1
|
Zhang Z, Yan L, Treebak JT, Li MD. Circadian nutrition: is meal timing an elixir for fatigue? Sci Bull (Beijing) 2025; 70:309-312. [PMID: 39676008 DOI: 10.1016/j.scib.2024.11.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/17/2024] [Accepted: 11/22/2024] [Indexed: 12/17/2024]
Affiliation(s)
- Zhihui Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education (MOE) Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Lu Yan
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education (MOE) Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Min-Dian Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China; Ministry of Education (MOE) Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Chongqing 400038, China.
| |
Collapse
|
2
|
Yamazaki T, Cable EE, Schnabl B. Peroxisome proliferator-activated receptor delta and liver diseases. Hepatol Commun 2025; 9:e0646. [PMID: 39899669 DOI: 10.1097/hc9.0000000000000646] [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: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 02/05/2025] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors involved in transcriptional regulation and play an important role in many physiological and metabolic processes. Unlike PPAR-alpha and PPAR-gamma, PPAR-delta is ubiquitously expressed, and its activity is key to maintaining proper metabolic homeostasis within the liver. PPAR-delta not only regulates physiologic processes of lipid, glucose, and bile acid metabolism but also attenuates pathologic responses to alcohol metabolism, inflammation, fibrosis, and carcinogenesis, and is considered an important therapeutic target in liver diseases. Promising results have been reported in clinical trials for PPAR-delta agonists in liver disease, and the selective agonist seladelpar was recently conditionally approved in the United States as a new treatment option for primary biliary cholangitis. This review provides an overview of PPAR-delta's function and biology in the liver, examines its kinetics and therapeutic potential across different liver diseases, and discusses the current status of clinical trials involving its agonists.
Collapse
Affiliation(s)
- Tomoo Yamazaki
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan
| | | | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, California, USA
| |
Collapse
|
3
|
Munns J, Beale AD, Michaelides IN, Peak-Chew SY, Mihut A, Major-Styles CT, Zeng A, Storer RI, Edgar RS, Moreau K, O'Neill JS. Development of compounds for targeted degradation of mammalian cryptochrome proteins. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230342. [PMID: 39842482 PMCID: PMC11753880 DOI: 10.1098/rstb.2023.0342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/29/2024] [Accepted: 08/14/2024] [Indexed: 01/24/2025] Open
Abstract
The mammalian cryptochrome proteins (CRY1 and CRY2) are transcriptional repressors most notable for their role in circadian transcriptional feedback. Not all circadian rhythms depend on CRY proteins, however, and the CRY proteins are promiscuous interactors that also regulate many other processes. In cells with chronic CRY deficiency, protein homeostasis is highly perturbed, with a basal increase in cellular stress and activation of key inflammatory signalling pathways. Here, we developed tools to delineate the specific effects of CRY reduction, rather than chronic deficiency, to better understand the direct functions of CRY proteins. Performing a bioluminescence screen and immunoblot validation, we identified compounds that resulted in CRY reduction. Using these compounds, we found that circadian PERIOD2 (PER2) protein rhythms persisted under CRY-depleted conditions. By quantitative mass spectrometry, we found that CRY-depleted cells partially phenocopied the proteomic dysregulation of CRY-deficient cells, but showed minimal circadian phenotypes. We did, however, also observe substantial off-target effects of these compounds on luciferase activity and could not ascertain a specific mechanism of action. This work therefore highlights both the utility and the challenges of targeted protein degradation and bioluminescence reporter approaches in disentangling the contribution of CRY proteins to circadian rhythmicity, homeostasis and innate immune regulation.This article is part of the Theo Murphy meeting issue 'Circadian rhythms in infection and immunity'.
Collapse
Affiliation(s)
- Jack Munns
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, CambridgeCB2 0QH, UK
| | - Andrew D. Beale
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, CambridgeCB2 0QH, UK
| | | | - Sew Y. Peak-Chew
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, CambridgeCB2 0QH, UK
| | - Andrei Mihut
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, CambridgeCB2 0QH, UK
| | - Christine T. Major-Styles
- Department of Infectious Disease, Imperial College London, LondonW2 1NY, UK
- Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Aiwei Zeng
- Department of Infectious Disease, Imperial College London, LondonW2 1NY, UK
- Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - R. Ian Storer
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, CambridgeCB2 0AA, UK
| | - Rachel S. Edgar
- Department of Infectious Disease, Imperial College London, LondonW2 1NY, UK
- Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Kevin Moreau
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, CambridgeCB2 0AA, UK
| | - John S. O'Neill
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, CambridgeCB2 0QH, UK
| |
Collapse
|
4
|
Fitzgerald LS, Reynoso Spurrier CS, Lau NJ, Melamed MD, Burnett LA, Meyer GA, Gui C, Hevener AL, Sanford JA, Schenk S. Markers of mitochondrial function and oxidative metabolism in female skeletal muscle do not display intrinsic circadian regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631231. [PMID: 39868315 PMCID: PMC11761028 DOI: 10.1101/2025.01.08.631231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Mitochondria are key regulators of metabolism and ATP supply in skeletal muscle, while circadian rhythms influence many physiological processes. However, whether mitochondrial function is intrinsically regulated in a circadian manner in mouse skeletal muscle is inadequately understood. Accordingly, we measured post-absorptive transcript abundance of markers of mitochondrial biogenesis, dynamics, and metabolism (extensor digitorum longus [EDL], soleus, gastrocnemius), protein abundance of electron transport chain complexes (EDL and soleus), enzymatic activity of SDH (tibialis anterior and plantaris), and maximum uncoupled respiration (tibialis anterior) in different skeletal muscles from female C57BL/6NJ mice at four zeitgeber times (ZT), ZT 1, 7, 13, and 19. Our findings demonstrate that markers of mitochondrial function and oxidative metabolism do not display intrinsic time-of-day regulation at the gene, protein, enzymatic, or functional level. The core-clock genes Bmal1 and Dbp exhibited intrinsic circadian rhythmicity in skeletal muscle (i.e., EDL, soleus, gastrocnemius) and circadian amplitude varied by muscle type. These findings demonstrate that female mouse skeletal muscle does not display circadian regulation of markers of mitochondrial function or oxidative metabolism over 24 hours.
Collapse
|
5
|
Liu P, Nadeef S, Serag MF, Paytuví-Gallart A, Abadi M, Della Valle F, Radío S, Roda X, Dilmé Capó J, Adroub S, Hosny El Said N, Fallatah B, Celii M, Messa GM, Wang M, Li M, Tognini P, Aguilar-Arnal L, Habuchi S, Masri S, Sassone-Corsi P, Orlando V. PRC2-EZH1 contributes to circadian gene expression by orchestrating chromatin states and RNA polymerase II complex stability. EMBO J 2024; 43:6052-6075. [PMID: 39433902 PMCID: PMC11612306 DOI: 10.1038/s44318-024-00267-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/04/2024] [Accepted: 09/27/2024] [Indexed: 10/23/2024] Open
Abstract
Circadian rhythmicity of gene expression is a conserved feature of cell physiology. This involves fine-tuning between transcriptional and post-transcriptional mechanisms and strongly depends on the metabolic state of the cell. Together these processes guarantee an adaptive plasticity of tissue-specific genetic programs. However, it is unclear how the epigenome and RNA Pol II rhythmicity are integrated. Here we show that the PcG protein EZH1 has a gateway bridging function in postmitotic skeletal muscle cells. On the one hand, the circadian clock master regulator BMAL1 directly controls oscillatory behavior and periodic assembly of core components of the PRC2-EZH1 complex. On the other hand, EZH1 is essential for circadian gene expression at alternate Zeitgeber times, through stabilization of RNA Polymerase II preinitiation complexes, thereby controlling nascent transcription. Collectively, our data show that PRC2-EZH1 regulates circadian transcription both negatively and positively by modulating chromatin states and basal transcription complex stability.
Collapse
Affiliation(s)
- Peng Liu
- King Abdullah University of Science and Technology, KAUST, Biological and Environmental Sciences and Engineering Division, KAUST Environmental Epigenetics Program, Thuwal, 23955-6900, Kingdom of Saudi Arabia.
| | - Seba Nadeef
- King Abdullah University of Science and Technology, KAUST, Biological and Environmental Sciences and Engineering Division, KAUST Environmental Epigenetics Program, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Maged F Serag
- King Abdullah University of Science and Technology, KAUST, Biological and Environmental Sciences and Engineering Division, Bioscience Program, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | | | - Maram Abadi
- King Abdullah University of Science and Technology, KAUST, Biological and Environmental Sciences and Engineering Division, Bioscience Program, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Francesco Della Valle
- King Abdullah University of Science and Technology, KAUST, Biological and Environmental Sciences and Engineering Division, KAUST Environmental Epigenetics Program, Thuwal, 23955-6900, Kingdom of Saudi Arabia
- Altos Labs, Institute of Science, San Diego, CA, 92121, USA
| | - Santiago Radío
- Sequentia Biotech SL, Carrer Comte D'Urgell 240, Barcelona, 08036, Spain
| | - Xènia Roda
- Sequentia Biotech SL, Carrer Comte D'Urgell 240, Barcelona, 08036, Spain
| | - Jaïr Dilmé Capó
- Sequentia Biotech SL, Carrer Comte D'Urgell 240, Barcelona, 08036, Spain
| | - Sabir Adroub
- King Abdullah University of Science and Technology, KAUST, Biological and Environmental Sciences and Engineering Division, KAUST Environmental Epigenetics Program, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Nadine Hosny El Said
- King Abdullah University of Science and Technology, KAUST, Biological and Environmental Sciences and Engineering Division, KAUST Environmental Epigenetics Program, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Bodor Fallatah
- King Abdullah University of Science and Technology, KAUST, Biological and Environmental Sciences and Engineering Division, KAUST Environmental Epigenetics Program, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Mirko Celii
- King Abdullah University of Science and Technology, KAUST, Biological and Environmental Sciences and Engineering Division, KAUST Environmental Epigenetics Program, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Gian Marco Messa
- King Abdullah University of Science and Technology, KAUST, Biological and Environmental Sciences and Engineering Division, KAUST Environmental Epigenetics Program, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Mengge Wang
- King Abdullah University of Science and Technology, KAUST, Biological and Environmental Sciences and Engineering Division, Bioscience Program, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Mo Li
- King Abdullah University of Science and Technology, KAUST, Biological and Environmental Sciences and Engineering Division, Bioscience Program, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Paola Tognini
- University of California, Irvine, Department of Biological Chemistry, School of Medicine, Center for Epigenetics and Metabolism, Irvine, CA, 92697, USA
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, 56126, Italy
| | - Lorena Aguilar-Arnal
- University of California, Irvine, Department of Biological Chemistry, School of Medicine, Center for Epigenetics and Metabolism, Irvine, CA, 92697, USA
- Universidad Nacional Autónoma de México, Instituto de Investigaciones Biomédicas, Mexico City, 04510, Mexico
| | - Satoshi Habuchi
- King Abdullah University of Science and Technology, KAUST, Biological and Environmental Sciences and Engineering Division, Bioscience Program, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Selma Masri
- University of California Irvine, Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, Irvine, CA, 92697, USA
| | - Paolo Sassone-Corsi
- University of California, Irvine, Department of Biological Chemistry, School of Medicine, Center for Epigenetics and Metabolism, Irvine, CA, 92697, USA
| | - Valerio Orlando
- King Abdullah University of Science and Technology, KAUST, Biological and Environmental Sciences and Engineering Division, KAUST Environmental Epigenetics Program, Thuwal, 23955-6900, Kingdom of Saudi Arabia.
| |
Collapse
|
6
|
Fitzgerald LS, Bremner SN, Ward SR, Cho Y, Schenk S. Intrinsic Skeletal Muscle Function and Contraction-Stimulated Glucose Uptake Do Not Vary by Time-of-Day in Mice. FUNCTION 2024; 5:zqae035. [PMID: 39134511 DOI: 10.1093/function/zqae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/13/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024] Open
Abstract
A growing body of data suggests that skeletal muscle contractile function and glucose metabolism vary by time-of-day, with chronobiological effects on intrinsic skeletal muscle properties being proposed as the underlying mediator. However, no studies have directly investigated intrinsic contractile function or glucose metabolism in skeletal muscle over a 24 h circadian cycle. To address this, we assessed intrinsic contractile function and endurance, as well as contraction-stimulated glucose uptake, in isolated extensor digitorum longus and soleus from mice at 4 times-of-day (zeitgeber times 1, 7, 13, 19). Significantly, though both muscles demonstrated circadian-related changes in gene expression, there were no differences between the 4 time points in intrinsic contractile function, endurance, and contraction-stimulated glucose uptake, regardless of sex. Overall, these results suggest that time-of-day variation in exercise performance and the glycemia-reducing benefits of exercise are not due to chronobiological effects on intrinsic muscle function or contraction-stimulated glucose uptake.
Collapse
Affiliation(s)
- Liam S Fitzgerald
- Department of Orthopaedic Surgery, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Biomedical Sciences Graduate Program, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Medical Scientist Training Program, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Shannon N Bremner
- Department of Orthopaedic Surgery, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Samuel R Ward
- Department of Orthopaedic Surgery, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Radiology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Yoshitake Cho
- Division of Cardiovascular Medicine-Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Simon Schenk
- Department of Orthopaedic Surgery, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Biomedical Sciences Graduate Program, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
7
|
Hui Y, Zhong Y, Kuang L, Xu J, Hao Y, Cao J, Zheng T. O-GlcNAcylation of circadian clock protein Bmal1 impairs cognitive function in diabetic mice. EMBO J 2024; 43:5667-5689. [PMID: 39375536 PMCID: PMC11574178 DOI: 10.1038/s44318-024-00263-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024] Open
Abstract
Neuronal damage in the hippocampus induced by high glucose has been shown to promote the onset and development of cognitive impairment in diabetes, but the underlying molecular mechanism remains unclear. Guided by single-cell RNA sequencing, we here report that high glucose increases O-GlcNAcylation of Bmal1 in hippocampal neurons. This glycosylation promotes the binding of Clock to Bmal1, resulting in the expression of transcription factor Bhlhe41 and its target Dnajb4. Upregulated Dnajb4 in turn leads to ubiquitination and degradation of the mitochondrial Na + /Ca2+ exchanger NCLX, thereby inducing mitochondrial calcium overload that causes neuronal damage and cognitive impairment in mice. Notably, Bhlhe41 downregulation or treatment with a short peptide that specifically blocks O-GlcNAcylation of Bmal1 on Ser424 mitigated these adverse effects in diabetic mouse models. These data highlight the crucial role of O-GlcNAcylation in circadian clock gene expression and may facilitate the design of targeted therapies for diabetes-associated cognitive impairment.
Collapse
Affiliation(s)
- Ya Hui
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Yuanmei Zhong
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Liuyu Kuang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Jingxi Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Yuqi Hao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Jingxue Cao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Tianpeng Zheng
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China.
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China.
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China.
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China.
| |
Collapse
|
8
|
Nakajima S, Hanzawa F, Ikeda S, Oda H. Physical inactivity and breakfast skipping caused visceral fat accumulation in rats. Sci Rep 2024; 14:22644. [PMID: 39349485 PMCID: PMC11442645 DOI: 10.1038/s41598-024-68058-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/19/2024] [Indexed: 10/02/2024] Open
Abstract
Physical inactivity as well as breakfast skipping is known as risk factor for various metabolic diseases, such as obesity and type 2 diabetes. We have previously reported that a breakfast skipping model, in which the timing of feeding is delayed, induces abnormal lipid metabolism by altering the circadian rhythm of lipid metabolism-related genes in rats. The purpose of this study was to elucidate the synergistic effect of physical inactivity and breakfast skipping on lipid metabolism. We adopted sciatic neurectomized rats as physically inactive models, because we confirmed that the rats mildly decreased their spontaneous locomotor activity compared to sham-operated rats. And then the physically inactive model rats were fed a mild high-fat diet during zeitgeber time (ZT) 12-0 in the control group and ZT16-0 in the breakfast skipping group for 11 days. Body weight gain and total food intake were similar in both groups. Breakfast skipping induced a significant visceral fat accumulation, which was not observed in our previous breakfast skipping or physically inactive studies. The mRNA levels of clock and lipogenesis-related genes were altered by breakfast skipping in the liver and epididymal adipose tissue, and serum insulin level was altered by breakfast skipping. These results suggest that physical inactivity and breakfast skipping synergistically induces drastic visceral fat accumulation due to the alteration of circadian clock and lipid metabolism in the liver and adipose tissue. Therefore, regular feeding timing plays an important role in the health of a sedentary modern society.
Collapse
Affiliation(s)
- Syunsuke Nakajima
- Laboratory of Nutritional Biochemistry, Department of Applied Molecular Biosciences, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8601, Japan
| | - Fumiaki Hanzawa
- Department of Nutritional Science, Nagoya University of Arts and Sciences, Nisshin, 470-0196, Japan
| | - Saiko Ikeda
- Department of Nutritional Science, Nagoya University of Arts and Sciences, Nisshin, 470-0196, Japan
| | - Hiroaki Oda
- Laboratory of Nutritional Biochemistry, Department of Applied Molecular Biosciences, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8601, Japan.
| |
Collapse
|
9
|
Lee K, Hong KS, Park J, Park W. Readjustment of circadian clocks by exercise intervention is a potential therapeutic target for sleep disorders: a narrative review. Phys Act Nutr 2024; 28:35-42. [PMID: 39097996 PMCID: PMC11298283 DOI: 10.20463/pan.2024.0014] [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: 03/21/2024] [Revised: 04/25/2024] [Accepted: 05/17/2024] [Indexed: 08/06/2024] Open
Abstract
PURPOSE Circadian clocks are evolved endogenous biological systems that communicate with environmental cues to optimize physiological processes, such as the sleep-wake cycle, which is nearly related to quality of life. Sleep disorders can be treated using pharmacological strategies targeting melatonin, orexin, or core clock genes. Exercise has been widely explored as a behavioral treatment because it challenges homeostasis in the human body and affects the regulation of core clock genes. Exercise intervention at the appropriate time of the day can induce a phase shift in internal clocks. Although exercise is a strong external time cue for resetting the circadian clock, exercise therapy for sleep disorders remains poorly understood. METHODS This review focused on exercise as a potential treatment for sleep disorders by tuning the internal circadian clock. We used scientific paper depositories, including Google Scholar, PubMed, and the Cochrane Library, to identify previous studies that investigated the effects of exercise on circadian clocks and sleep disorders. RESULTS The exercise-induced adjustment of the circadian clock phase depended on exercise timing and individual chronotypes. Adjustment of circadian clocks through scheduled morning exercises can be appropriately prescribed for individuals with delayed sleep phase disorders. Individuals with advanced sleep phase disorders can synchronize their internal clocks with their living environment by performing evening exercises. Exercise-induced physiological responses are affected by age, sex, and current fitness conditions. CONCLUSION Personalized approaches are necessary when implementing exercise interventions for sleep disorders.
Collapse
Affiliation(s)
- Kwangjun Lee
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Kwang-Seok Hong
- Department of Physical Education, College of Education, Chung-Ang University, Seoul, Republic of Korea
| | - Jonghoon Park
- Department of Physical Education, Korea University, Seoul, Republic of Korea
| | - Wonil Park
- Department of Physical Education, College of Education, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
10
|
Dial MB, Malek EM, Cooper AR, Neblina GA, Vasileva NI, Hines DJ, McGinnis GR. Social jet lag impairs exercise volume and attenuates physiological and metabolic adaptations to voluntary exercise training. J Appl Physiol (1985) 2024; 136:996-1006. [PMID: 38450426 PMCID: PMC11305643 DOI: 10.1152/japplphysiol.00632.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/31/2024] [Accepted: 02/26/2024] [Indexed: 03/08/2024] Open
Abstract
Social jet lag (SJL) is a misalignment between sleep and wake times on workdays and free days. SJL leads to chronic circadian rhythm disruption and may affect nearly 70% of the general population, leading to increased risk for cardiometabolic diseases. This study investigated the effects of SJL on metabolic health, exercise performance, and exercise-induced skeletal muscle adaptations in mice. Ten-week-old C57BL/6J mice (n = 40) were allocated to four groups: control sedentary (CON-SED), control exercise (CON-EX), social jet lag sedentary (SJL-SED), and social jet lag exercise (SJL-EX). CON mice were housed under a 12:12-h light-dark cycle. SJL was simulated by implementing a 4-h phase delay for 3 days to simulate "weekends," followed by a 4-h phase advance back to "weekdays," for 6 wk. EX mice had free access to a running wheel. Graded exercise tests (GXTs) and glucose tolerance tests (GTTs) were performed at baseline and after intervention to monitor the effects of exercise and social jet lag on cardiorespiratory and metabolic health, respectively. SJL led to alterations in activity and running patterns and clock gene expression in skeletal muscle and decreased average running distance (P < 0.05). SJL-SED mice gained significantly more weight compared with CON-SED and SJL-EX mice (P < 0.01). SJL impaired fasting blood glucose and glucose tolerance compared with CON mice (P < 0.05), which was partially restored by exercise in SJL-EX mice. SJL also blunted improvements in exercise performance and mitochondrial content in the quadriceps. These data suggest that SJL blunted some cardiometabolic adaptations to exercise and that proper circadian hygiene is necessary for maintaining health and performance.NEW & NOTEWORTHY In mice, disrupting circadian rhythms with social jet lag for 6 wk caused significant weight gain, higher fasting blood glucose, and impaired glucose tolerance compared with control. Voluntary exercise in mice experiencing social jet lag prevented weight gain, though the mice still experienced increased fasting blood glucose and impaired exercise performance compared with trained mice not experiencing social jet lag. Social jet lag seems to be a potent circadian rhythm disruptor that impacts exercise-induced training adaptations.
Collapse
Affiliation(s)
- Michael B Dial
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada, Las Vegas, Nevada, United States
| | - Elias M Malek
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada, Las Vegas, Nevada, United States
| | - Austin R Cooper
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada, Las Vegas, Nevada, United States
| | - Greco A Neblina
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada, Las Vegas, Nevada, United States
| | - Nikoleta I Vasileva
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada, Las Vegas, Nevada, United States
| | - Dustin J Hines
- Department of Psychology, Psychological and Brain Sciences and Interdisciplinary Neuroscience Programs, University of Nevada, Las Vegas, Nevada, United States
| | - Graham R McGinnis
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada, Las Vegas, Nevada, United States
| |
Collapse
|
11
|
Dobbins TW, Swanson RM, Dennis AA, Rivera JD, Dinh TTN, Lemley CO, Burnett DD. Melatonin supplementation to sows in mid to late gestation affects offspring circadian, myogenic, and growth factor transcript abundance in pre and postnatal skeletal muscle. J Anim Sci 2024; 102:skae377. [PMID: 39679952 DOI: 10.1093/jas/skae377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 12/15/2024] [Indexed: 12/17/2024] Open
Abstract
The neuroendocrine hormone melatonin is associated with circadian rhythms and has antioxidant and vasodilative properties. In cattle, melatonin rescues fetal growth during maternal nutrient restriction in a seasonally dependent manner, but melatonin research in swine is limited. The objective of this study was to evaluate the effects of dietary melatonin supplementation during mid to late gestation on circadian rhythm and muscle growth and development of the longissimus dorsi in utero and postnatally. Sows received 20 mg of dietary melatonin daily (MEL) or no melatonin supplement (CON). Experiment 1 supplemented sows from gestational age (dGA) 38 ± 1 to 99 ± 1, experiment 2 supplemented sows from 41 to 106 ± 1 dGA, and experiment 3 supplemented sows from 60 dGA to farrowing. At harvest, morphometric measurements of all fetuses were taken, while the small (SM), medium (MED), and large (LG) piglets from each litter were used for further analysis. Prenatal data were analyzed using the MIXED procedure of SAS, and postnatal data were analyzed using the GLIMMIX procedure. Fetal morphometrics were analyzed for fixed the effect of treatment, and transcript abundance was analyzed for treatment, time, and size. Postnatal parameters were analyzed for fixed effects of treatment, size, and production stage. In experiment 1, MEL increased (P = 0.016) Period 1 (PER1) transcript abundance in the evening (PM) compared to the morning (AM). In experiment 1, myogenin (MYOG) transcript abundance was increased (P = 0.033) in MEL fetuses in the AM compared to MEL in the PM. Myogenic factor 5 (MYF5) and paired box 7 (PAX7) were increased (P = 0.016) in the PM. Fetuses from MEL-treated sows had increased (P < 0.05) BW, curve crown-rump length, and head circumference in experiment 2. In experiment 2, MEL increased (P = 0.012) PER1 and Period 2 (PER2) transcript abundance in the PM. In experiment 2, myoblast differentiation 1 (MYOD) was increased (P = 0.016) in SM and MED fetuses, while MYF5 and PAX7 were increased (P = 0.019) in SM fetuses. Postnatal BW was increased (P = 0.025) in MED and LG MEL-treated offspring compared to CON. Insulin-like growth factor 1 (IGF1) was downregulated (P = 0.050) in MEL-treated offspring, while insulin-like growth factor 1 receptor (IGF1R) was upregulated (P = 0.009) in MEL offspring. These results indicate that maternal melatonin supplementation during gestation modulates fetal circadian regulatory genes and alters myogenic genes during growth.
Collapse
Affiliation(s)
- Thomas W Dobbins
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA
| | - Rebecca M Swanson
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA
| | - Amberly A Dennis
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA
| | - J Daniel Rivera
- Department of Animal Science, University of Arkansas System Division of Agriculture, Southwest Research and Extension Center, Hope, AR 71801, USA
| | - Thu T N Dinh
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA
| | - Caleb O Lemley
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA
| | - Derris D Burnett
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA
| |
Collapse
|
12
|
Zhang K, Mi F, Li X, Wang Z, Jiang F, Song E, Guo P, Lan X. Detection of genetic variation in bovine CRY1 gene and its associations with carcass traits. Anim Biotechnol 2023; 34:3387-3394. [PMID: 36448652 DOI: 10.1080/10495398.2022.2149547] [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: 12/02/2022]
Abstract
The biological clock (also known as circadian clock) is closely related to growth and development, metabolism, and diseases in animals. As a part of the circadian clock, the cryptochrome circadian regulator 1 (CRY1) gene is involved in the regulation of biological processes such as osteogenesis, energy metabolism and cell proliferation, however, few studies have been reported on the relationship between this gene and animal carcass traits. Herein, a total of four insertion/deletion (InDel) loci within the CRY1 gene were detected in Shandong Black Cattle Genetic Resource (SDBCGR) population (n = 433). Among them, the P1-6-bp-del locus was polymorphic in population of interest. Moreover, the P1-6-bp-del locus showed two genotypes, with a higher insertion/insertion (II) genotype frequency (0.751) than insertion/deletion (ID) genotype frequency (0.249). Correlation analysis showed that the P1-6-bp-del locus polymorphisms were significantly associated with twenty carcass traits (e.g., slaughter weight, limb weight, and belly meat weight). Individuals with II genotype were significantly better than those with ID genotype for eighteen carcass traits. Therefore, the P1-6-bp-del locus of the CRY1 gene can be used as a molecular marker for beef cattle breeding.
Collapse
Affiliation(s)
- Kejing Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Fang Mi
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuelan Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Zhiying Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Fugui Jiang
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Enliang Song
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Peng Guo
- College of Computer and Information Engineering, Tianjin Agricultural University, Tianjin, China
| | - Xianyong Lan
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
13
|
Hühne-Landgraf A, Laurent K, Frisch MK, Wehr MC, Rossner MJ, Landgraf D. Rescue of Comorbid Behavioral and Metabolic Phenotypes of Arrhythmic Mice by Restoring Circadian Cry1/2 Expression in the Suprachiasmatic Nucleus. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:632-641. [PMID: 37881564 PMCID: PMC10593920 DOI: 10.1016/j.bpsgos.2023.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 06/03/2023] [Accepted: 06/22/2023] [Indexed: 10/27/2023] Open
Abstract
Background Psychiatric and metabolic disorders occur disproportionately often comorbidly, which poses particular hurdles for patients and therapists. However, the mechanisms that promote such comorbidities are largely unknown and therefore cannot yet be therapeutically targeted for the simultaneous treatment of both conditions. Because circadian clocks regulate most physiological processes and their disruption is a risk factor for both psychiatric and metabolic disorders, they may be considered as a potential mechanism for the development of comorbidities and a therapeutic target. In the current study, we investigated the latter assumption in Cry1/2-/- mice, which exhibit substantially disrupted endogenous circadian rhythms and marked metabolic and behavioral deficits. Methods By targeted virus-induced restoration of circadian rhythms in their suprachiasmatic nucleus, we can restore behavioral as well as several metabolic processes of these animals to near-normal circadian rhythmicity. Results Importantly, by rescuing suprachiasmatic nucleus rhythms, several of their anxiety-like behavioral as well as diabetes- and energy homeostasis-related deficits were significantly improved. Interestingly, however, this did not affect all deficits typical of Cry1/2-/- mice; for example, restlessness and body weight remained unaffected. Conclusions Taken together, the results of this study demonstrate, on the one hand, that restoration of disturbed circadian rhythms can be used to simultaneously treat psychiatric and metabolic deficits. On the other hand, the results also allow us to distinguish processes that depend more on local canonical clocks from those that depend more on suprachiasmatic nucleus rhythms.
Collapse
Affiliation(s)
- Anisja Hühne-Landgraf
- Circadian Biology Group, Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Katharina Laurent
- Circadian Biology Group, Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Muriel K. Frisch
- Circadian Biology Group, Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Michael C. Wehr
- Cell Signaling Group, Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
- Systasy Bioscience GmbH, Munich, Germany
| | - Moritz J. Rossner
- Systasy Bioscience GmbH, Munich, Germany
- Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Dominic Landgraf
- Circadian Biology Group, Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
14
|
Laothamatas I, Rasmussen ES, Green CB, Takahashi JS. Metabolic and chemical architecture of the mammalian circadian clock. Cell Chem Biol 2023; 30:1033-1052. [PMID: 37708890 PMCID: PMC10631358 DOI: 10.1016/j.chembiol.2023.08.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/20/2023] [Accepted: 08/23/2023] [Indexed: 09/16/2023]
Abstract
Circadian rhythms are endogenous periodic biological processes that occur on a daily timescale. These rhythms are generated by a transcriptional/translational feedback loop that consists of the CLOCK-BMAL1 heterodimeric transcriptional activator complex and the PER1/2-CRY1/2-CK1δ/ε repressive complex. The output pathways of this molecular feedback loop generate circadian rhythmicity in various biological processes. Among these, metabolism is a primary regulatory target of the circadian clock which can also feedback to modulate clock function. This intertwined relationship between circadian rhythms and metabolism makes circadian clock components promising therapeutic targets. Despite this, pharmacological therapeutics that target the circadian clock are relatively rare. In this review, we hope to stimulate interest in chemical chronobiology by providing a comprehensive background on the molecular mechanism of mammalian circadian rhythms and their connection to metabolism, highlighting important studies in the chemical approach to circadian research, and offering our perspectives on future developments in the field.
Collapse
Affiliation(s)
- Isara Laothamatas
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Emil Sjulstok Rasmussen
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Carla B Green
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
15
|
Unver S, Yigit S, Tural E, Yigit E, Atan T. Evaluation of a circadian rhythm gene (PER3) VNTR variant in Turkish athletes. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2023; 43:173-183. [PMID: 37610137 DOI: 10.1080/15257770.2023.2248198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/24/2023]
Abstract
OBJECTIVE Circadian rhythmicity has been shown to contribute to the regulation of key physiological and cognitive processes related to performance. The period homolog 3 (PER3) is expressed in a circadian pattern in the suprachiasmatic nucleus. Therefore, in this study, we aimed to evaluate the role of the variable tandem repeat (VNTR) variant of the PER3 gene in athletic performance in the Turkish population. METHODS This study included 223 subjects, which consisted of 123 athletes and 100 sedentary controls. Blood samples were drawn from all subjects. DNA was extracted from whole-blood samples. The PER3 VNTR variant was genotyped using the polymerase chain reaction-restriction method (PCR). The results of the analyses were evaluated for statistical significance. RESULTS The mean ages of athletes and controls were 22 ± 2.814 and 23 ± 3.561, respectively. Endurance athletes in the group were 21.1%, and sprint athletes were 78.9%. There was no statistical significance in terms of PER3 VNTR genotype distribution or allele frequency. In the recessive model, a statistically significant association was observed when the athletes were compared with the controls according to 4/4 + 4/5 versus 5/5 genotype (p = 0.020). CONCLUSION In this case-control study, for the first time in our country, we obtained findings suggesting that the PER3 VNTR variant may affect sports performance in the Turkish population. Results need to be replicated in different ethnic and larger samples.
Collapse
Affiliation(s)
- Saban Unver
- Department of Coaching Education, Faculty of Sports Science, University of Ondokuz Mayis, Samsun, Turkey
| | - Serbulent Yigit
- Department of Genetics, Faculty of Veterinary, Ondokuz Mayıs University, Samsun, Turkey
- Department of Medical Biology, Graduate Institute, Ondokuz Mayıs University, Samsun, Turkey
| | - Ercan Tural
- Department of Physiotherapy and Rehabilitation, Faculty of Health Science, Ondokuz Mayıs University, Samsun, Turkey
| | - Ercument Yigit
- Department of Sports Management, School of Physical Education and Sports, Halic University, Istanbul, Turkey
| | - Tulin Atan
- Department of Coaching Education, Faculty of Sports Science, University of Ondokuz Mayis, Samsun, Turkey
| |
Collapse
|
16
|
Awuah WA, Huang H, Kalmanovich J, Mehta A, Mikhailova T, Ng JC, Abdul-Rahman T, Adebusoye FT, Tan JK, Kamanousa K, Ferreira T, Roy S, Kundu M, Yarlagadda R, Mukerjee N, Alexiou A, Papadakis M. Circadian rhythm in systemic autoimmune conditions: Potential of chrono-immunology in clinical practice: A narrative review. Medicine (Baltimore) 2023; 102:e34614. [PMID: 37565922 PMCID: PMC10419593 DOI: 10.1097/md.0000000000034614] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/14/2023] [Indexed: 08/12/2023] Open
Abstract
The circadian rhythm (CR) is a fundamental biological process regulated by the Earth's rotation and solar cycles. It plays a critical role in various bodily functions, and its dysregulation can have systemic effects. These effects impact metabolism, redox homeostasis, cell cycle regulation, gut microbiota, cognition, and immune response. Immune mediators, cycle proteins, and hormones exhibit circadian oscillations, supporting optimal immune function and defence against pathogens. Sleep deprivation and disruptions challenge the regulatory mechanisms, making immune responses vulnerable. Altered CR pathways have been implicated in diseases such as diabetes, neurological conditions, and systemic autoimmune diseases (SADs). SADs involve abnormal immune responses to self-antigens, with genetic and environmental factors disrupting self-tolerance and contributing to conditions like Systemic Lupus Erythematosus, Rheumatoid Arthritis, and Inflammatory Myositis. Dysregulated CR may lead to increased production of pro-inflammatory cytokines, contributing to the systemic responses observed in SADs. Sleep disturbances significantly impact the quality of life of patients with SADs; however, they are often overlooked. The relationship between sleep and autoimmune conditions, whether causal or consequential to CR dysregulation, remains unclear. Chrono-immunology investigates the role of CR in immunity, offering potential for targeted therapies in autoimmune conditions. This paper provides an overview of the connections between sleep and autoimmune conditions, highlighting the importance of recognizing sleep disturbances in SADs and the need for further research into the complex relationship between the CR and autoimmune diseases.
Collapse
Affiliation(s)
| | - Helen Huang
- Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | | | - Aashna Mehta
- University of Debrecen-Faculty of Medicine, Debrecen, Hungary
| | | | - Jyi Cheng Ng
- Faculty of Medicine and Health Sciences, University of Putra Malaysia, Serdang, Malaysia
| | | | | | | | | | - Tomas Ferreira
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Sakshi Roy
- School of Medicine, Queen’s University Belfast, Belfast, UK
| | - Mrinmoy Kundu
- Institute of Medical Sciences and SUM Hospital, Bhubaneswar, India
| | | | - Nobendu Mukerjee
- Department of Microbiology, West Bengal State University, Barasat, India
- Department of Health Sciences, Novel Global Community Educational Foundation, Hebersham, NSW
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, Wuppertal, Germany
| |
Collapse
|
17
|
Rajput P, Kumar D, Krishnamurthy S. Chronic exposure to dim artificial light disrupts the daily rhythm in mitochondrial respiration in mouse suprachiasmatic nucleus. Chronobiol Int 2023; 40:938-951. [PMID: 37483020 DOI: 10.1080/07420528.2023.2236708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 05/29/2023] [Accepted: 07/10/2023] [Indexed: 07/25/2023]
Abstract
Circadian rhythms of physiology, behavior, and metabolism have an endogenous 24 h period that synchronizes with environmental cycles of light/dark and food availability. Alterations in light cycles are stressful and disrupt such diurnal oscillations. Recently, we witnessed a sudden rise in studies describing the mechanisms behind the interaction between the key characteristics of mitochondrial functions, peripheral clocks, and stress responses. To our knowledge, there is no study in the suprachiasmatic nuclei (SCN) describing the dysregulated mitochondrial bioenergetics under abnormal lighting conditions, which is common in today's modern world. Thus, we aimed to investigate the existence of daily changes in mitochondrial bioenergetics (respiratory control rate, RCR), mitochondrial abundance (mtDNA/nDNA), plasma corticosterone, and to test whether disturbances in the lighting conditions might influence such rhythms. To confirm this, mice were sacrificed, mitochondria were isolated from the suprachiasmatic nuclei in the brain and blood was collected, every 3 h at various time points zeitgeber time/circadian time, (0, 3, 6, 9, 12, 15, 18, 21, and 24 h) under 12:12 h light-dark (LD, 150 lux L: 0 lux D) cycle and chronic artificial dim lighting (LL, 5 lux: 5lux) conditions, of a 24 h period, respectively. Our results demonstrate the existence of robust daily rhythmicity in RCR, mtDNA/nDNA and plasma CORT under a normal LD cycle. However, these rhythms were significantly disrupted and clock genes expressions were dysregulated under chronic dim LL. Furthermore, mitochondrial abundance was significantly reduced during LL compared to their numbers under LD cycle. Our data demonstrate that the circadian clock regulates mitochondrial functions (RCR, number), essential for accomplishing daily energy demands and supply by the SCN neurons. Abnormal light exposure dysregulates mitochondrial functions in the SCN and may alter metabolism, resulting in obesity, diabetes, and other metabolic disorders. Therefore, properly designing lighting conditions in workplaces is essential to mitigate the adverse consequences of light on humans.
Collapse
Affiliation(s)
- Prabha Rajput
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, (Banaras Hindu University), Varanasi, India
| | | | - Sairam Krishnamurthy
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, (Banaras Hindu University), Varanasi, India
| |
Collapse
|
18
|
Xin H, Huang R, Zhou M, Chen J, Zhang J, Zhou T, Ji S, Liu X, Tian H, Lam SM, Bao X, Li L, Tong S, Deng F, Shui G, Zhang Z, Wong CCL, Li MD. Daytime-restricted feeding enhances running endurance without prior exercise in mice. Nat Metab 2023; 5:1236-1251. [PMID: 37365376 DOI: 10.1038/s42255-023-00826-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 05/17/2023] [Indexed: 06/28/2023]
Abstract
Physical endurance and energy conservation are essential for survival in the wild. However, it remains unknown whether and how meal timing regulates physical endurance and muscle diurnal rhythms. Here, we show that day/sleep time-restricted feeding (DRF) enhances running endurance by 100% throughout the circadian cycle in both male and female mice, compared to either ad libitum feeding or night/wake time-restricted feeding. Ablation of the circadian clock in the whole body or the muscle abolished the exercise regulatory effect of DRF. Multi-omics analysis revealed that DRF robustly entrains diurnal rhythms of a mitochondrial oxidative metabolism-centric network, compared to night/wake time-restricted feeding. Remarkably, muscle-specific knockdown of the myocyte lipid droplet protein perilipin-5 completely mimics DRF in enhancing endurance, enhancing oxidative bioenergetics and outputting rhythmicity to circulating energy substrates, including acylcarnitine. Together, our work identifies a potent dietary regimen to enhance running endurance without prior exercise, as well as providing a multi-omics atlas of muscle circadian biology regulated by meal timing.
Collapse
Affiliation(s)
- Haoran Xin
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Rongfeng Huang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Meiyu Zhou
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jianghui Chen
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
- Department of Cardiology, Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianxin Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Tingting Zhou
- Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Shushen Ji
- Department of Bioinformatics, GFK Biotech, Shanghai, China
| | - Xiao Liu
- Department of Bioinformatics, GFK Biotech, Shanghai, China
| | - He Tian
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- LipidALL Technologies, Changzhou, China
| | - Xinyu Bao
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Lihua Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Shifei Tong
- Department of Cardiology, Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Deng
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Zhihui Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China.
| | - Catherine C L Wong
- Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China.
- Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China.
| | - Min-Dian Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
19
|
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
|
20
|
Martin RA, Viggars MR, Esser KA. Metabolism and exercise: the skeletal muscle clock takes centre stage. Nat Rev Endocrinol 2023; 19:272-284. [PMID: 36726017 PMCID: PMC11783692 DOI: 10.1038/s41574-023-00805-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/12/2023] [Indexed: 02/03/2023]
Abstract
Circadian rhythms that influence mammalian homeostasis and overall health have received increasing interest over the past two decades. The molecular clock, which is present in almost every cell, drives circadian rhythms while being a cornerstone of physiological outcomes. The skeletal muscle clock has emerged as a primary contributor to metabolic health, as the coordinated expression of the core clock factors BMAL1 and CLOCK with the muscle-specific transcription factor MYOD1 facilitates the circadian and metabolic programme that supports skeletal muscle physiology. The phase of the skeletal muscle clock is sensitive to the time of exercise, which provides a rationale for exploring the interactions between the skeletal muscle clock, exercise and metabolic health. Here, we review the underlying mechanisms of the skeletal muscle clock that drive muscle physiology, with a particular focus on metabolic health. Additionally, we highlight the interaction between exercise and the skeletal muscle clock as a means of reinforcing metabolic health and discuss the possible implications of the time of exercise as a chronotherapeutic approach.
Collapse
Affiliation(s)
- Ryan A Martin
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, FL, USA
| | - Mark R Viggars
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, FL, USA
| | - Karyn A Esser
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA.
- Myology Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
21
|
Bahiru MS, Bittman EL. Adult Neurogenesis Is Altered by Circadian Phase Shifts and the Duper Mutation in Female Syrian Hamsters. eNeuro 2023; 10:ENEURO.0359-22.2023. [PMID: 36878716 PMCID: PMC10062491 DOI: 10.1523/eneuro.0359-22.2023] [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: 09/05/2022] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 03/08/2023] Open
Abstract
Cell birth and survival in the adult hippocampus are regulated by a circadian clock. Rotating shift work and jet lag disrupt circadian rhythms and aggravate disease. Internal misalignment, a state in which abnormal phase relationships prevail between and within organs, is proposed to account for adverse effects of circadian disruption. This hypothesis has been difficult to test because phase shifts of the entraining cycle inevitably lead to transient desynchrony. Thus, it remains possible that phase shifts, regardless of internal desynchrony, account for adverse effects of circadian disruption and alter neurogenesis and cell fate. To address this question, we examined cell birth and differentiation in the duper Syrian hamster (Mesocricetus auratus), a Cry1-null mutant in which re-entrainment of locomotor rhythms is greatly accelerated. Adult females were subjected to alternating 8 h advances and delays at eight 16 d intervals. BrdU, a cell birth marker, was given midway through the experiment. Repeated phase shifts decreased the number of newborn non-neuronal cells in WT, but not in duper hamsters. The duper mutation increased the number of BrdU-IR cells that stained for NeuN, which marks neuronal differentiation. Immunocytochemical staining for proliferating cell nuclear antigen indicated no overall effect of genotype or repeated shifts on cell division rates after 131 days. Cell differentiation, assessed by doublecortin, was higher in duper hamsters but was not significantly altered by repeated phase shifts. Our results support the internal misalignment hypothesis and indicate that Cry1 regulates cell differentiation. Phase shifts may determine neuronal stem cell survival and time course of differentiation after cell birth. Figure created with BioRender.
Collapse
Affiliation(s)
- Michael Seifu Bahiru
- Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts 01003
| | - Eric L Bittman
- Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts 01003
- Department of Biology, University of Massachusetts, Amherst, Massachusetts 01003
| |
Collapse
|
22
|
Hao Y, Xue T, Liu S, Geng S, Shi X, Qian P, He W, Zheng J, Li Y, Lou J, Shi T, Wang G, Wang X, Wang Y, Li Y, Song Y. Loss of CRY2 promotes regenerative myogenesis by enhancing PAX7 expression and satellite cell proliferation. MedComm (Beijing) 2023; 4:e202. [PMID: 36636367 PMCID: PMC9830134 DOI: 10.1002/mco2.202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 01/11/2023] Open
Abstract
The regenerative capacity of skeletal muscle is dependent on satellite cells. The circadian clock regulates the maintenance and function of satellite cells. Cryptochrome 2 (CRY2) is a critical component of the circadian clock, and its role in skeletal muscle regeneration remains controversial. Using the skeletal muscle lineage and satellite cell-specific CRY2 knockout mice (CRY2scko), we show that the deletion of CRY2 enhances muscle regeneration. Single myofiber analysis revealed that deletion of CRY2 stimulates the proliferation of myoblasts. The differentiation potential of myoblasts was enhanced by the loss of CRY2 evidenced by increased expression of myosin heavy chain (MyHC) and myotube formation in CRY2-/- cells versus CRY2+/+ cells. Immunostaining revealed that the number of mononucleated paired box protein 7 (PAX7+) cells associated with myotubes formed by CRY2-/- cells was increased compared with CRY2+/+ cells, suggesting that more reserve cells were produced in the absence of CRY2. Loss of CRY2 leads to the activation of the ERK1/2 signaling pathway and ETS1, which binds to the promoter of PAX7 to induce its transcription. CRY2 deficient myoblasts survived better in ischemic muscle. Therefore, CRY2 is essential in regulating skeletal muscle repair.
Collapse
Affiliation(s)
- Yingxue Hao
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Ting Xue
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Song‐Bai Liu
- Suzhou Vocational Health College, Suzhou Key Laboratory of Biotechnology for Laboratory MedicineSuzhouJiangsuP. R. China
| | - Sha Geng
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Xinghong Shi
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Panting Qian
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Wei He
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Jiqing Zheng
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Yanfang Li
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Jing Lou
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Tianze Shi
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Ge Wang
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| | - Xiaoxiao Wang
- Suzhou Vocational Health College, Suzhou Key Laboratory of Biotechnology for Laboratory MedicineSuzhouJiangsuP. R. China
| | - Yanli Wang
- Institutefor Cardiovascular Science and Department of Cardiovascular SurgeryFirst Affiliated Hospital and Medical College of Soochow UniversitySuzhouJiangsuP. R. China
- Collaborative Innovation Center of HematologySoochow UniversitySuzhouJiangsuP. R. China
| | - Yangxin Li
- Institutefor Cardiovascular Science and Department of Cardiovascular SurgeryFirst Affiliated Hospital and Medical College of Soochow UniversitySuzhouJiangsuP. R. China
- Collaborative Innovation Center of HematologySoochow UniversitySuzhouJiangsuP. R. China
| | - Yao‐Hua Song
- Cyrus Tang Hematology CenterCollaborative Innovation Center of HematologySoochow UniversitySuzhouP. R. China
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouP. R. China
- State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouP. R. China
| |
Collapse
|
23
|
Sun Q, Zhao J, Liu L, Wang X, Gu X. Identification of the potential biomarkers associated with circadian rhythms in heart failure. PeerJ 2023; 11:e14734. [PMID: 36699999 PMCID: PMC9869779 DOI: 10.7717/peerj.14734] [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: 11/14/2022] [Accepted: 12/21/2022] [Indexed: 01/22/2023] Open
Abstract
Background Heart failure (HF) is a syndrome with multiple clinical symptoms resulting from damage to the heart's structure and/or function with various pathogenic factors, which has developed as one of the most severe threats to human health. Approximately 13% of genes and about 8% of proteins contained in the heart are rhythmic, which could lead to HF if disrupted. Herein, we aimed to identify the circadian rhythms-related hub genes as potential biomarkers contributing to the identification and treatment of HF. Methods Expression data of ischemic and dilated cardiomyopathy samples with or without HF were collected from the GEO database. First, genes with differential expression in HF and healthy samples were identified, named as differentially expressed genes (DEGs), which were then intersected with circadian rhythms-related genes to identify circadian rhythms-related DEGs. A protein-protein interaction (PPI) network was established to screen hub genes. The performance of the hub genes to identify HF among healthy controls was assessed by referring to the receiver operating characteristic (ROC) curve. Additionally, quantitative real-time polymerase chain reaction (RT-PCR) was run to further validate the hub genes depending on clinical human peripheral blood samples. Results A total of 10,163 DEGs were determined, composed of 4,615 up-regulated genes and 5,548 down-regulated genes in HF patients in comparison to healthy controls. By overlapping the circadian rhythms-related genes in the Circadian Gene DataBase (CGDB), 723 circadian rhythms-related DEGs were obtained, mainly enriched in regulating lipid metabolic process, circadian rhythm and AMPK signaling pathway. Eight hub genes were screened out through the PPI network. The ROC curve indicated the high accuracy of five hub genes with AUC > 0.7, which also showed high accuracy validated by the external validation dataset. Furthermore, according to the results of quantitative RT-PCR, the HF group showed significantly increased relative mRNA expression of CRY2 and BHLHE41 while the decreased ARNTL and NPAS2 in comparison to controls, indicating the four hub genes as potential biomarkers of HF. Conclusion Our study validated that ARNTL, CRY2, BHLHE41 and NPAS2 could serve as potential biomarkers of circadian rhythm in HF. These results may provide a reference for employing novel markers or targets for the diagnosis and treatment of HF.
Collapse
Affiliation(s)
- Qiang Sun
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China,Department of Cardiology, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Jun Zhao
- Department of Cardiology, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Li Liu
- Department of Cardiology, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Xiaoliang Wang
- Department of Cardiology, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Xinshun Gu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
24
|
Sleep, circadian biology and skeletal muscle interactions: Implications for metabolic health. Sleep Med Rev 2022; 66:101700. [PMID: 36272396 DOI: 10.1016/j.smrv.2022.101700] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 12/07/2022]
Abstract
There currently exists a modern epidemic of sleep loss, triggered by the changing demands of our 21st century lifestyle that embrace 'round-the-clock' remote working hours, access to energy-dense food, prolonged periods of inactivity, and on-line social activities. Disturbances to sleep patterns impart widespread and adverse effects on numerous cells, tissues, and organs. Insufficient sleep causes circadian misalignment in humans, including perturbed peripheral clocks, leading to disrupted skeletal muscle and liver metabolism, and whole-body energy homeostasis. Fragmented or insufficient sleep also perturbs the hormonal milieu, shifting it towards a catabolic state, resulting in reduced rates of skeletal muscle protein synthesis. The interaction between disrupted sleep and skeletal muscle metabolic health is complex, with the mechanisms underpinning sleep-related disturbances on this tissue often multifaceted. Strategies to promote sufficient sleep duration combined with the appropriate timing of meals and physical activity to maintain circadian rhythmicity are important to mitigate the adverse effects of inadequate sleep on whole-body and skeletal muscle metabolic health. This review summarises the complex relationship between sleep, circadian biology, and skeletal muscle, and discusses the effectiveness of several strategies to mitigate the negative effects of disturbed sleep or circadian rhythms on skeletal muscle health.
Collapse
|
25
|
Circadian clock controls rhythms in ketogenesis by interfering with PPARα transcriptional network. Proc Natl Acad Sci U S A 2022; 119:e2205755119. [PMID: 36161962 PMCID: PMC9546578 DOI: 10.1073/pnas.2205755119] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ketone bodies are energy-rich metabolites and signaling molecules whose production is mainly regulated by diet. Caloric restriction (CR) is a dietary intervention that improves metabolism and extends longevity across the taxa. We found that CR induced high-amplitude daily rhythms in blood ketone bodies (beta-hydroxybutyrate [βOHB]) that correlated with liver βOHB level. Time-restricted feeding, another periodic fasting-based diet, also led to rhythmic βOHB but with reduced amplitude. CR induced strong circadian rhythms in the expression of fatty acid oxidation and ketogenesis genes in the liver. The transcriptional factor peroxisome-proliferator-activated-receptor α (PPARα) and its transcriptional target hepatokine fibroblast growth factor 21 (FGF21) are primary regulators of ketogenesis. Fgf21 expression and the PPARα transcriptional network became highly rhythmic in the CR liver, which implicated the involvement of the circadian clock. Mechanistically, the circadian clock proteins CLOCK, BMAL1, and cryptochromes (CRYs) interfered with PPARα transcriptional activity. Daily rhythms in the blood βOHB level and in the expression of PPARα target genes were significantly impaired in circadian clock-deficient Cry1,2-/- mice. These data suggest that blood βOHB level is tightly controlled and that the circadian clock is a regulator of diet-induced ketogenesis.
Collapse
|
26
|
Yang H, Gu X, Chen H, Zeng Q, Mao Z, Jin M, Li H, Ge Y, Zha J, Martyniuk CJ. Transcriptome profiling reveals toxicity mechanisms following sertraline exposure in the brain of juvenile zebrafish (Danio rerio). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 242:113936. [PMID: 35930839 DOI: 10.1016/j.ecoenv.2022.113936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/24/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
Sertraline (SER) is one of the most commonly detected antidepressants in the aquatic environment that can negatively affect aquatic organisms at low concentrations. Despite some knowledge on its acute toxicity to fish, the effects of chronic SER exposure remain poorly understood along with any underlying mechanisms of SER-induced toxicity. To address this knowledge gap, the effects of chronic exposure to three SER concentrations from low to high were investigated in zebrafish. Juvenile zebrafish were exposed to three concentrations of 1, 10, or 100 μg/L of SER for 28 d, after which indicators of oxidative stress and neurotoxicity in the brain were measured. Superoxide dismutase (SOD) activity was significantly enhanced by SER at 1 up to 100 μg/L, and catalase (CAT) activity was significantly induced by SER at 1 or 10 μg/L. The activity of acetylcholinesterase (AChE) was significantly induced by 10 and 100 μg/L of SER, and the serotonin (5-HT) level was significantly increased by all three concentrations of SER. To ascertain mechanisms of SER-induced toxicity, transcriptomics was conducted in the brain of zebrafish following 100 μg/L SER exposure. The molecular signaling pathways connected with circadian system and the immune system were significantly altered in the zebrafish brain. Based on transcriptomic data, the expression levels of six circadian clock genes were measured, and three genes were significantly altered in relative abundance in fish from all experimental treatments with SER, including cryptochrome circadian regulator 2 (cry2), period circadian clock 2 (per2), and period circadian clock 3 (per3). We hypothesize that the circadian system may be related to SER-induced neurotoxicity and oxidative stress in the central nervous system. This study reveals potential mechanisms and key events (i.e., oxidative stress and neurotoxicity) associated with SER-induced toxicity, and improves understanding of the molecular and biochemical pathways putatively perturbed by SER.
Collapse
Affiliation(s)
- Huiting Yang
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaohong Gu
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian 223300, China.
| | - Huihui Chen
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China.
| | - Qingfei Zeng
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China
| | - Zhigang Mao
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China
| | - Miao Jin
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongmin Li
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - You Ge
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinmiao Zha
- University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Christopher J Martyniuk
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611 USA
| |
Collapse
|
27
|
Yin F, Wu MM, Wei XL, Ren RX, Liu MH, Chen CQ, Yang L, Xie RQ, Jiang SY, Wang XF, Wang H. Hepatic NCoR1 deletion exacerbates alcohol-induced liver injury in mice by promoting CCL2-mediated monocyte-derived macrophage infiltration. Acta Pharmacol Sin 2022; 43:2351-2361. [PMID: 35149852 PMCID: PMC9433401 DOI: 10.1038/s41401-022-00863-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/07/2022] [Indexed: 02/06/2023] Open
Abstract
Nuclear receptor corepressor 1 (NCoR1) is a corepressor of the epigenetic regulation of gene transcription that has important functions in metabolism and inflammation, but little is known about its role in alcohol-associated liver disease (ALD). In this study, we developed mice with hepatocyte-specific NCoR1 knockout (NCoR1Hep-/-) using the albumin-Cre/LoxP system and investigated the role of NCoR1 in the pathogenesis of ALD and the underlying mechanisms. The traditional alcohol feeding model and NIAAA model of ALD were both established in wild-type and NCoR1Hep-/- mice. We showed that after ALD was established, NCoR1Hep-/- mice had worse liver injury but less steatosis than wild-type mice. We demonstrated that hepatocyte-specific loss of NCoR1 attenuated liver steatosis by promoting fatty acid oxidation by upregulating BMAL1 (a circadian clock component that has been reported to promote peroxisome proliferator activated receptor alpha (PPARα)-mediated fatty β-oxidation by upregulating de novo lipid synthesis). On the other hand, hepatocyte-specific loss of NCoR1 exacerbated alcohol-induced liver inflammation and oxidative stress by recruiting monocyte-derived macrophages via C-C motif chemokine ligand 2 (CCL2). In the mouse hepatocyte line AML12, NCoR1 knockdown significantly increased ethanol-induced CCL2 release. These results suggest that hepatocyte NCoR1 plays distinct roles in controlling liver inflammation and steatosis, which provides new insights into the development of treatments for steatohepatitis induced by chronic alcohol consumption.
Collapse
Affiliation(s)
- Fan Yin
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Miao-Miao Wu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Xiao-Li Wei
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Rui-Xue Ren
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Meng-Hua Liu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Chong-Qing Chen
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Liu Yang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Rui-Qian Xie
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Shan-Yue Jiang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Xue-Fu Wang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China.
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
28
|
The duper mutation reveals previously unsuspected functions of Cryptochrome 1 in circadian entrainment and heart disease. Proc Natl Acad Sci U S A 2022; 119:e2121883119. [PMID: 35930669 PMCID: PMC9371649 DOI: 10.1073/pnas.2121883119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The Cryptochrome 1 (Cry1)-deficient duper mutant hamster has a short free-running period in constant darkness (τDD) and shows large phase shifts in response to brief light pulses. We tested whether this measure of the lability of the circadian phase is a general characteristic of Cry1-null animals and whether it indicates resistance to jet lag. Upon advance of the light:dark (LD) cycle, both duper hamsters and Cry1-/- mice re-entrained locomotor rhythms three times as fast as wild types. However, accelerated re-entrainment was dissociated from the amplified phase-response curve (PRC): unlike duper hamsters, Cry1-/- mice show no amplification of the phase response to 15' light pulses. Neither the amplified acute shifts nor the increased rate of re-entrainment in duper mutants is due to acceleration of the circadian clock: when mutants drank heavy water to lengthen the period, these aspects of the phenotype persisted. In light of the health consequences of circadian misalignment, we examined effects of duper and phase shifts on a hamster model of heart disease previously shown to be aggravated by repeated phase shifts. The mutation shortened the lifespan of cardiomyopathic hamsters relative to wild types, but this effect was eliminated when mutants experienced 8-h phase shifts every second week, to which they rapidly re-entrained. Our results reveal previously unsuspected roles of Cry1 in phase shifting and longevity in the face of heart disease. The duper mutant offers new opportunities to understand the basis of circadian disruption and jet lag.
Collapse
|
29
|
Mori C, Lee JY, Tokumoto M, Satoh M. Cadmium Toxicity Is Regulated by Peroxisome Proliferator-Activated Receptor δ in Human Proximal Tubular Cells. Int J Mol Sci 2022; 23:ijms23158652. [PMID: 35955783 PMCID: PMC9369238 DOI: 10.3390/ijms23158652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 12/13/2022] Open
Abstract
Cadmium (Cd) is a toxic heavy metal that is widely present in the environment. Renal proximal tubule disorder is the main symptom of Cd chronic poisoning. Our previous study demonstrated that Cd inhibits the total activities of peroxisome proliferator-activated receptor (PPAR) transcription factors in human and rat proximal tubular cells. In this study, we investigated the involvement of PPAR in Cd renal toxicity using the HK-2 human proximal tubular cell line. Among PPAR isoform genes, only PPARD knockdown significantly showed resistance to Cd toxicity in HK-2 cells. The transcriptional activity of PPARδ was decreased not only by PPARD knockdown but also by Cd treatment. DNA microarray analysis showed that PPARD knockdown changed the expression of apoptosis-related genes in HK-2 cells. PPARD knockdown decreased apoptosis signals and caspase-3 activity induced by Cd treatment. PPARD knockdown did not affect the intracellular Cd level after Cd treatment. These results suggest that PPARδ plays a critical role in the modification of susceptibility to Cd renal toxicity and that the apoptosis pathway may be involved in PPARδ-related Cd toxicity.
Collapse
|
30
|
The Circadian Regulation of Nutrient Metabolism in Diet-Induced Obesity and Metabolic Disease. Nutrients 2022; 14:nu14153136. [PMID: 35956312 PMCID: PMC9370226 DOI: 10.3390/nu14153136] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/18/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Obesity and other metabolic diseases are major public health issues that are particularly prevalent in industrialized societies where circadian rhythmicity is disturbed by shift work, jet lag, and/or social obligations. In mammals, daylight entrains the hypothalamic suprachiasmatic nucleus (SCN) to a ≈24 h cycle by initiating a transcription/translation feedback loop (TTFL) of molecular clock genes. The downstream impacts of the TTFL on clock-controlled genes allow the SCN to set the rhythm for the majority of physiological, metabolic, and behavioral processes. The TTFL, however, is ubiquitous and oscillates in tissues throughout the body. Tissues outside of the SCN are entrained to other signals, such as fed/fasting state, rather than light input. This system requires a considerable amount of biological flexibility as it functions to maintain homeostasis across varying conditions contained within a 24 h day. In the face of either circadian disruption (e.g., jet lag and shift work) or an obesity-induced decrease in metabolic flexibility, this finely tuned mechanism breaks down. Indeed, both human and rodent studies have found that obesity and metabolic disease develop when endogenous circadian pacing is at odds with the external cues. In the following review, we will delve into what is known on the circadian rhythmicity of nutrient metabolism and discuss obesity as a circadian disease.
Collapse
|
31
|
Scholtes C, Giguère V. Transcriptional control of energy metabolism by nuclear receptors. Nat Rev Mol Cell Biol 2022; 23:750-770. [DOI: 10.1038/s41580-022-00486-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2022] [Indexed: 12/11/2022]
|
32
|
Mansingh S, Handschin C. Time to Train: The Involvement of the Molecular Clock in Exercise Adaptation of Skeletal Muscle. Front Physiol 2022; 13:902031. [PMID: 35547572 PMCID: PMC9081842 DOI: 10.3389/fphys.2022.902031] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/08/2022] [Indexed: 11/23/2022] Open
Abstract
Circadian rhythms regulate a host of physiological processes in a time-dependent manner to maintain homeostasis in response to various environmental stimuli like day and night cycles, food intake, and physical activity. Disruptions in circadian rhythms due to genetic mutations, shift work, exposure to artificial light sources, aberrant eating habits, and abnormal sleep cycles can have dire consequences for health. Importantly, exercise training efficiently ameliorates many of these adverse effects and the role of skeletal muscle in mediating the benefits of exercise is a topic of great interest. However, the molecular and physiological interactions between the clock, skeletal muscle function and exercise are poorly understood, and are most likely a combination of molecular clock components directly acting in muscle as well as in concordance with other peripheral metabolic organ systems like the liver. This review aims to consolidate existing experimental evidence on the involvement of molecular clock factors in exercise adaptation of skeletal muscle and to highlight the existing gaps in knowledge that need to be investigated to develop therapeutic avenues for diseases that are associated with these systems.
Collapse
|
33
|
Casanova-Vallve N, Duglan D, Vaughan ME, Pariollaud M, Handzlik MK, Fan W, Yu RT, Liddle C, Downes M, Delezie J, Mello R, Chan AB, Westermark PO, Metallo CM, Evans RM, Lamia KA. Daily running enhances molecular and physiological circadian rhythms in skeletal muscle. Mol Metab 2022; 61:101504. [PMID: 35470095 PMCID: PMC9079800 DOI: 10.1016/j.molmet.2022.101504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/12/2022] [Accepted: 04/19/2022] [Indexed: 11/16/2022] Open
Abstract
Objective Exercise is a critical component of a healthy lifestyle and a key strategy for the prevention and management of metabolic disease. Identifying molecular mechanisms underlying adaptation in response to chronic physical activity is of critical interest in metabolic physiology. Circadian rhythms broadly modulate metabolism, including muscle substrate utilization and exercise capacity. Here, we define the molecular and physiological changes induced across the daily cycle by voluntary low intensity daily exercise. Methods Wildtype C57BL6/J male and female mice were housed with or without access to a running wheel for six weeks. Maximum running speed was measured at four different zeitgeber times (ZTs, hours after lights on) using either electrical or manual stimulation to motivate continued running on a motorized treadmill. RNA isolated from plantaris muscles at six ZTs was sequenced to establish the impact of daily activity on genome-wide transcription. Patterns of gene expression were analyzed using Gene Set Enrichment Analysis (GSEA) and Detection of Differential Rhythmicity (DODR). Blood glucose, lactate, and ketones, and muscle and liver glycogen were measured before and after exercise. Results We demonstrate that the use of mild electrical shocks to motivate running negatively impacts maximum running speed in mice, and describe a manual method to motivate running in rodent exercise studies. Using this method, we show that time of day influences the increase in exercise capacity afforded by six weeks of voluntary wheel running: when maximum running speed is measured at the beginning of the nighttime active period in mice, there is no measurable benefit from a history of daily voluntary running, while maximum increase in performance occurs at the end of the night. We show that daily voluntary exercise dramatically remodels the murine muscle circadian transcriptome. Finally, we describe daily rhythms in carbohydrate metabolism associated with the time-dependent response to moderate daily exercise in mice. Conclusions Collectively, these data indicate that chronic nighttime physical activity dramatically remodels daily rhythms of murine muscle gene expression, which in turn support daily fluctuations in exercise performance. Daily voluntary running dramatically remodels the mouse muscle circadian transcriptome. Daily voluntary running maximally increases mouse running speed in the late active period. Muscle and liver glycogen content exhibit robust daily rhythms in laboratory mice. Use of mild electric shocks to motivate running in mice impairs maximum running speed.
Collapse
Affiliation(s)
| | - Drew Duglan
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Megan E Vaughan
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Marie Pariollaud
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Michal K Handzlik
- Department of Bioengineering, University of California, La Jolla, San Diego, CA 92093, USA; Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Weiwei Fan
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ruth T Yu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Christopher Liddle
- Storr Liver Centre, Westmead Institute for Medical Research and University of Sydney School of Medicine, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Julien Delezie
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Rebecca Mello
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Alanna B Chan
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Pål O Westermark
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Christian M Metallo
- Department of Bioengineering, University of California, La Jolla, San Diego, CA 92093, USA; Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Katja A Lamia
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA.
| |
Collapse
|
34
|
Cheng Q, Lu C, Qian R. The circadian clock regulates metabolic responses to physical exercise. Chronobiol Int 2022; 39:907-917. [PMID: 35282722 DOI: 10.1080/07420528.2022.2050384] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
It has been proposed for years that physical exercise ameliorates metabolic diseases. Optimal exercise timing in humans and mammals has indicated that circadian clocks play a vital role in exercise and body metabolism. Skeletal muscle metabolism exhibits a robust circadian rhythm under the control of the suprachiasmatic nucleus of the hypothalamus. Clock genes also control the development, differentiation, and function of skeletal muscles. In this review, we aimed to clarify the relationship between exercise, skeletal muscles, and the circadian clock. Health benefits can be attained by the scheduling of exercise at the best circadian time. Exercise therapy for metabolic diseases and cardiovascular health is a key adjuvant method. This review highlights the importance of exercise timing in maintaining healthy metabolism and circadian clocks.
Collapse
Affiliation(s)
- Qianyun Cheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chao Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ruizhe Qian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Manoogian ENC, Chow LS, Taub PR, Laferrère B, Panda S. Time-restricted Eating for the Prevention and Management of Metabolic Diseases. Endocr Rev 2022; 43:405-436. [PMID: 34550357 PMCID: PMC8905332 DOI: 10.1210/endrev/bnab027] [Citation(s) in RCA: 158] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Indexed: 02/08/2023]
Abstract
Time-restricted feeding (TRF, animal-based studies) and time-restricted eating (TRE, humans) are an emerging behavioral intervention approach based on the understanding of the role of circadian rhythms in physiology and metabolism. In this approach, all calorie intake is restricted within a consistent interval of less than 12 hours without overtly attempting to reduce calories. This article will summarize the origin of TRF/TRE starting with concept of circadian rhythms and the role of chronic circadian rhythm disruption in increasing the risk for chronic metabolic diseases. Circadian rhythms are usually perceived as the sleep-wake cycle and dependent rhythms arising from the central nervous system. However, the recent discovery of circadian rhythms in peripheral organs and the plasticity of these rhythms in response to changes in nutrition availability raised the possibility that adopting a consistent daily short window of feeding can sustain robust circadian rhythm. Preclinical animal studies have demonstrated proof of concept and identified potential mechanisms driving TRF-related benefits. Pilot human intervention studies have reported promising results in reducing the risk for obesity, diabetes, and cardiovascular diseases. Epidemiological studies have indicated that maintaining a consistent long overnight fast, which is similar to TRE, can significantly reduce risks for chronic diseases. Despite these early successes, more clinical and mechanistic studies are needed to implement TRE alone or as adjuvant lifestyle intervention for the prevention and management of chronic metabolic diseases.
Collapse
Affiliation(s)
| | - Lisa S Chow
- University of Minnesota, Division of Diabetes, Endocrinology and Metabolism, Minneapolis, Minnesota 55455, USA
| | - Pam R Taub
- University of California, San Diego, Division of Cardiovascular Diseases, Department of Medicine, 9434 Medical Center Drive, La Jolla, California 92037, USA
| | - Blandine Laferrère
- New York Nutrition Obesity Research Center, Division of Endocrinology, Department of Medicine, Columbia University Irving Medical Center; New York, New York 10032, USA
| | | |
Collapse
|
36
|
Nose Y, Hiromatsu C, Hanzawa F, Yumen Y, Kotani K, Nagai N. Match or mismatch between chronotype and sleep-wake cycle and their association with lean body mass gain among male high-school baseball players. Chronobiol Int 2022; 39:848-857. [PMID: 35189763 DOI: 10.1080/07420528.2022.2041657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
For athletes, it is important to acquire lean body mass (LBM) involving the skeletal muscle mass during their growth periods; however, the influence of chronotype on LBM gain remains unclear. We therefore aimed to investigate whether chronotype, sleep-wake cycle on weekdays (SWC-W), and their interaction contribute to LBM gain among adolescent male athletes in a 4-month intervention study. The participants were 45 male high-school baseball players. The intervention, including exercise menu (running and muscle strength training) and nutritional education, was conducted during a 4-month period of season-off training. The chronotype, body composition, lifestyle, and dietary intake were investigated before intervention (baseline) and after 4 months. Among the participants [Morningness (n = 14), Eveningness (n = 15), Intermediate (n = 16); ME score based on the Morningness/Eveningness Scale for Children (MES-C)], the midpoint of sleep on weekdays (MSW) was calculated in the "Morningness" and "Eveningness" participants, respectively. They were divided into 4 groups based on a match/mismatch with the chronotype: Type M-match (n = 8), Type M-mismatch (n = 6), Type E-match (n = 7), and Type E-mismatch (n = 8) groups. The data were compared among the 4 groups. Moreover, multiple regression analysis was conducted using an increase (kg) LBM gain as a response variable. When comparing the data between the "Morningness" and "Eveningness" participants, there were no differences in nutrient intake, the duration of training, or each parameter of body composition (per body weight) at baseline or after 4 months. There were also no differences in the rates of change in the body weight or each parameter of body composition. In groups in which the chronotype was consistent with the SWC-W (the Type M-match and Type E-match groups), the LBM gain were slightly greater than in the Type M-mismatch and Type E-mismatch groups (Type M-match: 3.5 ± 2.0 kg, Type M-mismatch: 1.6 ± 1.7 kg, Type E-match: 3.4 ± 2.2 kg, and Type E-mismatch: 1.2 ± 1.8 kg, p = .057). Multiple regression analysis revealed that an extent of the LBM gain was associated with a match between the chronotype and SWC-W (ß = 0.37, p = .030), independent of a long duration of training (ß = 0.52, p = .004). The results suggested that training-related LBM gain is associated with interactions between the chronotype and SWC-W in adolescent male athletes.Abbreviations: LBM: Lean body mass; SWC-W: Sleep-wake cycle on weekdays; ME score: Morningness-eveningness score; MES-C: Morningness/Eveningness Scale for Children; MSW: Midpoint of sleep on weekdays; MSF: Midpoint of sleep on free days; MSFsc: Midpoint of sleep on free days corrected for sleep debt accumulated through weekdays.
Collapse
Affiliation(s)
- Yoko Nose
- Laboratory of Nutritional Physiology, Graduate School of Human Science and Environment, University of Hyogo, Hyogo, Japan.,Department of Nutrition, Osaka International College, Osaka, Japan
| | - Chiyori Hiromatsu
- Department of Food Science and Nutrition, School of Human Science and Environment, University of Hyogo, Hyogo, Japan
| | - Fumiaki Hanzawa
- Department of Food Science and Nutrition, School of Human Science and Environment, University of Hyogo, Hyogo, Japan
| | - Yukina Yumen
- Laboratory of Nutritional Physiology, Graduate School of Human Science and Environment, University of Hyogo, Hyogo, Japan
| | - Kazuhiko Kotani
- Division of Community and Family Medicine, Jichi Medical University, Tochigi, Japan
| | - Narumi Nagai
- Laboratory of Nutritional Physiology, Graduate School of Human Science and Environment, University of Hyogo, Hyogo, Japan.,Department of Food Science and Nutrition, School of Human Science and Environment, University of Hyogo, Hyogo, Japan
| |
Collapse
|
37
|
Quattrocelli M, Wintzinger M, Miz K, Levine DC, Peek CB, Bass J, McNally EM. Muscle mitochondrial remodeling by intermittent glucocorticoid drugs requires an intact circadian clock and muscle PGC1α. SCIENCE ADVANCES 2022; 8:eabm1189. [PMID: 35179955 PMCID: PMC8856622 DOI: 10.1126/sciadv.abm1189] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/23/2021] [Indexed: 06/14/2023]
Abstract
Exogenous glucocorticoids interact with the circadian clock, but little attention is paid to the timing of intake. We recently found that intermittent once-weekly prednisone improved nutrient oxidation in dystrophic muscle. Here, we investigated whether dosage time affected prednisone effects on muscle bioenergetics. In mice treated with once-weekly prednisone, drug dosing in the light-phase promoted nicotinamide adenine dinucleotide (NAD+) levels and mitochondrial function in wild-type muscle, while this response was lost with dark-phase dosing. These effects depended on a normal circadian clock since they were disrupted in muscle from [Brain and muscle Arnt-like protein-1 (Bmal1)]-knockout mice. The light-phase prednisone pulse promoted BMAL1-dependent glucocorticoid receptor recruitment on noncanonical targets, including Nampt and Ppargc1a [peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α)]. In mice with muscle-restricted inducible PGC1α ablation, bioenergetic stimulation by light-phase prednisone required PGC1α. These results demonstrate that glucocorticoid "chronopharmacology" for muscle bioenergetics requires an intact clock and muscle PGC1α activity.
Collapse
Affiliation(s)
- Mattia Quattrocelli
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Michelle Wintzinger
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Karen Miz
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Daniel C. Levine
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Clara Bien Peek
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joseph Bass
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
38
|
Krishnan RH, Sadu L, Das UR, Satishkumar S, Pranav Adithya S, Saranya I, Akshaya R, Selvamurugan N. Role of p300, a histone acetyltransferase enzyme, in osteoblast differentiation. Differentiation 2022; 124:43-51. [DOI: 10.1016/j.diff.2022.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 12/21/2022]
|
39
|
de Goede P, Wüst RCI, Schomakers BV, Denis S, Vaz FM, Pras-Raves ML, van Weeghel M, Yi CX, Kalsbeek A, Houtkooper RH. Time-restricted feeding during the inactive phase abolishes the daily rhythm in mitochondrial respiration in rat skeletal muscle. FASEB J 2022; 36:e22133. [PMID: 35032416 DOI: 10.1096/fj.202100707r] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 11/26/2021] [Accepted: 12/17/2021] [Indexed: 01/06/2023]
Abstract
Shift-workers show an increased incidence of type 2 diabetes mellitus (T2DM). A possible mechanism is the disruption of the circadian timing of glucose homeostasis. Skeletal muscle mitochondrial function is modulated by the molecular clock. We used time-restricted feeding (TRF) during the inactive phase to investigate how mistimed feeding affects muscle mitochondrial metabolism. Rats on an ad libitum (AL) diet were compared to those that could eat only during the light (inactive) or dark (active) phase. Mitochondrial respiration, metabolic gene expressions, and metabolite concentrations were determined in the soleus muscle. Rats on AL feeding or dark-fed TRF showed a clear daily rhythm in muscle mitochondrial respiration. This rhythm in mitochondrial oxidative phosphorylation capacity was abolished in light-fed TRF animals and overall 24h respiration was lower. The expression of several genes involved in mitochondrial biogenesis and the fission/fusion machinery was altered in light-fed animals. Metabolomics analysis indicated that light-fed animals had lost rhythmic levels of α-ketoglutarate and citric acid. Contrastingly, lipidomics showed that light-fed animals abundantly gained rhythmicity in levels of triglycerides. Furthermore, while the RER shifted entirely with the food intake in the light-fed animals, many measured metabolic parameters (e.g., activity and mitochondrial respiration) did not strictly align with the shifted timing of food intake, resulting in a mismatch between expected metabolic supply/demand (as dictated by the circadian timing system and light/dark-cycle) and the actual metabolic supply/demand (as dictated by the timing of food intake). These data suggest that shift-work impairs mitochondrial metabolism and causes metabolic inflexibility, which can predispose to T2DM.
Collapse
Affiliation(s)
- Paul de Goede
- Laboratory of Endocrinology, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Hypothalamic Integration Mechanisms Group, Netherlands Institute for Neuroscience (NIN), an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Rob C I Wüst
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bauke V Schomakers
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Core Facility Metabolomics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Simone Denis
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Frédéric M Vaz
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Core Facility Metabolomics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Mia L Pras-Raves
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Core Facility Metabolomics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Core Facility Metabolomics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Chun-Xia Yi
- Laboratory of Endocrinology, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Laboratory of Endocrinology, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Hypothalamic Integration Mechanisms Group, Netherlands Institute for Neuroscience (NIN), an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Wong DCS, Seinkmane E, Zeng A, Stangherlin A, Rzechorzek NM, Beale AD, Day J, Reed M, Peak‐Chew SY, Styles CT, Edgar RS, Putker M, O’Neill JS. CRYPTOCHROMES promote daily protein homeostasis. EMBO J 2022; 41:e108883. [PMID: 34842284 PMCID: PMC8724739 DOI: 10.15252/embj.2021108883] [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: 06/03/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 11/29/2022] Open
Abstract
The daily organisation of most mammalian cellular functions is attributed to circadian regulation of clock-controlled protein expression, driven by daily cycles of CRYPTOCHROME-dependent transcriptional feedback repression. To test this, we used quantitative mass spectrometry to compare wild-type and CRY-deficient fibroblasts under constant conditions. In CRY-deficient cells, we found that temporal variation in protein, phosphopeptide, and K+ abundance was at least as great as wild-type controls. Most strikingly, the extent of temporal variation within either genotype was much smaller than overall differences in proteome composition between WT and CRY-deficient cells. This proteome imbalance in CRY-deficient cells and tissues was associated with increased susceptibility to proteotoxic stress, which impairs circadian robustness, and may contribute to the wide-ranging phenotypes of CRY-deficient mice. Rather than generating large-scale daily variation in proteome composition, we suggest it is plausible that the various transcriptional and post-translational functions of CRY proteins ultimately act to maintain protein and osmotic homeostasis against daily perturbation.
Collapse
Affiliation(s)
| | | | - Aiwei Zeng
- MRC Laboratory of Molecular BiologyCambridgeUK
| | | | | | | | - Jason Day
- Department of Earth SciencesUniversity of CambridgeCambridgeUK
| | - Martin Reed
- MRC Laboratory of Molecular BiologyCambridgeUK
| | | | | | - Rachel S Edgar
- Department of Infectious DiseasesImperial CollegeLondonUK
| | - Marrit Putker
- MRC Laboratory of Molecular BiologyCambridgeUK
- Present address:
Crown BioscienceUtrechtthe Netherlands
| | | |
Collapse
|
41
|
Sun Y, Wang Z, Nie C, Xue L, Wang Y, Song C, Fan M, Qian H, Ying H, Li Y, Wang L. Hydroxysafflor Yellow A Alters Fuel Selection From Glucose to Fat by Activating the PPARδ Pathway in Myocytes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:13838-13848. [PMID: 34757740 DOI: 10.1021/acs.jafc.1c06034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Modulation of fuel selection is critical in skeletal muscle function. Hydroxysafflor yellow A (HSYA) is the major bioactive component in safflower (Carthamus tinctorius L.) and, in our previous study, has been demonstrated to promote a shift from fast to slow myofiber. However, the effects of HSYA on fuel selection in skeletal muscle and its underlying mechanisms remain unclear. In this study, the in vitro experiments found that water extracts of safflower, rich in HSYA, significantly suppressed the expressions of the genes related to glucose utilization and activated the expressions of the lipolysis genes. Furthermore, HSYA resulted in a shift in substrate utilization toward fat relative to carbohydrates in C2C12 myotubes. Animal tests showed HSYA could significantly reduce the respiratory exchange ratio and prolonge endurance performance in mice and also trigger a switch in intramuscular fuel selection preference from carbohydrates to fat at rest and during exercise. Mechanistic studies revealed that HSYA converted this fuel selection by activating peroxisome proliferator activated receptor δ (PPARδ), and these effects of HSYA could be reversed by specific suppression of PPARδ by PPARδ siRNA. Collectively, our study demonstrated that HSYA can switch substrate utilization from glucose to fat in myocytes by activating PPARδ signaling, resulting in prolonged endurance performance. These findings provided direct evidence for the endurance performance enhancement effect of HSYA and explored new perspectives for the innovation and application of HSYA in the health care industry.
Collapse
Affiliation(s)
- Yujie Sun
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhijun Wang
- COFCO Aerocean Oils & Grain Industrial Co., Ltd, Shawan, NO.1 West Park Road, West Urumqi Road, Shawan County, Tacheng District, Xinjiang Province 832100, China
| | - Chenzhipeng Nie
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Lamei Xue
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yu Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Chunmei Song
- Food & Pharmacy College, Xuchang University, Xuchang 461000, China
| | - Mingcong Fan
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Haifeng Qian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hao Ying
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, People's Republic of China
| | - Yan Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Li Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
42
|
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
|
43
|
Expression of cell proliferation regulatory factors bricd5, tnfrsf21, cdk1 correlates with expression of clock gene cry1 in testes of Hu rams during puberty. Mol Biol Rep 2021; 48:7379-7385. [PMID: 34626314 DOI: 10.1007/s11033-021-06747-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/04/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Cryptochrome 1 (cry1), the core regulator of the circadian clock, is essential for ontogeny and mammalian reproduction. Unlike in other tissues, the cry1 gene have noncircadian functions in spermatogenesis, which implies the unique role of cry1 gene in the development of testis. The role of cry1 during the puberty has not been described yet. This study aimed to explore the relationship between cry1 expression and spermatogenic cell numbers. METHODS AND RESULTS We analyzed testicular tissues from Hu sheep aged 0-180 days by hematoxylin and eosin staining, measured cry1 and cell proliferation regulatory factors (bricd5, tnfrsf21, cdk1) expression by quantitative real-time PCR and characterized the transcription factor in the 5' flanking region of cry1 gene. The data revealed that the number of spermatocytes and early spermatocytes increased rapidly from 90 to 120 dpp (day postpartum). Correspondingly, there was a marked variation in the cry1 and cell proliferation related genes (bricd5, tnfrsf21, cdk1) mRNA expression in the testes from the age of 90 days to 180 days (p < 0.05). We also identified some transcription factors (tcfl5) related to cell proliferation. CONCLUSIONS There is a significant causal relationship between the transcription level of cry1 gene in Hu sheep testes and the number of spermatogenic cells. It is speculated that cry1 gene may regulate the proliferation of spermatogenic cells by regulating the expression of cell proliferation related genes such as bricd5, tnfrsf21 and cdk1.
Collapse
|
44
|
Li MD, Xin H, Yuan Y, Yang X, Li H, Tian D, Zhang H, Zhang Z, Han TL, Chen Q, Duan G, Ju D, Chen K, Deng F, He W. Circadian Clock-Controlled Checkpoints in the Pathogenesis of Complex Disease. Front Genet 2021; 12:721231. [PMID: 34557221 PMCID: PMC8452875 DOI: 10.3389/fgene.2021.721231] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/16/2021] [Indexed: 12/26/2022] Open
Abstract
The circadian clock coordinates physiology, metabolism, and behavior with the 24-h cycles of environmental light. Fundamental mechanisms of how the circadian clock regulates organ physiology and metabolism have been elucidated at a rapid speed in the past two decades. Here we review circadian networks in more than six organ systems associated with complex disease, which cluster around metabolic disorders, and seek to propose critical regulatory molecules controlled by the circadian clock (named clock-controlled checkpoints) in the pathogenesis of complex disease. These include clock-controlled checkpoints such as circadian nuclear receptors in liver and muscle tissues, chemokines and adhesion molecules in the vasculature. Although the progress is encouraging, many gaps in the mechanisms remain unaddressed. Future studies should focus on devising time-dependent strategies for drug delivery and engagement in well-characterized organs such as the liver, and elucidating fundamental circadian biology in so far less characterized organ systems, including the heart, blood, peripheral neurons, and reproductive systems.
Collapse
Affiliation(s)
- Min-Dian Li
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Haoran Xin
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yinglin Yuan
- Medical Center of Hematology, The Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Xinqing Yang
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hongli Li
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dingyuan Tian
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Zhang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhihui Zhang
- Department of Cardiology and the Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ting-Li Han
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Chen
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Guangyou Duan
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Dapeng Ju
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ka Chen
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Fang Deng
- Key Laboratory of Extreme Environmental Medicine, Department of Pathophysiology, College of High Altitude Military Medicine, Ministry of Education of China, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of High Altitude Medicine, PLA, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wenyan He
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | |
Collapse
|
45
|
Adamovich Y, Dandavate V, Ezagouri S, Manella G, Zwighaft Z, Sobel J, Kuperman Y, Golik M, Auerbach A, Itkin M, Malitsky S, Asher G. Clock proteins and training modify exercise capacity in a daytime-dependent manner. Proc Natl Acad Sci U S A 2021; 118:e2101115118. [PMID: 34426495 PMCID: PMC8536342 DOI: 10.1073/pnas.2101115118] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Exercise and circadian biology are closely intertwined with physiology and metabolism, yet the functional interaction between circadian clocks and exercise capacity is only partially characterized. Here, we tested different clock mutant mouse models to examine the effect of the circadian clock and clock proteins, namely PERIODs and BMAL1, on exercise capacity. We found that daytime variance in endurance exercise capacity is circadian clock controlled. Unlike wild-type mice, which outperform in the late compared with the early part of their active phase, PERIODs- and BMAL1-null mice do not show daytime variance in exercise capacity. It appears that BMAL1 impairs and PERIODs enhance exercise capacity in a daytime-dependent manner. An analysis of liver and muscle glycogen stores as well as muscle lipid utilization suggested that these daytime effects mostly relate to liver glycogen levels and correspond to the animals' feeding behavior. Furthermore, given that exercise capacity responds to training, we tested the effect of training at different times of the day and found that training in the late compared with the early part of the active phase improves exercise performance. Overall, our findings suggest that clock proteins shape exercise capacity in a daytime-dependent manner through changes in liver glycogen levels, likely due to their effect on animals' feeding behavior.
Collapse
Affiliation(s)
- Yaarit Adamovich
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Vaishnavi Dandavate
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Saar Ezagouri
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Gal Manella
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Ziv Zwighaft
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Jonathan Sobel
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Yael Kuperman
- Department of Veterinary Resources, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Marina Golik
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Asher Auerbach
- Department of Chemical Research Support, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Maxim Itkin
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Sergey Malitsky
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Gad Asher
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel;
| |
Collapse
|
46
|
Abstract
Disruption of circadian rhythms increases the risk of several types of cancer. Mammalian cryptochromes (CRY1 and CRY2) are circadian transcriptional repressors that are related to DNA-repair enzymes. While CRYs lack DNA-repair activity, they modulate the transcriptional response to DNA damage, and CRY2 can promote SKP1 cullin 1-F-box (SCF)FBXL3-mediated ubiquitination of c-MYC and other targets. Here, we characterize five mutations in CRY2 observed in human cancers in The Cancer Genome Atlas. We demonstrate that two orthologous mutations of mouse CRY2 (D325H and S510L) accelerate the growth of primary mouse fibroblasts expressing high levels of c-MYC. Neither mutant affects steady-state levels of overexpressed c-MYC, and they have divergent impacts on circadian rhythms and on the ability of CRY2 to interact with SCFFBXL3 Unexpectedly, stable expression of either CRY2 D325H or of CRY2 S510L robustly suppresses P53 target-gene expression, suggesting that this may be a primary mechanism by which they influence cell growth.
Collapse
|
47
|
Kinouchi K, Mikami Y, Kanai T, Itoh H. Circadian rhythms in the tissue-specificity from metabolism to immunity; insights from omics studies. Mol Aspects Med 2021; 80:100984. [PMID: 34158177 DOI: 10.1016/j.mam.2021.100984] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/04/2021] [Accepted: 06/07/2021] [Indexed: 12/31/2022]
Abstract
Creatures on earth have the capacity to preserve homeostasis in response to changing environments. The circadian clock enables organisms to adapt to daily predictable rhythms in surrounding conditions. In mammals, circadian clocks constitute hierarchical network, where the central pacemaker in hypothalamic suprachiasmatic nucleus (SCN) serves as a time-keeping machinery and governs peripheral clocks in every other organ through descending neural and humoral factors. The central clock in SCN is reset by light, whilst peripheral clocks are entrained by feeding-fasting rhythms, emphasizing the point that temporal patterns of nutrient availability specifies peripheral clock functions. Indeed, emerging evidence revealed various types of diets or timing of food intake reprogram circadian rhythms in a tissue specific manner. This advancement in understanding of mechanisms underlying tissue specific responsiveness of circadian oscillators to nutrients at the genomic and epigenomic levels is largely owing to employment of state-of-the-art technologies. Specifically, high-throughput transcriptome, proteome, and metabolome have provided insights into how genes, proteins, and metabolites behave over circadian cycles in a given tissue under a certain dietary condition in an unbiased fashion. Additionally, combinations with specialized types of sequencing such as nascent-seq and ribosomal profiling allow us to dissect how circadian rhythms are generated or obliterated at each step of gene regulation. Importantly, chromatin immunoprecipitation followed by deep sequencing methods provide chromatin landscape in terms of regulatory mechanisms of circadian gene expression. In this review, we outline recent discoveries on temporal genomic and epigenomic regulation of circadian rhythms, discussing entrainment of the circadian rhythms by feeding as a fundamental new comprehension of metabolism and immune response, and as a potential therapeutic strategy of metabolic and inflammatory diseases.
Collapse
Affiliation(s)
- Kenichiro Kinouchi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| | - Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, 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, 160-8582, Japan
| |
Collapse
|
48
|
Gabriel BM, Zierath JR. Zeitgebers of skeletal muscle and implications for metabolic health. J Physiol 2021; 600:1027-1036. [PMID: 33963554 DOI: 10.1113/jp280884] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/04/2021] [Indexed: 12/25/2022] Open
Abstract
Metabolic health is a crucial area of current research, and is an outcome of innate physiology, and interactions with the environment. Environmental cues, such as the Earth's day-night rhythm, partly regulate diurnal hormones and metabolites. Circadian physiology consists of highly conserved biological processes over ∼24-h cycles, which are influenced by external cues (Zeitgebers - 'time-keepers'). Skeletal muscle has diurnal variations of a large magnitude, owing in part to the strong nature of physical activity throughout the day and other external Zeitgebers. The orchestration of whole-body and skeletal muscle metabolism is a complex, finely tuned process, and molecular diurnal variations are regulated by a transcription-translation feedback loop controlled by the molecular clock, as well as non-transcriptional metabolic processes. The mitochondrion may play an important role in regulating diurnal metabolites within skeletal muscle, given its central role in the regulation of NAD+ /NADH, O2 , reactive oxygen species and redox metabolism. These molecular pathways display diurnal variation and illustrate the complex orchestration of circadian metabolism in skeletal muscle. Probably the most robust Zeitgeber of skeletal muscle is exercise, which alters glucose metabolism and flux, in addition to a range of other diurnal metabolic pathways. Indeed, performing exercise at different times of the day may alter metabolism and health outcomes in some cohorts. The objective of this Symposium Review is to briefly cover the current literature, and to speculate regarding future areas of research. Thus, we postulate that metabolic health may be optimized by altering the timing of external cues such as diet and exercise.
Collapse
Affiliation(s)
- Brendan M Gabriel
- Aberdeen Cardiovascular & Diabetes Centre, The Rowett Institute, University of Aberdeen, Aberdeen, UK.,Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
49
|
Assessment of Selected Clock Proteins (CLOCK and CRY1) and Their Relationship with Biochemical, Anthropometric, and Lifestyle Parameters in Hypertensive Patients. Biomolecules 2021; 11:biom11040517. [PMID: 33808431 PMCID: PMC8067097 DOI: 10.3390/biom11040517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 03/29/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Circadian rhythms misalignment is associated with hypertension. The aim of the study was to evaluate the concentration of selected clock proteins-cryptochrome 1 (CRY1) and circadian locomotor output cycles kaput (CLOCK) to determine their relationships with biochemical and anthropometric parameters and lifestyle elements (diet, physical activity, and quality of sleep) in hypertensive patients. METHODS In 31 females with hypertension (HT) and 55 non-hypertensive women (NHT) the CRY1 and CLOCK concentrations, total antioxidant status (TAS), lipid profile, and glycemia were analyzed. Blood pressure and anthropometric measurements, nutritional, exercise, and sleep analyses were performed. RESULTS In the HT group, the CRY1 level was 37.38% lower than in the NHT group. No differences were noted in CLOCK concentration between groups. BMI, FBG, and TG were higher in the HT group compared to the NHT group, while TC, LDL, and HDL levels were similar. The study showed no relationship between CRY1 or CLOCK concentrations and glucose or lipids profile, amount of physical activity, or sleep quality, although CRY1 was associated with some anthropometric indicators. In the HT group, increased CLOCK and CRY1 values were associated with a high TAS level. CONCLUSIONS The serum level of CRY1 could be considered in a detailed diagnostic of hypertension risk in populations with abnormal anthropometric indices.
Collapse
|
50
|
Adamovich Y, Ezagouri S, Dandavate V, Asher G. Monitoring daytime differences in moderate intensity exercise capacity using treadmill test and muscle dissection. STAR Protoc 2021; 2:100331. [PMID: 33598660 PMCID: PMC7868630 DOI: 10.1016/j.xpro.2021.100331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There is growing interest in medicine and sports in uncovering exercise modifiers that enhance or limit exercise capacity. Here, we detail a protocol for testing the daytime effect on running capacity in mice using a moderate intensity treadmill effort test. Instructions for dissecting soleus, gastrocnemius plantaris, and quadriceps muscles for further analysis are provided as well. This experimental setup is optimized for addressing questions regarding the involvement of daytime and circadian clocks in regulating exercise capacity. For complete details on the use and execution of this protocol, please refer to Ezagouri et al. (2019). Exercise capacity is influenced by the time of day Protocol for determining moderate intensity exercise capacity using treadmill test Instructions for muscle dissection
Collapse
Affiliation(s)
- Yaarit Adamovich
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Saar Ezagouri
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Vaishnavi Dandavate
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Gad Asher
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| |
Collapse
|