercise), or internal (aging) conditions on Earth. Our analysis identified alterations of the clock network and skeletal muscle-associated pathways, as a result of spaceflight duration in mice, which resembles aging-related gene expression changes observed in humans on Earth (e.g., ATF4 downregulation, associated with muscle atrophy). Furthermore, according to our results, external factors such as exercise or fasting lead to molecular changes in the core-clock network, which may compensate for the circadian disruption observed during spaceflights. Thus, maintaining circadian functioning is crucial to ameliorate unphysiological alterations and musculoskeletal atrophy reported among astronauts.
Collapse
Affiliation(s)
- Deeksha Malhan
- Institute for Theoretical Biology (ITB), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, 20457, Germany
| | - Müge Yalçin
- Institute for Theoretical Biology (ITB), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, 20457, Germany
| | - Britt Schoenrock
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany
| | - Dieter Blottner
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany
- Neuromuscular System and Neuromuscular Signaling, Berlin Center of Space Medicine & Extreme Environments, Berlin, 10115, Germany
| | - Angela Relógio
- Institute for Theoretical Biology (ITB), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany.
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany.
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, 20457, Germany.
| |
Collapse
|
20
|
Tang D, Tang Q, Huang W, Zhang Y, Tian Y, Fu X. Fasting: From Physiology to Pathology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204487. [PMID: 36737846 PMCID: PMC10037992 DOI: 10.1002/advs.202204487] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/06/2023] [Indexed: 06/18/2023]
Abstract
Overnutrition is a risk factor for various human diseases, including neurodegenerative diseases, metabolic disorders, and cancers. Therefore, targeting overnutrition represents a simple but attractive strategy for the treatment of these increasing public health threats. Fasting as a dietary intervention for combating overnutrition has been extensively studied. Fasting has been practiced for millennia, but only recently have its roles in the molecular clock, gut microbiome, and tissue homeostasis and function emerged. Fasting can slow aging in most species and protect against various human diseases, including neurodegenerative diseases, metabolic disorders, and cancers. These centuried and unfading adventures and explorations suggest that fasting has the potential to delay aging and help prevent and treat diseases while minimizing side effects caused by chronic dietary interventions. In this review, recent animal and human studies concerning the role and underlying mechanism of fasting in physiology and pathology are summarized, the therapeutic potential of fasting is highlighted, and the combination of pharmacological intervention and fasting is discussed as a new treatment regimen for human diseases.
Collapse
Affiliation(s)
- Dongmei Tang
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuan610041China
| | - Qiuyan Tang
- Neurology Department of Integrated Traditional Chinese and Western Medicine, School of Clinical MedicineChengdu University of Traditional Chinese MedicineChengduSichuan610075China
| | - Wei Huang
- West China Centre of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineWest China‐Liverpool Biomedical Research CentreWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Yuwei Zhang
- Division of Endocrinology and MetabolismWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Yan Tian
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuan610041China
| | - Xianghui Fu
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuan610041China
| |
Collapse
|
21
|
Zhu WZ, He QY, Feng DC, Wei Q, Yang L. Circadian rhythm in prostate cancer: time to take notice of the clock. Asian J Androl 2023; 25:184-191. [PMID: 36073562 PMCID: PMC10069698 DOI: 10.4103/aja202255] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The circadian clock is an evolutionary molecular product that is associated with better adaptation to changes in the external environment. Disruption of the circadian rhythm plays a critical role in tumorigenesis of many kinds of cancers, including prostate cancer (PCa). Integrating circadian rhythm into PCa research not only brings a closer understanding of the mechanisms of PCa but also provides new and effective options for the precise treatment of patients with PCa. This review begins with patterns of the circadian clock, highlights the role of the disruption of circadian rhythms in PCa at the epidemiological and molecular levels, and discusses possible new approaches to PCa therapy that target the circadian clock.
Collapse
Affiliation(s)
- Wei-Zhen Zhu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qi-Ying He
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - De-Chao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lu Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
22
|
Marciniak M, Sato M, Rutkowski R, Zawada A, Juchacz A, Mahadea D, Grzymisławski M, Dobrowolska A, Kawka E, Korybalska K, Bręborowicz A, Witowski J, Kanikowska D. Effect of the one-day fasting on cortisol and DHEA daily rhythm regarding sex, chronotype, and age among obese adults. Front Nutr 2023; 10:1078508. [PMID: 36814510 PMCID: PMC9940638 DOI: 10.3389/fnut.2023.1078508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/10/2023] [Indexed: 02/10/2023] Open
Abstract
Introduction Physiological and biochemical processes in the human body occur in a specific order and show rhythmic variability. Time dependence characterizes the secretion of cortisol and dehydroepiandrosterone (DHEA). One-day fasting implies alternating fasting days and eating days. The study aimed to determine how 24-h fasting affects the daily rhythm of cortisol and DHEA levels in obese people while taking into account gender and chronotype. Methods Forty-nine obese patients (BMI 32.2-67.1 kg/m2; 25 women and 24 men) underwent a 3-week hospital-controlled calorie restriction diet to reduce body weight. During hospitalization, patients fasted for 1 day, during which only water could be consumed. Samples of whole mixed unstimulated saliva were collected at 2-3-h intervals over a 64-h period and analyzed for cortisol and DHEA by immunoassays. The individual chronotypes were assessed by the morning and evening questionnaire, according to Horne and Östberg. Three components of daily rhythm were evaluated: amplitude, acrophase, and the so-called MESOR. Results Cortisol rhythm showed differences in amplitude (p = 0.0127) and acrophase (p = 0.0005). The amplitude on the fasting day was 11% higher (p = 0.224) than the day after. The acrophase advanced on the day of fasting, 48 min earlier than the day before (p = 0.0064), and by 39 min to the day after fasting (p = 0.0005). In the rhythm of DHEA, differences were found in the MESOR (p = 0.0381). The MESOR on the fasting day increased. Discussion Our results obtained during 64 consecutive hours of saliva sampling suggest that one-day fasting may affect three components of cortisol and DHEA daily rhythm. Additionally, no differences were found in the daily rhythm between the morning and evening chronotypes and between females and males. Although aging did not influence daily cortisol rhythm, DHEA amplitude, MESOR, and acrophase changed with age. To the best of our knowledge, this is the first presentation of changes in DHEA rhythm during one-day fasting.
Collapse
Affiliation(s)
- Martyna Marciniak
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland,Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Science, Poznan, Poland
| | - Maki Sato
- Institutional Research, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Rafał Rutkowski
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Agnieszka Zawada
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Science, Poznan, Poland
| | - Aldona Juchacz
- Greater Poland Center of Pulmonology and Thoracic Surgery of Eugenia and Janusz Zeyland, Poznan, Poland
| | - Dagmara Mahadea
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Science, Poznan, Poland
| | - Marian Grzymisławski
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Science, Poznan, Poland
| | - Agnieszka Dobrowolska
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Science, Poznan, Poland
| | - Edyta Kawka
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Korybalska
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Andrzej Bręborowicz
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland,Collegium Medicum, Zielona Góra University, Zielona Góra, Poland
| | - Janusz Witowski
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Dominika Kanikowska
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland,*Correspondence: Dominika Kanikowska,
| |
Collapse
|
23
|
Kavanagh K, Bashore AC, Davis MA, Jorgensen MJ, McClouth CJ, Beavers DA, Parks JS. Early time-restricted feeding improves high-density lipoprotein amount and function in nonhuman primates, without effects on body composition. Obesity (Silver Spring) 2023; 31 Suppl 1:75-84. [PMID: 36229981 PMCID: PMC9877107 DOI: 10.1002/oby.23564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/04/2022] [Accepted: 08/01/2022] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Time-restricted feeding (TRF), whereby caloric intake is limited to a <12-hour window, is a potential regimen to ameliorate metabolic syndrome and cardiovascular disease (CVD) risk co-occurring with aging and with obesity. Early TRF (eTRF; early morning feeding followed by overnight fasting) times calorie consumption with hepatic circadian gene expression rhythms. Brief TRF trials demonstrate that high-density lipoprotein (HDL) cholesterol increases similar to diet/exercise interventions, which may impart beneficial CVD effects. Using a nonhuman primate (NHP) model, the efficacy of eTRF to raise HDL and increase plasma cholesterol efflux capacity (CEC) (primarily mediated by cholesterol efflux to HDL particles, a process that is inversely associated with CVD risk) was examined. METHODS Adult (8-16 years old, n = 25) and geriatric (≥17 years old) NHPs were randomized to ad libitum feeding or eTRF for 12 months, and relevant body composition, glycemic control, and plasma HDL cholesterol levels and CEC were measured. RESULTS Impaired CEC was found in geriatric NHPs. eTRF induced larger-sized HDL particles, increased HDL apolipoprotein A-1 content, lowered triglyceride concentrations, and increased plasma CEC (primarily to HDL particles) in both adult and geriatric NHPs without changes in glycemic control or body composition. CONCLUSIONS A beneficial effect of eTRF on increasing HDL CEC in NHPs was demonstrated.
Collapse
Affiliation(s)
- Kylie Kavanagh
- Department of PathologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
- College of Health and MedicineUniversity of TasmaniaHobartAustralia
- Department of Internal Medicine‐Section on Molecular MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Alexander C. Bashore
- Department of Internal Medicine‐Section on Molecular MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Matthew A. Davis
- Department of Internal Medicine‐Section on Molecular MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Matthew J. Jorgensen
- Department of PathologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Christopher J. McClouth
- Department of Public Health SciencesWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Daniel A. Beavers
- Department of Public Health SciencesWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - John S. Parks
- Department of Internal Medicine‐Section on Molecular MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
24
|
Abstract
Biomedical research on mammals has traditionally neglected females, raising the concern that some scientific findings may generalize poorly to half the population. Although this lack of sex inclusion has been broadly documented, its extent within circadian genomics remains undescribed. To address this gap, we examined sex inclusion practices in a comprehensive collection of publicly available transcriptome studies on daily rhythms. Among 148 studies having samples from mammals in vivo, we found strong underrepresentation of females across organisms and tissues. Overall, only 23 of 123 studies in mice, 0 of 10 studies in rats, and 9 of 15 studies in humans included samples from females. In addition, studies having samples from both sexes tended to have more samples from males than from females. These trends appear to have changed little over time, including since 2016, when the US National Institutes of Health began requiring investigators to consider sex as a biological variable. Our findings highlight an opportunity to dramatically improve representation of females in circadian research and to explore sex differences in daily rhythms at the genome level.
Collapse
Affiliation(s)
- Dora Obodo
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee,Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Elliot H. Outland
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jacob J. Hughey
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee,Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, Tennessee,Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee,Jacob J. Hughey, Department of Biomedical Informatics, Vanderbilt University Medical Center, 2525 West End Ave., Suite 1475, Nashville, TN 37232, USA; e-mail:
| |
Collapse
|
25
|
Cornuti S, Chen S, Lupori L, Finamore F, Carli F, Samad M, Fenizia S, Caldarelli M, Damiani F, Raimondi F, Mazziotti R, Magnan C, Rocchiccioli S, Gastaldelli A, Baldi P, Tognini P. Brain histone beta-hydroxybutyrylation couples metabolism with gene expression. Cell Mol Life Sci 2023; 80:28. [PMID: 36607453 PMCID: PMC11072080 DOI: 10.1007/s00018-022-04673-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023]
Abstract
Little is known about the impact of metabolic stimuli on brain tissue at a molecular level. The ketone body beta-hydroxybutyrate (BHB) can be a signaling molecule regulating gene transcription. Thus, we assessed lysine beta-hydroxybutyrylation (K-bhb) levels in proteins extracted from the cerebral cortex of mice undergoing a ketogenic metabolic challenge (48 h fasting). We found that fasting enhanced K-bhb in a variety of proteins including histone H3. ChIP-seq experiments showed that K9 beta-hydroxybutyrylation of H3 (H3K9-bhb) was significantly enriched by fasting on more than 8000 DNA loci. Transcriptomic analysis showed that H3K9-bhb on enhancers and promoters correlated with active gene expression. One of the most enriched functional annotations both at the epigenetic and transcriptional level was "circadian rhythms''. Indeed, we found that the diurnal oscillation of specific transcripts was modulated by fasting at distinct zeitgeber times both in the cortex and suprachiasmatic nucleus. Moreover, specific changes in locomotor activity daily features were observed during re-feeding after 48-h fasting. Thus, our results suggest that fasting remarkably impinges on the cerebral cortex transcriptional and epigenetic landscape, and BHB acts as a powerful epigenetic molecule in the brain through direct and specific histone marks remodeling in neural tissue cells.
Collapse
Affiliation(s)
- Sara Cornuti
- Bio@SNS Lab, Scuola Normale Superiore, Pisa, Italy
| | - Siwei Chen
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA, USA
| | | | - Francesco Finamore
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Fabrizia Carli
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Muntaha Samad
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA, USA
| | - Simona Fenizia
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Matteo Caldarelli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | | | - Raffaele Mazziotti
- Institute of Neuroscience, National Research Council, Pisa, Italy
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, Calambrone, Pisa, Italy
| | - Christophe Magnan
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA, USA
| | | | - Amalia Gastaldelli
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Pierre Baldi
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA, USA
| | - Paola Tognini
- Bio@SNS Lab, Scuola Normale Superiore, Pisa, Italy.
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.
| |
Collapse
|
26
|
The circadian rhythm regulates branched-chain amino acids metabolism in fast muscle of Chinese perch ( Siniperca chuatsi) during short-term fasting by Clock-KLF15-Bcat2 pathway. Br J Nutr 2022:1-12. [PMID: 36373572 DOI: 10.1017/s0007114522003646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
As an internal time-keeping mechanism, circadian rhythm plays crucial role in maintaining homoeostasis when in response to nutrition change; meanwhile, branched-chain amino acids (BCAA) in skeletal muscle play an important role in preserving energy homoeostasis during fasting. Previous results from our laboratory suggested that fasting can influence peripheral circadian rhythm and BCAA metabolism in fish, but the relationship between circadian rhythm and BCAA metabolism, and whether circadian rhythm regulates BCAA metabolism to maintain physiological homoeostasis during fasting remains unclear. This study shows that the expression of fifteen core clock genes as well as KLF15 and Bcat2 is highly responsive to short-term fasting in fast muscle of Siniperca chuatsi, and the correlation coefficient between Clock and KLF15 expression is enhanced after fasting treatment. Furthermore, we demonstrate that the transcriptional expression of KLF15 is regulated by Clock, and the transcriptional expression of Bcat2 is regulated by KLF15 by using dual-luciferase reporter gene assay and Vivo-morpholinos-mediated gene knockdown technique. Therefore, fasting imposes a dynamic coordination of transcription between the circadian rhythm and BCAA metabolic pathways. The findings highlight the interaction between circadian rhythm and BCAA metabolism and suggest that fasting induces a switch in KLF15 expression through affecting the rhythmic expression of Clock, and then KLF15 promotes the transcription of Bcat2 to enhance the metabolism of BCAA, thus maintaining energy homoeostasis and providing energy for skeletal muscle as well as other tissues.
Collapse
|
27
|
Bideyan L, López Rodríguez M, Priest C, Kennelly JP, Gao Y, Ferrari A, Rajbhandari P, Feng AC, Tevosian SG, Smale ST, Tontonoz P. Hepatic GATA4 regulates cholesterol and triglyceride homeostasis in collaboration with LXRs. Genes Dev 2022; 36:1129-1144. [PMID: 36522129 PMCID: PMC9851399 DOI: 10.1101/gad.350145.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/02/2022] [Indexed: 12/16/2022]
Abstract
GATA4 is a transcription factor known for its crucial role in the development of many tissues, including the liver; however, its role in adult liver metabolism is unknown. Here, using high-throughput sequencing technologies, we identified GATA4 as a transcriptional regulator of metabolism in the liver. GATA4 expression is elevated in response to refeeding, and its occupancy is increased at enhancers of genes linked to fatty acid and lipoprotein metabolism. Knocking out GATA4 in the adult liver (Gata4LKO) decreased transcriptional activity at GATA4 binding sites, especially during feeding. Gata4LKO mice have reduced plasma HDL cholesterol and increased liver triglyceride levels. The expression of a panel of GATA4 binding genes involved in hepatic cholesterol export and triglyceride hydrolysis was down-regulated in Gata4LKO mice. We further demonstrate that GATA4 collaborates with LXR nuclear receptors in the liver. GATA4 and LXRs share a number of binding sites, and GATA4 was required for the full transcriptional response to LXR activation. Collectively, these results show that hepatic GATA4 contributes to the transcriptional control of hepatic and systemic lipid homeostasis.
Collapse
Affiliation(s)
- Lara Bideyan
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, USA
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, USA
| | - Maykel López Rodríguez
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, USA
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, USA
| | - Christina Priest
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, USA
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, USA
| | - John P Kennelly
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, USA
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, USA
| | - Yajing Gao
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, USA
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, USA
| | - Alessandra Ferrari
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, USA
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, USA
| | - Prashant Rajbhandari
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, USA
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, USA
| | - An-Chieh Feng
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, USA
| | - Sergei G Tevosian
- Department of Physiological Sciences, University of Florida, Gainesville, Florida 32610, USA
| | - Stephen T Smale
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, USA
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, USA
| |
Collapse
|
28
|
Miao L, Batty KR, Jackson AN, Pieno HA, Rhoades MW, Kojima S. Genetic and environmental perturbations alter the rhythmic expression pattern of a circadian long non-coding RNA, Per2AS, in mouse liver. F1000Res 2022; 11:1073. [PMID: 36250003 PMCID: PMC9551389 DOI: 10.12688/f1000research.125628.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/26/2022] [Indexed: 11/23/2022] Open
Abstract
Background: Long non-coding RNAs (lncRNAs) play a wide variety of biological roles without encoding a protein. Although the functions of many lncRNAs have been uncovered in recent years, the regulatory mechanism of lncRNA expression is still poorly understood despite that the expression patterns of lncRNAs are much more specific compared to mRNAs. Here, we investigated the rhythmic expression of Per2AS, a novel lncRNA that regulates circadian rhythms. Given that Per2AS expression is antiphasic to Period2 ( Per2), a core circadian clock gene, and transcribed from the antisense strand of Per2, we hypothesized that the rhythmic Per2AS expression is driven either by its own promoter or by the rhythmic Per2 transcription via transcriptional interference. Methods: We leveraged existing circadian RNA-seq datasets and analyzed the expression patterns of Per2AS and Per2 in response to the genetic or environmental disruption of the circadian rhythm in mouse liver. We tested our hypotheses by comparing the changes in the expression patterns of Per2AS and Per2. Conclusions: We found that, in some cases, Per2AS expression is independently controlled by other circadian transcription factors. In other cases, the pattern of expression change is consistent with both transcriptional interference and independent regulation hypotheses. Although additional experiments will be necessary to distinguish these possibilities, findings from this work contribute to a deeper understanding of the mechanism of how the expression of lncRNA is regulated.
Collapse
Affiliation(s)
- Lin Miao
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061, USA,Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Kyle R. Batty
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, 24061, USA,Division of Systems Biology, Academy of Integrated Science, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Ayana N. Jackson
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061, USA,Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Heather A. Pieno
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Maisy W. Rhoades
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Shihoko Kojima
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061, USA,Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, 24061, USA,Division of Systems Biology, Academy of Integrated Science, Virginia Tech, Blacksburg, VA, 24061, USA,
| |
Collapse
|
29
|
Petrus P, Cervantes M, Samad M, Sato T, Chao A, Sato S, Koronowski KB, Park G, Alam Y, Mejhert N, Seldin MM, Monroy Kuhn JM, Dyar KA, Lutter D, Baldi P, Kaiser P, Jang C, Sassone-Corsi P. Tryptophan metabolism is a physiological integrator regulating circadian rhythms. Mol Metab 2022; 64:101556. [PMID: 35914650 PMCID: PMC9382333 DOI: 10.1016/j.molmet.2022.101556] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/15/2022] [Accepted: 07/15/2022] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE The circadian clock aligns physiology with the 24-hour rotation of Earth. Light and food are the main environmental cues (zeitgebers) regulating circadian rhythms in mammals. Yet, little is known about the interaction between specific dietary components and light in coordinating circadian homeostasis. Herein, we focused on the role of essential amino acids. METHODS Mice were fed diets depleted of specific essential amino acids and their behavioral rhythms were monitored and tryptophan was selected for downstream analyses. The role of tryptophan metabolism in modulating circadian homeostasis was studied using isotope tracing as well as transcriptomic- and metabolomic- analyses. RESULTS Dietary tryptophan depletion alters behavioral rhythms in mice. Furthermore, tryptophan metabolism was shown to be regulated in a time- and light- dependent manner. A multi-omics approach and combinatory diet/light interventions demonstrated that tryptophan metabolism modulates temporal regulation of metabolism and transcription programs by buffering photic cues. Specifically, tryptophan metabolites regulate central circadian functions of the suprachiasmatic nucleus and the core clock machinery in the liver. CONCLUSIONS Tryptophan metabolism is a modulator of circadian homeostasis by integrating environmental cues. Our findings propose tryptophan metabolism as a potential point for pharmacologic intervention to modulate phenotypes associated with disrupted circadian rhythms.
Collapse
Affiliation(s)
- Paul Petrus
- Center for Epigenetics and Metabolism, U1233 INSERM, Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA.
| | - Marlene Cervantes
- Center for Epigenetics and Metabolism, U1233 INSERM, Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA.
| | - Muntaha Samad
- Institute for Genomics and Bioinformatics, Department of Computer Science, University of California Irvine (UCI), Irvine, CA, USA
| | - Tomoki Sato
- Center for Epigenetics and Metabolism, U1233 INSERM, Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Alina Chao
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92697, USA
| | - Shogo Sato
- Center for Epigenetics and Metabolism, U1233 INSERM, Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Kevin B Koronowski
- Center for Epigenetics and Metabolism, U1233 INSERM, Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Grace Park
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92697, USA
| | - Yasmine Alam
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92697, USA
| | - Niklas Mejhert
- Department of Medicine (H7), Karolinska Institutet, C2-94, Karolinska University Hospital, 141 86 Stockholm, Sweden
| | - Marcus M Seldin
- Center for Epigenetics and Metabolism, Department of Biological Chemistry, University of California, Irvine, CA, USA
| | - José Manuel Monroy Kuhn
- Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum Munich - German Research Center for Environmental Health, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Kenneth A Dyar
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Metabolic Physiology, Institute for Diabetes and Cancer (IDC), Helmholtz Diabetes Center, Helmholtz Zentrum Munich - German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Dominik Lutter
- Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum Munich - German Research Center for Environmental Health, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Pierre Baldi
- Institute for Genomics and Bioinformatics, Department of Computer Science, University of California Irvine (UCI), Irvine, CA, USA
| | - Peter Kaiser
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92697, USA
| | - Paolo Sassone-Corsi
- Center for Epigenetics and Metabolism, U1233 INSERM, Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
30
|
Wei F, Gong L, Lu S, Zhou Y, Liu L, Duan Z, Xiang R, Gonzalez FJ, Li G. Circadian transcriptional pathway atlas highlights a proteasome switch in intermittent fasting. Cell Rep 2022; 41:111547. [PMID: 36288692 PMCID: PMC9671760 DOI: 10.1016/j.celrep.2022.111547] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 08/11/2022] [Accepted: 09/30/2022] [Indexed: 12/01/2022] Open
Abstract
While intermittent fasting is a safe strategy to benefit health, it remains unclear whether a “timer” exists in vivo to record fasting duration and trigger a transcriptional switch. Here, we map a circadian transcriptional pathway atlas from 600 samples across four metabolic tissues of mice under five feeding regimens. Results show that 95.6% of detected canonical pathways are rhythmic in a tissue-specific and feeding-regimen-specific manner, while only less than 25% of them induce changes in transcriptional function. Fasting for 16 h initiates a circadian resonance of 43 pathways in the liver, and the resonance punctually switches following refeeding. The hepatic proteasome coordinates the resonance, and most genes encoding proteasome subunits display a 16-h fasting-dependent transcriptional switch. These findings indicate that the hepatic proteasome may serve as a fasting timer and a coordinator of pathway transcriptional resonance, which provide a target for revealing the underlying mechanism of intermittent fasting. While intermittent fasting benefits health, the optimal duration of each fasting remains an open question. Wei et al. map an atlas of canonical pathways in intermittent fasting, find that fasting for 16 h initiates circadian resonance of pathways in the liver, and identify the proteasome as a liver-specific fasting “timer”.
Collapse
Affiliation(s)
- Fang Wei
- Center for Biomedical Aging, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Lijun Gong
- Center for Biomedical Aging, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Siyu Lu
- Center for Biomedical Aging, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Yiming Zhou
- Center for Biomedical Aging, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Li Liu
- Center for Biomedical Aging, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Zhigui Duan
- Center for Biomedical Aging, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Rong Xiang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 41001, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Guolin Li
- Center for Biomedical Aging, National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, Hunan 410081, China.
| |
Collapse
|
31
|
Chu Y, Li J, Feng L, Zhang G, Wu H, Jiang T, Wang H, Feng J. Comparative analysis of the daily liver transcriptomes in wild nocturnal bats. BMC Genomics 2022; 23:572. [PMID: 35948882 PMCID: PMC9367025 DOI: 10.1186/s12864-022-08823-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 08/03/2022] [Indexed: 11/15/2022] Open
Abstract
Background Mammals rely on the circadian clock network to regulate daily systemic metabolism and physiological activities. The liver is an important peripheral organ in mammals, and it has a unique circadian rhythm regulation process. As the only mammals that can fly, bats have attracted much research attention due to their nocturnal habits and life histories. However, few research reports exist concerning the circadian rhythms of bat liver gene expression and the relevant biological clock regulation mechanisms in the liver. Results In this study, the expression levels of liver genes of Asian particolored bats were comparatively analyzed using RNA-seq at four different time points across 24 h. A total of 996 genes were found to be rhythmic, accounting for 65% of the total number of expressed genes. The critical circadian rhythm genes Bmal1, Rev-erbα, Cry, and Ror in the liver exhibited different expression patterns throughout the day, and participated in physiological processes with rhythmic changes, including Th17 cell differentiation (ko04659), antigen processing and presentation (ko04612), the estrogen signaling pathway (ko04915), and insulin resistance (ko04931). In addition, previous studies have found that the peroxisome proliferator-activated receptor (PPAR) metabolic signaling pathway (ko03320) may play a vital role in the rhythmic regulation of the metabolic network. Conclusions This study is the first to demonstrate diurnal changes in bat liver gene expression and related physiological processes. The results have thus further enriched our understanding of bats’ biological clocks. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08823-y.
Collapse
Affiliation(s)
- Yujia Chu
- College of Life Science, Jilin Agricultural University, Changchun, 130118, China
| | - Jingjing Li
- College of Life Science, Jilin Agricultural University, Changchun, 130118, China
| | - Lei Feng
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, 130117, China
| | - Guoting Zhang
- College of Life Science, Jilin Agricultural University, Changchun, 130118, China
| | - Hui Wu
- College of Life Science, Jilin Agricultural University, Changchun, 130118, China
| | - Tinglei Jiang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, 130117, China
| | - Hui Wang
- College of Life Science, Jilin Agricultural University, Changchun, 130118, China.
| | - Jiang Feng
- College of Life Science, Jilin Agricultural University, Changchun, 130118, China. .,Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, 130117, China.
| |
Collapse
|
32
|
ISX-9 potentiates CaMKIIδ-mediated BMAL1 activation to enhance circadian amplitude. Commun Biol 2022; 5:750. [PMID: 35902736 PMCID: PMC9334596 DOI: 10.1038/s42003-022-03725-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 07/15/2022] [Indexed: 11/09/2022] Open
Abstract
Circadian dysregulation associates with numerous diseases including metabolic dysfunction, sleep disorder, depression and aging. Given that declined circadian amplitude is a trait commonly found with compromised health, interventions that design in precluding circadian amplitude from dampening will aid to mitigate complex, circadian-related diseases. Here we identify a neurogenic small molecule ISX-9 that is able to support persistent and higher amplitude of circadian oscillations. ISX-9 improves diurnal metabolic rhythms in middle-aged mice. Moreover, the ISX-9-treated mice show better sleep homeostasis with increased delta power during the day time and higher locomotive activity in the dark period. ISX-9 augments CaMKIIδ expression and increases BMAL1 activity via eliciting CaMKIIδ-mediated phosphorylation on BMAL1 residues S513/S515/S516, accordingly composes a positive feedback effect on enhancing circadian amplitude. CaMKIIδ-targeting, and the use of ISX-9 may serve as decent choices for treating circadian-related disorders.
Collapse
|
33
|
Pickel L, Lee JH, Maughan H, Shi IQ, Verma N, Yeung C, Guttman D, Sung H. Circadian rhythms in metabolic organs and the microbiota during acute fasting in mice. Physiol Rep 2022; 10:e15393. [PMID: 35851583 PMCID: PMC9295129 DOI: 10.14814/phy2.15393] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 04/16/2023] Open
Abstract
The circadian clock regulates metabolism in anticipation of regular changes in the environment. It is found throughout the body, including in key metabolic organs such as the liver, adipose tissues, and intestine, where the timing of the clock is set largely by nutrient signaling. However, the circadian clocks of these tissues during the fasted state have not been completely characterized. Moreover, the sufficiency of a functioning host clock to produce diurnal rhythms in the composition of the microbiome in fasted animals has not been explored. To this end, mice were fasted 24 h prior to collection of key metabolic tissues and fecal samples for the analysis of circadian clock gene expression and microbiome composition. Rhythm characteristics were determined using CircaCompare software. We identify tissue-specific changes to circadian clock rhythms upon fasting, particularly in the brown adipose tissue, and for the first time demonstrate the rhythmicity of the microbiome in fasted animals.
Collapse
Affiliation(s)
- Lauren Pickel
- Translational Medicine Program, The Hospital for Sick ChildrenTorontoOntarioUSA
| | - Ju Hee Lee
- Translational Medicine Program, The Hospital for Sick ChildrenTorontoOntarioUSA
- Department of Laboratory Medicine and PathologyUniversity of TorontoTorontoOntarioUSA
| | | | - Irisa Qianwen Shi
- Translational Medicine Program, The Hospital for Sick ChildrenTorontoOntarioUSA
| | - Navkiran Verma
- Translational Medicine Program, The Hospital for Sick ChildrenTorontoOntarioUSA
- Department of Laboratory Medicine and PathologyUniversity of TorontoTorontoOntarioUSA
| | - Christy Yeung
- Translational Medicine Program, The Hospital for Sick ChildrenTorontoOntarioUSA
- Department of Laboratory Medicine and PathologyUniversity of TorontoTorontoOntarioUSA
| | - David Guttman
- Centre for the Analysis of Genome Evolution & FunctionUniversity of TorontoTorontoOntarioUSA
| | - Hoon‐Ki Sung
- Translational Medicine Program, The Hospital for Sick ChildrenTorontoOntarioUSA
- Department of Laboratory Medicine and PathologyUniversity of TorontoTorontoOntarioUSA
| |
Collapse
|
34
|
Fan L, Sweet DR, Fan EK, Prosdocimo DA, Madera A, Jiang Z, Padmanabhan R, Haldar SM, Vinayachandran V, Jain MK. Transcription factors KLF15 and PPARδ cooperatively orchestrate genome-wide regulation of lipid metabolism in skeletal muscle. J Biol Chem 2022; 298:101926. [PMID: 35413288 PMCID: PMC9190004 DOI: 10.1016/j.jbc.2022.101926] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022] Open
Abstract
Skeletal muscle dynamically regulates systemic nutrient homeostasis through transcriptional adaptations to physiological cues. In response to changes in the metabolic environment (e.g., alterations in circulating glucose or lipid levels), networks of transcription factors and coregulators are recruited to specific genomic loci to fine-tune homeostatic gene regulation. Elucidating these mechanisms is of particular interest as these gene regulatory pathways can serve as potential targets to treat metabolic disease. The zinc-finger transcription factor Krüppel-like factor 15 (KLF15) is a critical regulator of metabolic homeostasis; however, its genome-wide distribution in skeletal muscle has not been previously identified. Here, we characterize the KLF15 cistrome in vivo in skeletal muscle and find that the majority of KLF15 binding is localized to distal intergenic regions and associated with genes related to circadian rhythmicity and lipid metabolism. We also identify critical interdependence between KLF15 and the nuclear receptor PPARδ in the regulation of lipid metabolic gene programs. We further demonstrate that KLF15 and PPARδ colocalize genome-wide, physically interact, and are dependent on one another to exert their transcriptional effects on target genes. These findings reveal that skeletal muscle KLF15 plays a critical role in metabolic adaptation through its direct actions on target genes and interactions with other nodal transcription factors such as PPARδ.
Collapse
Affiliation(s)
- Liyan Fan
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA; Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - David R Sweet
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA; Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Erica K Fan
- University of Pittsburgh School of Medicine, Department of Physical Medicine and Rehabilitation, Pittsburgh, Pennsylvania, USA
| | - Domenick A Prosdocimo
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA; The Webb Law Firm, Pittsburgh, Pennsylvania, USA
| | - Annmarie Madera
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Zhen Jiang
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Roshan Padmanabhan
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Saptarsi M Haldar
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA; Department of Medicine, Division of Cardiology, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Vinesh Vinayachandran
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA.
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Case Western Reserve University, and Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA.
| |
Collapse
|
35
|
Chambers L, Seidler K, Barrow M. Nutritional entrainment of circadian rhythms under alignment and misalignment: a mechanistic review. Clin Nutr ESPEN 2022; 51:50-71. [DOI: 10.1016/j.clnesp.2022.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 06/20/2022] [Indexed: 10/17/2022]
|
36
|
Goldberg D, Charni-Natan M, Buchshtab N, Bar-Shimon M, Goldstein I. Hormone-controlled cooperative binding of transcription factors drives synergistic induction of fasting-regulated genes. Nucleic Acids Res 2022; 50:5528-5544. [PMID: 35556130 PMCID: PMC9177981 DOI: 10.1093/nar/gkac358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/22/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
During fasting, hepatocytes produce glucose in response to hormonal signals. Glucagon and glucocorticoids are principal fasting hormones that cooperate in regulating glucose production via gluconeogenesis. However, how these hormone signals are integrated and interpreted to a biological output is unknown. Here, we use genome-wide profiling of gene expression, enhancer dynamics and transcription factor (TF) binding in primary mouse hepatocytes to uncover the mode of cooperation between glucagon and glucocorticoids. We found that compared to a single treatment with each hormone, a dual treatment directs hepatocytes to a pro-gluconeogenic gene program by synergistically inducing gluconeogenic genes. The cooperative mechanism driving synergistic gene expression is based on ‘assisted loading’ whereby a glucagon-activated TF (cAMP responsive element binding protein; CREB) leads to enhancer activation which facilitates binding of the glucocorticoid receptor (GR) upon glucocorticoid stimulation. Glucagon does not only activate single enhancers but also activates enhancer clusters, thereby assisting the loading of GR also across enhancer units within the cluster. In summary, we show that cells integrate extracellular signals by an enhancer-specific mechanism: one hormone-activated TF activates enhancers, thereby assisting the loading of a TF stimulated by a second hormone, leading to synergistic gene induction and a tailored transcriptional response to fasting.
Collapse
Affiliation(s)
- Dana Goldberg
- Institute of Biochemistry, Food Science and Nutrition. The Robert H. Smith Faculty of Agriculture, Food and Environment. The Hebrew University of Jerusalem. POB 12, Rehovot 7610001, Israel
| | - Meital Charni-Natan
- Institute of Biochemistry, Food Science and Nutrition. The Robert H. Smith Faculty of Agriculture, Food and Environment. The Hebrew University of Jerusalem. POB 12, Rehovot 7610001, Israel
| | - Nufar Buchshtab
- Institute of Biochemistry, Food Science and Nutrition. The Robert H. Smith Faculty of Agriculture, Food and Environment. The Hebrew University of Jerusalem. POB 12, Rehovot 7610001, Israel
| | - Meirav Bar-Shimon
- Institute of Biochemistry, Food Science and Nutrition. The Robert H. Smith Faculty of Agriculture, Food and Environment. The Hebrew University of Jerusalem. POB 12, Rehovot 7610001, Israel
| | - Ido Goldstein
- Institute of Biochemistry, Food Science and Nutrition. The Robert H. Smith Faculty of Agriculture, Food and Environment. The Hebrew University of Jerusalem. POB 12, Rehovot 7610001, Israel
| |
Collapse
|
37
|
Sato T, Sassone-Corsi P. Nutrition, metabolism, and epigenetics: pathways of circadian reprogramming. EMBO Rep 2022; 23:e52412. [PMID: 35412705 PMCID: PMC9066069 DOI: 10.15252/embr.202152412] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 10/28/2021] [Accepted: 03/30/2022] [Indexed: 01/07/2023] Open
Abstract
Food intake profoundly affects systemic physiology. A large body of evidence has indicated a link between food intake and circadian rhythms, and ~24-h cycles are deemed essential for adapting internal homeostasis to the external environment. Circadian rhythms are controlled by the biological clock, a molecular system remarkably conserved throughout evolution. The circadian clock controls the cyclic expression of numerous genes, a regulatory program common to all mammalian cells, which may lead to various metabolic and physiological disturbances if hindered. Although the circadian clock regulates multiple metabolic pathways, metabolic states also provide feedback on the molecular clock. Therefore, a remarkable feature is reprogramming by nutritional challenges, such as a high-fat diet, fasting, ketogenic diet, and caloric restriction. In addition, various factors such as energy balance, histone modifications, and nuclear receptor activity are involved in the remodeling of the clock. Herein, we review the interaction of dietary components with the circadian system and illustrate the relationships linking the molecular clock to metabolism and critical roles in the remodeling process.
Collapse
Affiliation(s)
- Tomoki Sato
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, School of Medicine, INSERM U1233, University of California, Irvine, CA, USA
| | - Paolo Sassone-Corsi
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, School of Medicine, INSERM U1233, University of California, Irvine, CA, USA
| |
Collapse
|
38
|
Vaca-Dempere M, Kumar A, Sica V, Muñoz-Cánoves P. Running skeletal muscle clocks on time- the determining factors. Exp Cell Res 2022; 413:112989. [PMID: 35081395 DOI: 10.1016/j.yexcr.2021.112989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 12/08/2021] [Accepted: 12/19/2021] [Indexed: 11/23/2022]
Abstract
Circadian rhythms generate 24 h-long oscillations, which are key regulators of many aspects of behavior and physiology. Recent circadian transcriptome studies have discovered rhythmicity at the transcriptional level of hundreds of skeletal muscle genes, with roles in skeletal muscle growth, maintenance, and metabolic functions. These rhythms allow this tissue to perform molecular functions at the appropriate time of the day in order to anticipate environmental changes. However, while the last decade of research has characterized several aspects of the skeletal muscle molecular clock, many still are unexplored, including its functions, regulatory mechanisms, and interactions with other tissues. The central clock is believed to be located in the suprachiasmatic nucleus (SCN) of the brain hypothalamus, providing entrainment to peripheral organs through humoral and neuronal signals. However, these mechanisms of action are still unknown. Conversely, muscle tissue can be entrained through extrinsic, SCN-independent factors, such as feeding and physical activity. In this review, we provide an overview of the recent research about the extrinsic and intrinsic factors required for skeletal muscle clock regulation. Furthermore, we discuss the need for future studies to elucidate the mechanisms behind this regulation, which will in turn help dissect the role of circadian disruption at the onset of aging and diseases.
Collapse
Affiliation(s)
- Mireia Vaca-Dempere
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), 08003, Barcelona, Spain
| | - Arun Kumar
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), 08003, Barcelona, Spain
| | - Valentina Sica
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), 08003, Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), 08003, Barcelona, Spain; ICREA, 08010, Barcelona, Spain; Spanish National Center on Cardiovascular Research (CNIC), 28029, Madrid, Spain.
| |
Collapse
|
39
|
Ng GYQ, Sheng DPLK, Bae HG, Kang SW, Fann DYW, Park J, Kim J, Alli-Shaik A, Lee J, Kim E, Park S, Han JW, Karamyan V, Okun E, Dheen T, Hande MP, Vemuganti R, Mallilankaraman K, Lim LHK, Kennedy BK, Drummond GR, Sobey CG, Gunaratne J, Mattson MP, Foo RSY, Jo DG, Arumugam TV. Integrative epigenomic and transcriptomic analyses reveal metabolic switching by intermittent fasting in brain. GeroScience 2022; 44:2171-2194. [PMID: 35357643 DOI: 10.1007/s11357-022-00537-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/21/2022] [Indexed: 11/04/2022] Open
Abstract
Intermittent fasting (IF) remains the most effective intervention to achieve robust anti-aging effects and attenuation of age-related diseases in various species. Epigenetic modifications mediate the biological effects of several environmental factors on gene expression; however, no information is available on the effects of IF on the epigenome. Here, we first found that IF for 3 months caused modulation of H3K9 trimethylation (H3K9me3) in the cerebellum, which in turn orchestrated a plethora of transcriptomic changes involved in robust metabolic switching processes commonly observed during IF. Second, a portion of both the epigenomic and transcriptomic modulations induced by IF was remarkably preserved for at least 3 months post-IF refeeding, indicating that memory of IF-induced epigenetic changes was maintained. Notably, though, we found that termination of IF resulted in a loss of H3K9me3 regulation of the transcriptome. Collectively, our study characterizes the novel effects of IF on the epigenetic-transcriptomic axis, which controls myriad metabolic processes. The comprehensive analyses undertaken in this study reveal a molecular framework for understanding how IF impacts the metabolo-epigenetic axis of the brain and will serve as a valuable resource for future research.
Collapse
Affiliation(s)
- Gavin Yong-Quan Ng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Han-Gyu Bae
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sung Wook Kang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Yang-Wei Fann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jinsu Park
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Joonki Kim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Natural Products Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, Republic of Korea
| | - Asfa Alli-Shaik
- Translational Biomedical Proteomics Group, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Jeongmi Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Eunae Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sunyoung Park
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jeung-Whan Han
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Vardan Karamyan
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Science Center, Amarillo, TX, USA
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-IIan University, Ramat Gan, Israel
| | - Thameem Dheen
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Manoor Prakash Hande
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Karthik Mallilankaraman
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lina H K Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Brian K Kennedy
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School Medicine, National University of Singapore, Singapore, Singapore.,Buck Institute for Research On Aging, Novato, USA
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Jayantha Gunaratne
- Translational Biomedical Proteomics Group, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Roger Sik-Yin Foo
- Genome Institute of Singapore, Singapore, Singapore. .,Centre for Translational Medicine, Cardiovascular Research Institute, National University Health Systems, National University of Singapore, Singapore, Singapore.
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
| | - Thiruma V Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea. .,Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia.
| |
Collapse
|
40
|
Tuning up an aged clock: Circadian clock regulation in metabolism and aging. TRANSLATIONAL MEDICINE OF AGING 2022. [DOI: 10.1016/j.tma.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
41
|
Kinouchi K, Miyashita K, Itoh H. Chromatin Immunoprecipitation and Circadian Rhythms. Methods Mol Biol 2022; 2482:341-351. [PMID: 35610438 DOI: 10.1007/978-1-0716-2249-0_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Organisms exhibit daily changes of physiology and behavior to exert homeostatic adaptations to day-night cycles. The cyclic fluctuation also takes place at transcriptional levels, giving rise to rhythmic gene expression. Central to this oscillatory transcription is the core clock machinery which constitutes a circuit of transcriptional-translational feedback and achieves circadian functions accordingly. Chromatin immunoprecipitation provides understanding of such mechanisms that clock and non-clock transcription factors along with co-regulators and chromatin modifications dictate circadian epigenome through cyclic alterations of chromatin structures and molecular functions in a concerted fashion. Besides, innovation of high-throughput sequencing technology has broadened our horizon and renewed perspectives in circadian research. This article summarizes the methodology of a chromatin immunoprecipitation experiment in light of circadian rhythm research.
Collapse
Affiliation(s)
- Kenichiro Kinouchi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Kazutoshi Miyashita
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Itoh
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
42
|
Abstract
Isolation of primary hepatocytes and culturing these cells ex vivo provides a powerful platform to model liver physiology in vivo. Primary hepatocytes can be cultured for several days, the circadian clock can be synchronized, and these primary cells can be utilized for functional gene regulation analysis and metabolic studies. In this chapter, we describe detailed methodology for isolation of viable primary hepatocytes, techniques for culturing these cells, methods for synchronization of the circadian clock, transfection and luciferase reporter analysis, as well as glucose production assays as a functional readout of metabolic state.
Collapse
Affiliation(s)
- Sung Kook Chun
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine (UCI), Irvine, CA, USA
| | - Selma Masri
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine (UCI), Irvine, CA, USA.
| |
Collapse
|
43
|
Kumar D, Jahan S. Circadian Liver Metabolism Affects Management of Type 2 Diabetes Mellitus During Cytokine Storm Due to COVID-19. CHRONOBIOLOGY IN MEDICINE 2021; 3:129-136. [DOI: 10.33069/cim.2021.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 10/30/2021] [Indexed: 01/03/2025]
Abstract
Diabetes is managed to keep the blood sugar in normal range. This involves liver as glucose metabolizing organ and sensitization of somatic cells to utilize this glucose for daily energy requirements. The management is subjected to the rhythmic glucose intake as diet and liver circadian cycles that runs parallel to this zeitgeber. COVID-19 patients having diabetes as comorbid condition face the challenges of inflammatory cytokine management along with the organization of glucose. Increased blood glucose level during the cytokine storm further aggravates the pathophysiology of COVID-19 patients leading to high morbidity and mortality in such patients. Clinical treatment of these patients requires multidimensional approach involving circadian variation of hepatic physiology, glucose intake, and inflammatory cytokine release.
Collapse
|
44
|
Yadav VK, Berger JM, Singh P, Nagarajan P, Karsenty G. Embryonic osteocalcin signalling determines lifelong adrenal steroidogenesis and homeostasis in the mouse. J Clin Invest 2021; 132:153752. [PMID: 34905510 PMCID: PMC8843753 DOI: 10.1172/jci153752] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/30/2021] [Indexed: 11/17/2022] Open
Abstract
Through their ability to regulate gene expression in most organs, glucocorticoid hormones influence numerous physiological processes and therefore are key regulators of organismal homeostasis. In bone, glucocorticoid hormones inhibit the expression of the hormone Osteocalcin for poorly understood reasons. Here we show that in a classical endocrine feedback loop, osteocalcin in return enhances the biosynthesis of glucocorticoid but also mineralocorticoid hormones (adrenal steroidogenesis) in rodents and primates. Conversely, inactivating osteocalcin signalling in adrenal glands significantly impairs adrenal growth and steroidogenesis in mice. Embryo-made osteocalcin is necessary for normal Sf1 expression in foetal adrenal cells and adrenal cell steroidogenic differentiation, it therefore determines the number of steroidogenic cells present in adrenal glands of adult animals. Embryonic not postnatal osteocalcin also governs adrenal growth, adrenal steroidogenesis, blood pressure, electrolyte equilibrium and the rise of circulating corticosterone during the acute stress response in adult offspring. This osteocalcin-dependent regulation of adrenal development and steroidogenesis occurs even in the absence of a functional of hypothalamus-pituitary-adrenal axis; this explains why osteocalcin administration during pregnancy promotes adrenal growth and steroidogenesis and improves survival of adrenocorticotropic hormone signalling-deficient animals. This study reveals that a bone-derived, embryonic hormone influences lifelong adrenal functions and organismal homeostasis in the mouse.
Collapse
Affiliation(s)
- Vijay K Yadav
- Department of Genetics and Development, Columbia University, New York, United States of America
| | - Julian M Berger
- Department of Genetics and Development, Columbia University, New York, United States of America
| | - Parminder Singh
- Metabolic Research Laboratory, National Institute of Immunology, New Delhi, India
| | - Perumal Nagarajan
- Experimental Animal Facility, National Institute of Immunology, New Delhi, India
| | - Gerard Karsenty
- Department of Genetics and Development, Columbia University, New York, United States of America
| |
Collapse
|
45
|
Rong B, Wu Q, Saeed M, Sun C. Gut microbiota-a positive contributor in the process of intermittent fasting-mediated obesity control. ACTA ACUST UNITED AC 2021; 7:1283-1295. [PMID: 34786501 PMCID: PMC8567329 DOI: 10.1016/j.aninu.2021.09.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 09/12/2021] [Accepted: 09/25/2021] [Indexed: 12/12/2022]
Abstract
Historically, intermittent fasting (IF) has been considered as an effective strategy for controlling the weight of athletes before competition. Along with excellent insight into its application in various spaces by numerous studies, increasing IF-mediated positive effects have been reported, including anti-aging, neuroprotection, especially obesity control. Recently, the gut microbiota has been considered as an essential manipulator for host energy metabolism and its structure has been reported to be sensitive to dietary structure and habits, indicating that there is a potential and strong association between IF and gut microbiota. In this paper, we focus on the crosstalk between these symbionts and energy metabolism during IF which hold the promise to optimize host energy metabolism at various physical positions, including adipose tissue, liver and intestines, and further improve milieu internal homeostasis. Moreover, this paper also discusses the positive function of a potential recommendatory strain (Akkermansia muciniphila) based on the observational data for IF-mediated alternated pattern of gut microbiota and a hopefully regulatory pathway (circadian rhythm) for gut microbiota in IF-involved improvement on host energy metabolism. Finally, this review addresses the limitation and perspective originating from these studies, such as the association with tissue-specific bio-clock and single strain research, which may continuously reveal novel viewpoints and mechanisms to understand the energy metabolism and develop new strategies for treating obesity, diabetes, and metabolic disorders.
Collapse
Affiliation(s)
- Bohan Rong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Qiong Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Department of Pathophysiology, Qinghai University Medical College, Xining, Qinghai, China
| | - Muhammad Saeed
- Faculty of Animal Production & Technology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, 63100, Pakistan
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| |
Collapse
|
46
|
Tice AL, Laudato JA, Rossetti ML, Wolff CA, Esser KA, Lee C, Lang CH, Vied C, Gordon BS, Steiner JL. Binge alcohol disrupts skeletal muscle core molecular clock independent of glucocorticoids. Am J Physiol Endocrinol Metab 2021; 321:E606-E620. [PMID: 34541876 PMCID: PMC8791790 DOI: 10.1152/ajpendo.00187.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 01/11/2023]
Abstract
Circadian rhythms are central to optimal physiological function, as disruption contributes to the development of several chronic diseases. Alcohol (EtOH) intoxication disrupts circadian rhythms within liver, brain, and intestines, but it is unknown whether alcohol also disrupts components of the core clock in skeletal muscle. Female C57BL/6Hsd mice were randomized to receive either saline (control) or alcohol (EtOH) (5 g/kg) via intraperitoneal injection at the start of the dark cycle [Zeitgeber time (ZT12)], and gastrocnemius was collected every 4 h from control and EtOH-treated mice for the next 48 h following isoflurane anesthetization. In addition, metyrapone was administered before alcohol intoxication in separate mice to determine whether the alcohol-induced increase in serum corticosterone contributed to circadian gene regulation. Finally, synchronized C2C12 myotubes were treated with alcohol (100 mM) to assess the influence of centrally or peripherally mediated effects of alcohol on the muscle clock. Alcohol significantly disrupted mRNA expression of Bmal1, Per1/2, and Cry1/2 in addition to perturbing the circadian pattern of clock-controlled genes, Myod1, Dbp, Tef, and Bhlhe40 (P < 0.05), in muscle. Alcohol increased serum corticosterone levels and glucocorticoid target gene, Redd1, in muscle. Metyrapone prevented the EtOH-mediated increase in serum corticosterone but did not normalize the EtOH-induced change in Per1, Cry1 and Cry2, and Myod1 mRNA expression. Core clock gene expression (Bmal, Per1/2, and Cry1/2) was not changed following 4, 8, or 12 h of alcohol treatment on synchronized C2C12 myotubes. Therefore, binge alcohol disrupted genes of the core molecular clock independently of elevated serum corticosterone or direct effects of EtOH on the muscle.NEW & NOTEWORTHY Alcohol is a myotoxin that impairs skeletal muscle metabolism and function following either chronic consumption or acute binge drinking; however, mechanisms underlying alcohol-related myotoxicity have not been fully elucidated. Herein, we demonstrate that alcohol acutely interrupts oscillation of skeletal muscle core clock genes, and this is neither a direct effect of ethanol on the skeletal muscle, nor an effect of elevated serum corticosterone, a major clock regulator.
Collapse
Affiliation(s)
- Abigail L Tice
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida
| | - Joseph A Laudato
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida
| | - Michael L Rossetti
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida
| | - Christopher A Wolff
- Department of Physiology and Functional Genomics, University of Florida, Gainesville Florida
| | - Karyn A Esser
- Department of Physiology and Functional Genomics, University of Florida, Gainesville Florida
| | - Choogon Lee
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee Florida
| | - Charles H Lang
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Cynthia Vied
- Translational Science Laboratory, Florida State University College of Medicine, Tallahassee Florida
| | - Bradley S Gordon
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida
- Institute of Sports Sciences and Medicine, Florida State University, Tallahassee, Florida
| | - Jennifer L Steiner
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida
- Institute of Sports Sciences and Medicine, Florida State University, Tallahassee, Florida
| |
Collapse
|
47
|
Ribas-Aulinas F, Parra-Vargas M, Ramon-Krauel M, Diaz R, Lerin C, Cambras T, Jimenez-Chillaron JC. Time-Restricted Feeding during Puberty Ameliorates Adiposity and Prevents Hepatic Steatosis in a Mouse Model of Childhood Obesity. Nutrients 2021; 13:3579. [PMID: 34684586 PMCID: PMC8538558 DOI: 10.3390/nu13103579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Time restricted feeding (TRF) refers to dietary interventions in which food access is limited during a specific timeframe of the day. TRFs have proven useful in improving metabolic health in adult subjects with obesity. Their beneficial effects are mediated, in part, through modulating the circadian rhythm. Nevertheless, the translation of these dietary interventions onto obese/overweight children and adolescents remains uncharacterized. The objective of this study is to explore the feasibility of temporal dietary interventions for improving metabolic health in the context of childhood obesity. METHODS We have previously developed a mouse model of early adiposity (i.e., childhood obesity) through litter size reduction. Mice raised in small litters (SL) became obese as early as by two weeks of age, and as adults, they developed several obesity-related co-morbidities, including insulin resistance, glucose intolerance and hepatic steatosis. Here, we explored whether two independent short-term chrono-nutritional interventions might improve metabolic health in 1-month-old pre-pubertal SL mice. Both TRFs comprised 8 h feeding/14 h fasting. In the first one (TRF1) Control and SL mice had access to the diet for 8 h during the dark phase. In the second intervention (TRF2) food was available during the light:dark transitions. RESULTS TRF1 did not alter food intake nor ameliorate adiposity in SL-TRF1. In contrast, SL-TRF2 mice showed unintentional reduction of caloric intake, which was accompanied by reduced total body weight and adiposity. Strikingly, hepatic triglyceride content was completely normalized in SL-TRF1 and SL-TRF2 mice, when compared to the ad lib-fed SL mice. These effects were partially mediated by (i) clock-dependent signals, which might modulate the expression of Pparg or Cpt1a, and (ii) clock-independent signals, such as fasting itself, which could influence Fasn expression. CONCLUSIONS Time-restricted feeding is an effective and feasible nutritional intervention to improve metabolic health, namely hepatic steatosis, in a model of childhood obesity. These data open new avenues for future safe and efficient chrono-nutritional interventions aimed to improve metabolic health in children with overweight/obesity.
Collapse
Affiliation(s)
- Francesc Ribas-Aulinas
- Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues, 08950 Barcelona, Spain; (F.R.-A.); (M.P.-V.); (M.R.-K.); (R.D.); (C.L.)
| | - Marcela Parra-Vargas
- Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues, 08950 Barcelona, Spain; (F.R.-A.); (M.P.-V.); (M.R.-K.); (R.D.); (C.L.)
| | - Marta Ramon-Krauel
- Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues, 08950 Barcelona, Spain; (F.R.-A.); (M.P.-V.); (M.R.-K.); (R.D.); (C.L.)
- School of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | - Ruben Diaz
- Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues, 08950 Barcelona, Spain; (F.R.-A.); (M.P.-V.); (M.R.-K.); (R.D.); (C.L.)
- School of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | - Carles Lerin
- Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues, 08950 Barcelona, Spain; (F.R.-A.); (M.P.-V.); (M.R.-K.); (R.D.); (C.L.)
| | - Trinitat Cambras
- Department of Biochemistry and Physiology, School of Pharmacy, University of Barcelona, 08028 Barcelona, Spain;
| | - Josep C. Jimenez-Chillaron
- Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues, 08950 Barcelona, Spain; (F.R.-A.); (M.P.-V.); (M.R.-K.); (R.D.); (C.L.)
| |
Collapse
|
48
|
Ulgherait M, Midoun AM, Park SJ, Gatto JA, Tener SJ, Siewert J, Klickstein N, Canman JC, Ja WW, Shirasu-Hiza M. Circadian autophagy drives iTRF-mediated longevity. Nature 2021; 598:353-358. [PMID: 34588695 PMCID: PMC9395244 DOI: 10.1038/s41586-021-03934-0] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/19/2021] [Indexed: 01/13/2023]
Abstract
Time-restricted feeding (TRF) has recently gained interest as a potential anti-ageing treatment for organisms from Drosophila to humans1-5. TRF restricts food intake to specific hours of the day. Because TRF controls the timing of feeding, rather than nutrient or caloric content, TRF has been hypothesized to depend on circadian-regulated functions; the underlying molecular mechanisms of its effects remain unclear. Here, to exploit the genetic tools and well-characterized ageing markers of Drosophila, we developed an intermittent TRF (iTRF) dietary regimen that robustly extended fly lifespan and delayed the onset of ageing markers in the muscles and gut. We found that iTRF enhanced circadian-regulated transcription and that iTRF-mediated lifespan extension required both circadian regulation and autophagy, a conserved longevity pathway. Night-specific induction of autophagy was both necessary and sufficient to extend lifespan on an ad libitum diet and also prevented further iTRF-mediated lifespan extension. By contrast, day-specific induction of autophagy did not extend lifespan. Thus, these results identify circadian-regulated autophagy as a critical contributor to iTRF-mediated health benefits in Drosophila. Because both circadian regulation and autophagy are highly conserved processes in human ageing, this work highlights the possibility that behavioural or pharmaceutical interventions that stimulate circadian-regulated autophagy might provide people with similar health benefits, such as delayed ageing and lifespan extension.
Collapse
Affiliation(s)
- Matt Ulgherait
- Department of Genetics and Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Adil M Midoun
- Department of Biology, École Normale Supérieure, PSL Research University, Paris, France
| | - Scarlet J Park
- Skaggs Graduate School, The Scripps Research Institute, Jupiter, FL, USA
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Jared A Gatto
- Department of Genetics and Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Samantha J Tener
- Department of Genetics and Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Julia Siewert
- Department of Genetics and Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Naomi Klickstein
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - William W Ja
- Skaggs Graduate School, The Scripps Research Institute, Jupiter, FL, USA
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
49
|
Wang H, Chan TW, Vashisht AA, Drew BG, Calkin AC, Harris TE, Wohlschlegel JA, Xiao X, Reue K. Lipin 1 modulates mRNA splicing during fasting adaptation in liver. JCI Insight 2021; 6:e150114. [PMID: 34494556 PMCID: PMC8492312 DOI: 10.1172/jci.insight.150114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/23/2021] [Indexed: 01/03/2023] Open
Abstract
Lipin 1 regulates cellular lipid homeostasis through roles in glycerolipid synthesis (through phosphatidic acid phosphatase activity) and transcriptional coactivation. Lipin 1-deficient individuals exhibit episodic disease symptoms that are triggered by metabolic stress, such as stress caused by prolonged fasting. We sought to identify critical lipin 1 activities during fasting. We determined that lipin 1 deficiency induces widespread alternative mRNA splicing in liver during fasting, much of which is normalized by refeeding. The role of lipin 1 in mRNA splicing was largely independent of its enzymatic function. We identified interactions between lipin 1 and spliceosome proteins, as well as a requirement for lipin 1 to maintain homeostatic levels of spliceosome small nuclear RNAs and specific RNA splicing factors. In fasted Lpin1-/- liver, we identified a correspondence between alternative splicing of phospholipid biosynthetic enzymes and dysregulated phospholipid levels; splicing patterns and phospholipid levels were partly normalized by feeding. Thus, lipin 1 influences hepatic lipid metabolism through mRNA splicing, as well as through enzymatic and transcriptional activities, and fasting exacerbates the deleterious effects of lipin 1 deficiency on metabolic homeostasis.
Collapse
Affiliation(s)
- Huan Wang
- Human Genetics, David Geffen School of Medicine at UCLA
| | | | - Ajay A Vashisht
- Biological Chemistry, University of California, Los Angeles, California, USA
| | - Brian G Drew
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Anna C Calkin
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia
| | - Thurl E Harris
- Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - James A Wohlschlegel
- Biological Chemistry, University of California, Los Angeles, California, USA.,Molecular Biology Institute and
| | - Xinshu Xiao
- Bioinformatics Interdepartmental Program and.,Molecular Biology Institute and.,Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Karen Reue
- Human Genetics, David Geffen School of Medicine at UCLA,,Molecular Biology Institute and
| |
Collapse
|
50
|
Wahl D, LaRocca TJ. Transcriptomic Effects of Healthspan-Promoting Dietary Interventions: Current Evidence and Future Directions. Front Nutr 2021; 8:712129. [PMID: 34447778 PMCID: PMC8383293 DOI: 10.3389/fnut.2021.712129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
Aging is the greatest risk factor most diseases, including cardiovascular disorders, cancers, diabetes, and neurodegeneration, but select nutritional interventions may profoundly reduce the risk for these conditions. These interventions include calorie restriction, intermittent fasting, protein restriction, and reducing intake of certain amino acids. Certain ad libitum diets, including the Mediterranean, Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability, and Okinawan diets also promote healthy aging. Evidence indicates that these dietary strategies influence aging and healthspan by acting on the biological "hallmarks of aging" and especially upstream nutrient sensing pathways. Recent advances in "omics" technologies, including RNA-sequencing (transcriptomics), have increased our understanding of how such nutritional interventions may influence gene expression related to these biological mediators of aging, primarily in pre-clinical studies. However, whether these effects are also reflected in the human transcriptome, which may provide insight on other downstream/related cellular processes with aging, is an emerging topic. Broadly, the investigation of how these nutritional interventions influence the transcriptome may provide novel insight into pathways associated with aging, and potential targets to treat age-associated disease and increase healthspan. Therefore, the purpose of this mini review is to summarize what is known about the transcriptomic effects of key dietary/nutritional interventions in both pre-clinical models and humans, address gaps in the literature, and provide insight into future research directions.
Collapse
Affiliation(s)
- Devin Wahl
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, CO, United States
| | - Thomas J. LaRocca
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|