1
|
Capone F, Vacca A, Bidault G, Sarver D, Kaminska D, Strocchi S, Vidal-Puig A, Greco CM, Lusis AJ, Schiattarella GG. Decoding the Liver-Heart Axis in Cardiometabolic Diseases. Circ Res 2025; 136:1335-1362. [PMID: 40403112 DOI: 10.1161/circresaha.125.325492] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
The liver and heart are closely interconnected organs, and their bidirectional interaction plays a central role in cardiometabolic disease. In this review, we summarize current evidence linking liver dysfunction-particularly metabolic dysfunction-associated steatotic liver disease, alcohol-associated liver disease, and cirrhosis-with an increased risk of heart failure and other cardiovascular diseases. We discuss how these liver conditions contribute to cardiac remodeling, systemic inflammation, and hemodynamic stress and how cardiac dysfunction in turn impairs liver perfusion and promotes hepatic injury. Particular attention is given to the molecular mediators of liver-heart communication, including hepatokines and cardiokines, as well as the emerging role of advanced research methodologies, including omics integration, proximity labeling, and organ-on-chip platforms, that are redefining our understanding of interorgan cross talk. By integrating mechanistic insights with translational tools, this review aims to support the development of multiorgan therapeutic strategies for cardiometabolic disease.
Collapse
Affiliation(s)
- Federico Capone
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Department of Medicine, Unit of Internal Medicine III, Padua University Hospital, University of Padua, Padova, Italy (F.C.)
- Department of Biomedical Sciences, University of Padova, Italy (F.C.)
| | - Antonio Vacca
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Clinica Medica, Department of Medicine, University of Udine, Italy (A.V.)
| | - Guillaume Bidault
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, United Kingdom (G.B., A.V.-P.)
| | - Dylan Sarver
- Division of Cardiology, Department of Medicine (D.S., D.K., A.J.L.), University of California, Los Angeles
- Department of Microbiology, Immunology and Molecular Genetics (D.S., A.J.L.), University of California, Los Angeles
- Department of Human Genetics (D.S., A.J.L.), University of California, Los Angeles
| | - Dorota Kaminska
- Division of Cardiology, Department of Medicine (D.S., D.K., A.J.L.), University of California, Los Angeles
| | - Stefano Strocchi
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Deutsches Herzzentrum der Charité, Charité-Universitätsmedizin Berlin, Germany (S.S., G.G.S.)
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, United Kingdom (G.B., A.V.-P.)
- Centro de Investigacion Principe Felipe, Valencia, Spain (A.V.-P.)
| | - Carolina M Greco
- Department of Biomedical Sciences, Humanitas University, Milan, Italy (C.M.G.)
- IRCCS Humanitas Research Hospital, Milan, Italy (C.M.G.)
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine (D.S., D.K., A.J.L.), University of California, Los Angeles
- Department of Microbiology, Immunology and Molecular Genetics (D.S., A.J.L.), University of California, Los Angeles
- Department of Human Genetics (D.S., A.J.L.), University of California, Los Angeles
| | - Gabriele G Schiattarella
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Deutsches Herzzentrum der Charité, Charité-Universitätsmedizin Berlin, Germany (S.S., G.G.S.)
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany (G.G.S.)
- Friede Springer Cardiovascular Prevention Center at Charité-Universitätsmedizin Berlin, Germany (G.G.S.)
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (G.G.S.)
| |
Collapse
|
2
|
Tong CY, Li C, Hurni C, Jacq A, Nie XY, Guy CR, Suh JH, Wong RKW, Merlin C, Naef F, Menet JS, Jiang Y. Single-Cell Multiomic Analysis of Circadian Rhythmicity in Mouse Liver. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.647044. [PMID: 40291723 PMCID: PMC12026578 DOI: 10.1101/2025.04.03.647044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
From bacteria to humans, most organisms showcase inherent 24-hour circadian rhythms, best exemplified by the sleep-wake cycle. These rhythms are remarkably widespread, governing hormonal, metabolic, physiological, and behavioral oscillations, and are driven by "molecular clocks" that orchestrate the rhythmic expression of thousands of genes throughout the body. Here, we generate single-cell RNA and ATAC multiomic data to simultaneously characterize gene expression and chromatin accessibility of ~33,000 mouse liver cells across the 24-hour day. Our study yields several key insights, including: (i) detecting circadian rhythmicity in both discretized liver cell types and transient sub-lobule cell states, capturing space-time RNA and ATAC profiles in a cell-type- and cell-state-specific manner; (ii) delving beyond mean cyclic patterns to characterize distributions, accounting for gene expression stochasticity due to transcriptional bursting; (iii) interrogating multimodal circadian rhythmicity, encompassing RNAs, DNA regulatory elements, and transcription factors (TFs), while examining priming and lagging effects across modalities; and (iv) inferring spatiotemporal gene regulatory networks involving target genes, TFs, and cis-regulatory elements that controls circadian rhythmicity and liver physiology. Our findings apply to existing single-cell data of mouse and Drosophila brains and are further validated by time-series single molecule fluorescence in situ hybridization, as well as vast amounts of existing and orthogonal high-throughput data from chromatin immunoprecipitation followed by sequencing, capture Hi-C, and TF knockout experiments. Altogether, our study constructs a comprehensive map of the time-series transcriptomic and epigenomic landscapes that elucidate the function and mechanism of the liver peripheral clocks.
Collapse
|
3
|
Lin Q, Du X, Ren F, Liu Y, Gong G, Ge S, Li W, Li Z, Zhou L, Duan M, Li XY, Wang GZ, Xiao R, Gui JF, Mei J. Anti-Müllerian hormone signalling sustains circadian homeostasis in zebrafish. Nat Commun 2025; 16:4359. [PMID: 40348785 PMCID: PMC12065890 DOI: 10.1038/s41467-025-59528-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 04/28/2025] [Indexed: 05/14/2025] Open
Abstract
Circadian clocks temporally orchestrate the behavioural and physiological rhythms. The core molecules establishing the circadian clock are clear; however, the critical signalling pathways that cause or favour the homeostasis are poorly understood. Here, we report that anti-Müllerian hormone (Amh)-mediated signalling plays an important role in sustaining circadian homeostasis in zebrafish. Notably, amh knockout dampens molecular clock oscillations and disrupts both behavioural and hormonal circadian rhythms, which are recapitulated in bmpr2a null mutants. Somatotropes and gonadotropes are identified as Amh-targeted pituitary cell populations. Single-cell transcriptome analysis further reveals a lineage-specific regulation of pituitary clock by Amh. Moreover, Amh-induced effect on clock gene expression can be abolished by blocking Smad1/5/9 phosphorylation and bmpr2a knockout. Mechanistically, Amh binds to its receptors, Bmpr2a/Bmpr1bb, which in turn activate Smad1/5/9 by phosphorylation and promote circadian gene expression. Our findings reveal a key hormone signalling pathway for circadian homeostasis in zebrafish with implications for rhythmic organ functions and circadian health.
Collapse
Affiliation(s)
- Qiaohong Lin
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, University of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Xian Du
- Department of Hematology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Department of Laboratory, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Ren
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, University of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Fisheries, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Ying Liu
- College of Fisheries, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Gaorui Gong
- College of Fisheries, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Si Ge
- College of Fisheries, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Weiwei Li
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, University of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Zhi Li
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, University of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Li Zhou
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, University of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Ming Duan
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, University of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Xi-Yin Li
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, University of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Guang-Zhong Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui Xiao
- Department of Hematology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| | - Jian-Fang Gui
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, University of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.
- College of Fisheries, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China.
| | - Jie Mei
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, University of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.
- College of Fisheries, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
4
|
Chen Y, Liu P, Sabo A, Guan D. Human genetic variation determines 24-hour rhythmic gene expression and disease risk. Nat Commun 2025; 16:4270. [PMID: 40341583 PMCID: PMC12062405 DOI: 10.1038/s41467-025-59524-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 04/24/2025] [Indexed: 05/10/2025] Open
Abstract
24-hour biological rhythms are essential to maintain physiological homeostasis. Disruption of these rhythms increases the risks of multiple diseases. Biological rhythms are known to have a genetic basis formed by core clock genes, but how individual genetic variation shapes the oscillating transcriptome and contributes to human chronophysiology and disease risk is largely unknown. Here, we mapped interactions between temporal gene expression and genotype to identify quantitative trait loci (QTLs) contributing to rhythmic gene expression. These newly identified QTLs were termed as rhythmic QTLs (rhyQTLs), which determine previously unappreciated rhythmic genes in human subpopulations with specific genotypes. Functionally, rhyQTLs and their associated rhythmic genes contribute extensively to essential chronophysiological processes, including bile acid and lipid metabolism. The identification of rhyQTLs sheds light on the genetic mechanisms of gene rhythmicity, offers mechanistic insights into variations in human disease risk, and enables precision chronotherapeutic approaches for patients.
Collapse
Affiliation(s)
- Ying Chen
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Panpan Liu
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Aniko Sabo
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Dongyin Guan
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
5
|
Gaspar LS, Pyakurel S, Xu N, D'Souza SP, Koritala BSC. Circadian Biology in Obstructive Sleep Apnea-Associated Cardiovascular Disease. J Mol Cell Cardiol 2025; 202:116-132. [PMID: 40107345 DOI: 10.1016/j.yjmcc.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/16/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
A dysregulated circadian system is independently associated with both Obstructive Sleep Apnea (OSA) and cardiovascular disease (CVD). OSA and CVD coexistence is often seen in patients with prolonged untreated OSA. However, the role of circadian dysregulation in their relationship is unclear. Half of the human genes, associated biological pathways, and physiological functions exhibit circadian rhythms, including blood pressure and heart rate regulation. Mechanisms related to circadian dysregulation and heart function are potentially involved in the coexistence of OSA and CVD. In this article, we provide a comprehensive overview of circadian dysregulation in OSA and associated CVD. We also discuss feasible animal models and new avenues for future research to understand their relationship. Oxygen-sensing pathways, inflammation, dysregulation of cardiovascular processes, oxidative stress, metabolic regulation, hormone signaling, and epigenetics are potential clock-regulated mechanisms connecting OSA and CVD.
Collapse
Affiliation(s)
- Laetitia S Gaspar
- Centre for Neuroscience and Cell Biology, University of Coimbra, Portugal; Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
| | - Santoshi Pyakurel
- Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Na Xu
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Shane P D'Souza
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Bala S C Koritala
- Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America; Department of Otolaryngology-Head and Neck Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America.
| |
Collapse
|
6
|
Ruan W, Li T, Bang IH, Lee J, Deng W, Ma X, Luo C, Du F, Yoo SH, Kim B, Li J, Yuan X, Figarella K, An YA, Wang YY, Liang Y, DeBerge M, Zhang D, Zhou Z, Wang Y, Gorham JM, Seidman JG, Seidman CE, Aranki SF, Nair R, Li L, Narula J, Zhao Z, Gorfe AA, Muehlschlegel JD, Tsai KL, Eltzschig HK. BMAL1-HIF2A heterodimer modulates circadian variations of myocardial injury. Nature 2025; 641:1017-1028. [PMID: 40269168 DOI: 10.1038/s41586-025-08898-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/14/2025] [Indexed: 04/25/2025]
Abstract
Acute myocardial infarction is a leading cause of morbidity and mortality worldwide1. Clinical studies have shown that the severity of cardiac injury after myocardial infarction exhibits a circadian pattern, with larger infarcts and poorer outcomes in patients experiencing morning-onset events2-7. However, the molecular mechanisms underlying these diurnal variations remain unclear. Here we show that the core circadian transcription factor BMAL17-11 regulates circadian-dependent myocardial injury by forming a transcriptionally active heterodimer with a non-canonical partner-hypoxia-inducible factor 2 alpha (HIF2A)12-16-in a diurnal manner. To substantiate this finding, we determined the cryo-EM structure of the BMAL1-HIF2A-DNA complex, revealing structural rearrangements within BMAL1 that enable cross-talk between circadian rhythms and hypoxia signalling. BMAL1 modulates the circadian hypoxic response by enhancing the transcriptional activity of HIF2A and stabilizing the HIF2A protein. We further identified amphiregulin (AREG)16,17 as a rhythmic target of the BMAL1-HIF2A complex, critical for regulating daytime variations of myocardial injury. Pharmacologically targeting the BMAL1-HIF2A-AREG pathway provides cardioprotection, with maximum efficacy when aligned with the pathway's circadian phase. These findings identify a mechanism governing circadian variations of myocardial injury and highlight the therapeutic potential of clock-based pharmacological interventions for treating ischaemic heart disease.
Collapse
Affiliation(s)
- Wei Ruan
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA.
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, China.
| | - Tao Li
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - In Hyuk Bang
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Jaewoong Lee
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT, USA
| | - Wankun Deng
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xinxin Ma
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Cong Luo
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Fang Du
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Boyun Kim
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Major in Aquaculture and Applied Life Sciences, College of Fisheries Science, Pukyong National University, Busan, Republic of Korea
| | - Jiwen Li
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Department of Cardiac Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyi Yuan
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Katherine Figarella
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Yin-Ying Wang
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yafen Liang
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Center for Outcomes Research, UTHealth Houston, Houston, TX, USA
| | - Matthew DeBerge
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Dongze Zhang
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Zhen Zhou
- Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Yanyu Wang
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Joshua M Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | | | | | - Sary F Aranki
- Department of Surgery, Division of Cardiac Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ragini Nair
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Lei Li
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jagat Narula
- Division of Cardiology, Department of Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Memorial Hermann Hospital, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alemayehu A Gorfe
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Jochen D Muehlschlegel
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kuang-Lei Tsai
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA.
- MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA.
| | - Holger K Eltzschig
- Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA.
- Center for Outcomes Research, UTHealth Houston, Houston, TX, USA.
| |
Collapse
|
7
|
Dong Y, Lam SM, Li Y, Li MD, Shui G. The circadian clock at the intersection of metabolism and aging - emerging roles of metabolites. J Genet Genomics 2025:S1673-8527(25)00123-7. [PMID: 40306487 DOI: 10.1016/j.jgg.2025.04.014] [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: 04/06/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025]
Abstract
The circadian clock is a highly hierarchical network of endogenous pacemakers that primarily maintains and directs oscillations through transcriptional and translational feedback loops, which modulates an approximately 24-hour cycle of endocrine and metabolic rhythms within cells and tissues. While circadian clocks regulate metabolic processes and related physiology, emerging evidence indicates that metabolism and circadian rhythm are intimately intertwined. In this review, we highlight the concept of metabolites, including lipids and other polar metabolites generated from intestinal microbial metabolism and nutrient intake, as circadian pacemakers that drive changes in circadian rhythms, which in turn influence metabolism and aging. Furthermore, we discuss the roles of functional metabolites as circadian pacemakers, paving a new direction on potential intervention targets of circadian disruption, pathological aging, as well as metabolic diseases that are clinically important.
Collapse
Affiliation(s)
- Yue Dong
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; LipidALL Technologies Company Limited, Changzhou, Jiangsu 213022, China
| | - Yan Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China.
| | - Min-Dian Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, MOE Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China.
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of the Chinese Academy of Sciences, Beijing 100049, China; Guangzhou National Laboratory, Guangzhou, Guangdong 510005, China.
| |
Collapse
|
8
|
Liu J, Xia P, Qu Y, Zhang X, Shen R, Yang P, Tan H, Chen H, Deng Y. Long-Term Exposure to Environmentally Realistic Doses of Starch-Based Microplastics Suggests Widespread Health Effects. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9867-9878. [PMID: 40202198 DOI: 10.1021/acs.jafc.4c10855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
There is a growing consensus on addressing the global plastic pollution problem by advocating for bioplastics. While starch-based plastics are prevalent, the potential health implications of starch-based microplastics (SMPs) remain largely unexplored. This is particularly concerning given their potential for accidental ingestion and subsequent interference with blood glucose metabolism. Our research provides the first investigation into the distribution and adverse effects of long-term exposure to environmentally relevant doses of SMPs in female mice, approximately 14-81 particles per mouse per day. After three months of exposure, SMPs were found to infiltrate the liver, intestine, and ovarian tissues, causing microstructural lesions. Exposure to SMPs also resulted in elevated blood glucose levels, increased hepatic oxidative stress, and disrupted lipid metabolism. A multiomics analysis further uncovered abnormalities in gene expression and microbiota, as well as enriched pathways related to insulin regulation and circadian rhythms in the exposed mice. Our results indicate that prolonged exposure to environmentally relevant doses of SMPs can have widespread health effects in mice, potentially disrupting circadian rhythms by inducing insulin resistance. This suggests that the safety of bioplastics requires further evaluation before their large-scale application in food packages.
Collapse
Affiliation(s)
- Jing Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Peng Xia
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Yi Qu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Xue Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Ruqin Shen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
- Department of Environmental Science and Engineering, Fudan University, Shanghai 200438, China
| | - Pan Yang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Hongli Tan
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Hexia Chen
- School of Environment and Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, China
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yongfeng Deng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| |
Collapse
|
9
|
Smyllie NJ, Koch AA, Adamson AD, Patton AP, Johnson A, Bagnall JS, Johnson O, Meng QJ, Loudon ASI, Hastings MH. Quantitative measures of clock protein dynamics in the mouse suprachiasmatic nucleus extends the circadian time-keeping model. EMBO J 2025:10.1038/s44318-025-00426-z. [PMID: 40247113 DOI: 10.1038/s44318-025-00426-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 04/19/2025] Open
Abstract
The suprachiasmatic nucleus (SCN) synchronises circadian rhythmicity (~24 h) across the body. The SCN cell-autonomous clock is modelled qualitatively as a transcriptional-translational feedback loop (TTFL), with heteromeric complexes of transcriptional activator and repressor proteins driving cyclical gene expression. How these proteins really behave within the SCN, individually and in relation to each other, is poorly understood. Imaging SCN slices from a novel array of knock-in reporter mice, we quantify the dynamic behaviours of combined repressors PERIOD2 (PER2) and CRYPTOCHROME1 (CRY1), and activator BMAL1. We reveal a spectrum of protein-specific intracellular and spatiotemporal behaviours that run counter to the qualitative TTFL model. We also show that PER and CRY1 exert independent actions on TTFL oscillations, and that their individual stabilities play a critical role in SCN circadian dynamics. These results reveal a rich and unanticipated complexity in the dynamic behaviours and functions of endogenous circadian proteins, prompting re-appraisal of current transcriptional-translational feedback loop models of the suprachiasmatic nucleus.
Collapse
Affiliation(s)
- Nicola J Smyllie
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK.
| | - Alex A Koch
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Antony D Adamson
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Andrew P Patton
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Adam Johnson
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - James S Bagnall
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Olivia Johnson
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Qing-Jun Meng
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Andrew S I Loudon
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Michael H Hastings
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
10
|
Zhang Z, Luo Y, Zhang H, Zeng Z, Zheng W, Zhao Y, Huang Y, Shen L. Exploring the mechanisms of cow placental peptides in delaying liver aging based on mitochondrial energy metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119593. [PMID: 40064320 DOI: 10.1016/j.jep.2025.119593] [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: 11/19/2024] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Placenta is a kind of traditional Chinese medicine, known as "Ziheche". The role of cow placental peptides (CPP) in delaying liver aging has been reported, and in-depth exploration of the specific regulatory mechanisms is of great significance for the recycling and utilization of CPP and the development of natural anti-aging drugs. AIM OF THE STUDY To investigate the protective effects and mechanisms of CPP on liver aging induced by D-galactose (D-gal) in mice from the perspective of mitochondrial energy metabolism. METHODS An aging model was induced in mice using D-gal. The body weight and liver index of mice were measured, followed by staining and electron microscopy to observe liver morphology and aging markers. Reactive oxygen species (ROS) levels and antioxidant-related indicators were assessed, and mitochondrial function was evaluated. Finally, changes and mechanisms in liver transcriptomics and targeted mitochondrial energy metabolomics were analyzed and integrated to elucidate the regulatory pathways through which CPP delays liver aging. RESULTS CPP improved liver structural damage, oxidative stress, and mitochondrial dysfunction induced by D-galactose in aging mice. It increased the final body weight and liver index, alleviated hepatocyte swelling and degeneration, enhanced liver antioxidant capacity, and restored normal mitochondrial morphology and function. The combined analysis of targeted mitochondrial energy metabolomics and liver transcriptomics revealed that CPP directly or indirectly regulated mitochondrial energy metabolism and delayed aging by influencing the cAMP signaling pathway, PI3K-Akt signaling pathway, oxidative phosphorylation, and other pathways, thereby modulating related genes and metabolites.
Collapse
Affiliation(s)
- Zeru Zhang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yuxin Luo
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Hanwen Zhang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhi Zeng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Weijian Zheng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yuquan Zhao
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yixin Huang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Liuhong Shen
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
11
|
Shanmugam DAS, Balaraman AD, Kar A, Franco A, Balaji BAC, Meenakumari S, Praveenkumar PK, Gayathri R, Ganesan VK, Kumar MV, Senthilkumar K, Shanthi B. Mini review: Bidirectional Regulation of Circadian Rhythm by Suprachiasmatic Nucleus and Nuclear Receptors in Female Mammals. J Circadian Rhythms 2025; 23:4. [PMID: 40225034 PMCID: PMC11987856 DOI: 10.5334/jcr.245] [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: 07/02/2024] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
The anterior region of the hypothalamus accommodates a bilateral structure called the suprachiasmatic nucleus (SCN), which controls, modulates, and perpetuates the homeostasis of circadian rhythm and sleep hormone release. These SCN have a predominance over multitudinous peripheral tissues like the uterus, liver, intestine, pancreas, endocrine system, immune system, reproductive system, and cardiovascular system. This peripheral clock acts as a pacemaker for circadian rhythm timing, which regulates crucial metabolic pathways and organizes numerous activities in the female reproductive network of mammals. The circadian CLOCK genes are expressed in various reproductive organs. The CLOCK, BMAL1, CRY, and PER genes harmonize the balance and manifestation of nuclear receptors (NRs) expression, and the other way round, NRs regulate these circadian genes. Several NRs, in particular estrogen, progesterone, androgen, and PPARs, nurture the ovary and uterus. Bidirectional coordination between SCN and NRs maintains the circadian rhythm of the hypothalamic-pituitary-gonadal (HPG) axis of the female reproductive organs.
Collapse
Affiliation(s)
- Dharani Abirama Sundari Shanmugam
- Department of Endocrinology, Dr. ALM. PG. Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai – 600113, Tamil Nadu, India
| | - Ashwini Devi Balaraman
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur – 603202, Tamil Nadu, India
| | - Abhijit Kar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur – 603202, Tamil Nadu, India
| | - Abishek Franco
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur – 603202, Tamil Nadu, India
| | | | - S. Meenakumari
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur – 603202, Tamil Nadu, India
| | - P. K. Praveenkumar
- Department of Biotechnology, Sri Venkateswara College of Engineering, Sriperumbudur Tk – 602117, Tamil Nadu, India
| | - R. Gayathri
- Department of Biotechnology, St Joseph’s College of Engineering, Old Mahabalipuram Road, Kamaraj Nagar, Semmancheri, Chennai – 600119, Tamil Nadu, India
| | - Vinoth Kumar Ganesan
- Department of Health Research (DHR-ICMR), Multi-Disciplinary Research Unit (MRU), Rangaraya Medical College, Kakinada – 533003, Andhra Pradesh, India
| | - Merugumolu Vijay Kumar
- Department of Pharmacology, Dayananda Sagar University, Bengaluru – 560078, Karnataka, India
| | - K. Senthilkumar
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region of China
| | - B. Shanthi
- Department of Biotechnology, JAASB Institute and Research Academia, Valasaravakkam, Chennai – 600087, Tamil Nadu, India
| |
Collapse
|
12
|
Spangler RK, Jonnalagadda K, Ward JD, Partch CL. A wrinkle in timers: evolutionary rewiring of conserved biological timekeepers. Trends Biochem Sci 2025; 50:344-355. [PMID: 39952882 DOI: 10.1016/j.tibs.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/17/2025]
Abstract
Biological timing mechanisms are intrinsic to all organisms, orchestrating the temporal coordination of biological events through complex genetic networks. Circadian rhythms and developmental timers utilize distinct timekeeping mechanisms. This review summarizes the molecular basis for circadian rhythms in mammals and Drosophila, and recent work leveraging these clocks to understand temporal regulation in Caenorhabditis elegans development. We describe the evolutionary connections between distinct timing mechanisms and discuss recent insights into the rewiring of core clock components in development. By integrating findings from circadian and developmental studies with biochemical and structural analyses of conserved components, we aim to illuminate the molecular basis of nematode timing mechanisms and highlight broader insights into biological timing across species.
Collapse
Affiliation(s)
- Rebecca K Spangler
- Department of Chemistry and Biochemistry, University of California - Santa Cruz, Santa Cruz, CA 95064, USA
| | - Keya Jonnalagadda
- Department of Molecular, Cell, and Developmental Biology, University of California - Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jordan D Ward
- Department of Molecular, Cell, and Developmental Biology, University of California - Santa Cruz, Santa Cruz, CA 95064, USA
| | - Carrie L Partch
- Department of Chemistry and Biochemistry, University of California - Santa Cruz, Santa Cruz, CA 95064, USA; Center for Circadian Biology, University of California - Santa Diego, La Jolla, CA 92093, USA; Howard Hughes Medical Institute, University of California - Santa Cruz, Santa Cruz, CA 95064, USA.
| |
Collapse
|
13
|
Akyel YK, Seyhan NO, Gül Ş, Çelik M, Taşkın AC, Selby CP, Sancar A, Kavakli IH, Okyar A. The impact of circadian rhythm disruption on oxaliplatin tolerability and pharmacokinetics in Cry1 -/-Cry2 -/- mice under constant darkness. Arch Toxicol 2025; 99:1417-1429. [PMID: 39903276 PMCID: PMC11968489 DOI: 10.1007/s00204-025-03968-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Circadian rhythms, the 24-h oscillations of biological activities guided by the molecular clock, play a pivotal role in regulating various physiological processes in organisms. The intricate relationship between the loss of circadian rhythm and its influence on the tolerability and pharmacokinetic properties of anticancer drugs is poorly understood. In our study, we investigated the effects of oxaliplatin, a commonly used anticancer drug, on Cry1-/- and Cry2-/- mice (Cry DKO mice) under darkness conditions, where they exhibit free-running phenotype. We administered oxaliplatin at a dosage of 12 mg/kg/day at two distinct circadian times, CT8 and CT16, under constant darkness conditions to Cry DKO mice and their wild type littermates. Our results revealed a striking disparity in oxaliplatin tolerance between Cry DKO mice and their wild-type counterparts. Oxaliplatin exhibited severe toxicity in Cry DKO mice at both CT8 and CT16, in contrast to the wild type mice. Pharmacokinetic analyses suggested that such toxicity was a result of high concentrations of oxaliplatin in the serum and liver of Cry DKO mice after repeated dose injections. To understand the molecular basis of such intolerance, we performed RNA-seq studies using mouse livers. Our findings from the RNA-seq analysis highlighted the substantial impact of circadian rhythm disruption on gene expression, particularly affecting genes involved in detoxification and xenobiotic metabolism, such as the Gstm gene family. This dysregulation in detoxification pathways in Cry DKO mice likely contributes to the increased toxicity of oxaliplatin. In conclusion, our study highlights the crucial role of an intact molecular clock in dictating the tolerability of oxaliplatin. These findings emphasize the necessity of considering circadian rhythms in the administration of anticancer drugs, providing valuable insights into optimizing treatment strategies for cancer patients.
Collapse
Affiliation(s)
- Yasemin Kubra Akyel
- Department of Medical Pharmacology, School of Medicine, Istanbul Medipol University, Istanbul, Türkiye
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, TR-34116, Beyazit-Istanbul, Türkiye
| | - Narin Ozturk Seyhan
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, TR-34116, Beyazit-Istanbul, Türkiye
| | - Şeref Gül
- Biotechnology Division, Department of Biology, Faculty of Science, Istanbul University, Istanbul, Türkiye
- Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Istanbul, Türkiye
| | - Melis Çelik
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Türkiye
| | - Ali Cihan Taşkın
- Department of Laboratory Animal Science, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Türkiye
- Translational Medicine Research Center, Experimental Animals Laboratory, Koc University, Istanbul, Türkiye
| | - Christopher P Selby
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Aziz Sancar
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Ibrahim Halil Kavakli
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Türkiye.
- Department of Chemical and Biological Engineering, Koc University, Istanbul, Türkiye.
| | - Alper Okyar
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, TR-34116, Beyazit-Istanbul, Türkiye.
| |
Collapse
|
14
|
Hunter AL, Bechtold DA. The metabolic significance of peripheral tissue clocks. Commun Biol 2025; 8:497. [PMID: 40140664 PMCID: PMC11947457 DOI: 10.1038/s42003-025-07932-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
The circadian clock is a transcriptional-translational feedback loop which oscillates in virtually all nucleated cells of the body. In the decades since its discovery, it has become evident that the molecular clockwork is inextricably linked to energy metabolism. Given the frequency with which metabolic dysfunction and clock disruption co-occur, understanding why and how clock and metabolic processes are reciprocally coupled will have important implications for supporting human health and wellbeing. Here, we discuss the relevance of molecular clock function in metabolic tissues and explore its role not only as a driver of day-night variation in gene expression, but as a key mechanism for maintaining metabolic homeostasis in the face of fluctuating energy supply and demand.
Collapse
Affiliation(s)
- A Louise Hunter
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK.
- Diabetes, Endocrinology & Metabolism Centre, Oxford Road Campus, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK.
| | - David A Bechtold
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
15
|
Zhu W, Gong A, Zhang B, Cheng H, Huang L, Wu X, Zhang D, Dai W, Li S, Xu H. The Chronobiological and Neuroprotective Mechanisms of Resveratrol in Improving Sleep. Mediators Inflamm 2025; 2025:4954030. [PMID: 40144750 PMCID: PMC11944795 DOI: 10.1155/mi/4954030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/01/2025] [Indexed: 03/28/2025] Open
Abstract
According to statistics, more than one-third of the global population currently experiences sleep problems, and about 10% of adults have been diagnosed with insomnia, a proportion that is increasing annually. Most currently used insomnia medications are not specifically developed but are discovered by chance, often resulting in unavoidable side effects like addiction. Thus, there is an urgent need to find safer and more effective therapeutic options. Resveratrol, a natural polyphenolic compound, shows significant potential in improving insomnia. Research shows that its effects may be achieved through multiple biological processes, including antiapoptosis, antioxidant activity, anti-inflammation, circadian rhythm regulation, modulation of neurotransmitters (gamma-aminobutyric acid (GABA), DA, 5-HT, cortisol), and increased levels of neurotrophic factor BDNF. Additionally, resveratrol's treatment of insomnia is closely linked to the SIRT1, AMPK, NF-κB, mTOR, PI3K/Akt, and MAPK pathways. This review summarizes the mechanisms of resveratrol in treating insomnia to provide researchers with a deeper understanding of its action, which can aid in the development of novel targeted drugs and offer innovative ideas and methods for clinical insomnia treatment.
Collapse
Affiliation(s)
- Wenwen Zhu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Ailin Gong
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Bin Zhang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Hanxing Cheng
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Lishan Huang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiao Wu
- Department of Acupuncture and Tuina, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Dechou Zhang
- Department of Acupuncture and Tuina, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Wenbin Dai
- Department of Respiratory medicine, Luzhou Longmatan District People's Hospital, Luzhou, Sichuan, China
| | - Sen Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Houping Xu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
16
|
Ning G, Li BN, Wu H, Shi RB, Peng AJ, Wang HY, Zhou X. Regulation of testosterone synthesis by circadian clock genes and its research progress in male diseases. Asian J Androl 2025:00129336-990000000-00298. [PMID: 40101130 DOI: 10.4103/aja20258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/20/2025] [Indexed: 03/20/2025] Open
Abstract
ABSTRACT The circadian clock is an important internal time regulatory system for a range of physiological and behavioral rhythms within living organisms. Testosterone, as one of the most critical sex hormones, is essential for the development of the reproductive system, maintenance of reproductive function, and the overall health of males. The secretion of testosterone in mammals is characterized by distinct circadian rhythms and is closely associated with the regulation of circadian clock genes. Here we review the central and peripheral regulatory mechanisms underlying the influence of circadian clock genes upon testosterone synthesis. We also examined the specific effects of these genes on the occurrence, development, and treatment of common male diseases, including late-onset hypogonadism, erectile dysfunction, male infertility, and prostate cancer.
Collapse
Affiliation(s)
- Gang Ning
- The First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Bo-Nan Li
- Affiliated Changsha Hospital of Hunan Normal University, Changsha 410023, China
| | - Hui Wu
- The First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ruo-Bing Shi
- The First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - A-Jian Peng
- The First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Hao-Yu Wang
- The First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xing Zhou
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| |
Collapse
|
17
|
Tomatsu S, Abbott SM, Attarian H. Clinical Chronobiology: Circadian Rhythms in Health and Disease. Semin Neurol 2025. [PMID: 39961369 DOI: 10.1055/a-2538-3259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
Circadian rhythms (CRs) are entrainable endogenous rhythms that respond to external stimuli and regulate physiological functions. The suprachiasmatic nucleus (SCN) in the hypothalamus is the mammalian master clock that synchronizes all other tissue-specific peripheral clocks, primarily through gamma-aminobutyric acid (GABA) and vasoactive intestinal polypeptide (VIP). The SCN follows Earth's 24-hour cycle by light entrainment through the retinohypothalamic tract. At the cellular level, the core clock genes CLOCK, BMAL1, PER1-PER3, CRY1, and CRY2 regulate CRs in a negative feedback loop. The circadian disruption of the sleep-wake cycle manifests in at least six distinct clinical conditions. These are the circadian rhythm sleep-wake disorders (CRSWDs). Their diagnosis is made by history, sleep diaries, and actigraphy. Treatment involves a combination of timed light exposure, melatonin/melatonin agonists, and behavioral interventions. In addition, CR disturbances and subsequent misalignment can increase the risk of a variety of illnesses. These include infertility and menstrual irregularities as well as diabetes, obesity, fatty liver disease, and other metabolic syndromes. In addition, a disruption in the gut microbiome creates a proinflammatory environment. CR disturbances increase the risk for mood disorders, hence the utility of light-based therapies in depression. People with neurodegenerative disorders demonstrate significant disturbances in their CRs, and in their sleep-wake cycles. Circadian realignment therapies can also help decrease the symptomatic burden of these disorders. Certain epilepsy syndromes, such as juvenile myoclonic epilepsy (JME), have a circadian pattern of seizures. Circadian disturbances in epilepsy can be both the consequence and cause for breakthrough seizures. The immune system has its own CR. Disturbances in these due to shift work, for instance, can increase the risk of infections. CR disturbances can also increase the risk of cancer by impacting DNA repair, apoptosis, immune surveillance, and cell cycle regulation. Moreover, the timing of chemotherapeutic agents has been shown to increase their therapeutic impact in certain cancers.
Collapse
Affiliation(s)
- Shizuka Tomatsu
- Department of Neurology, Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Sabra M Abbott
- Department of Neurology, Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Hrayr Attarian
- Department of Neurology, Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
18
|
Xu X, Xu L, Lang Z, Sun G, Pan J, Li X, Bian Z, Wu X. Identification of potential susceptibility loci for non-small cell lung cancer through whole genome sequencing in circadian rhythm genes. Sci Rep 2025; 15:7825. [PMID: 40050692 PMCID: PMC11885630 DOI: 10.1038/s41598-025-92083-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
Lung cancer is a malignant tumor with a high morbidity and mortality rate worldwide, causing an increasing disease burden. Of these, the most common type is non-small cell lung cancer (NSCLC), which accounts for 80-85% of all lung cancer cases. Genetic research is crucial for continuously discovering susceptibility genes related to lung cancer for in-depth study. The role of genetic predisposition in the development of NSCLC, particularly within circadian rhythm pathways known to govern various physiological processes, is increasingly acknowledged. Yet, the association between genetic variants of circadian rhythm-related genes and NSCLC susceptibility among Chinese populations is not fully understood. This study carried out a two-phase (discovery and validation stages) research design to identify genetic variants associated with NSCLC risk within the circadian rhythm pathway. We employed extensive whole-genome sequencing (WGS) for 1,104 NSCLC cases and 9,635 controls. FastGWA-GLMM was used for single-locus risk association analysis of NSCLC, and we screened candidate SNPs in the validation set that comprised 4,444 cases and 174,282 controls from the Biobank Japan Project (BBJ). Furthermore, GCTA-COJO conditional analysis was utilized to confirm SNPs related to NSCLC risk. Finally, potential genetic variations that may regulate gene expression were explored in GTEx and QTLbase. RNA sequencing data were utilized for transcriptomic verification. Our study identified eight candidate SNPs associated with NSCLC susceptibility within the circadian rhythm pathway that met the requirement with P < 0.05 in both the discovery and validation populations. After conditional analysis, five of these SNPs remained. The A allele of CUL1 rs78524436 (ORmeta = 1.18, 95%CI: 1.09-1.29, Pmeta = 7.99e-5) and the A allele of TEF rs9611588 (ORmeta = 1.06, 95%CI: 1.02-1.10, Pmeta = 1.28e-3) were associated with an increased risk of NSCLC. The A allele of FBXL21 rs2069868 (ORmeta = 0.86, 95%CI: 0.80-0.96, Pmeta = 4.78e-4), the T allele of CSNK1D rs147316973 (ORmeta = 0.76, 95%CI: 0.65-0.88, Pmeta = 5.93e-4), and the A allele of RORA rs1589701 (ORmeta = 0.94, 95%CI: 0.91-0.98, Pmeta = 3.40e-3) were associated with a lower risk of NSCLC, separately. The eQTL results revealed an association between RORA rs1589701 and TEF rs9611588 with the expression levels of RORA and TEF, respectively. Transcriptome data indicated that RORA and TEF showed lower expression levels in tumor tissues compared to normal tissues (P < 0.001). Moreover, poorer survival was observed in patients with lower RORA and TEF expressions (log-rank P < 0.05). Our findings spotlight potential susceptibility loci within circadian rhythm pathway genes that modulate NSCLC carcinogenesis, which enriches the understanding of the genetic susceptibility of NSCLC in the Chinese population and provides a more solid basis for exploring the biological mechanism of circadian rhythm genes in NSCLC.
Collapse
Affiliation(s)
- Xiaohang Xu
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Intelligent Preventive Medicine, Hangzhou, 310058, China
| | - Luopiao Xu
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Intelligent Preventive Medicine, Hangzhou, 310058, China
| | - Zeyong Lang
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Gege Sun
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Junlong Pan
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xue Li
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Zhejiang Key Laboratory of Intelligent Preventive Medicine, Hangzhou, 310058, China
| | - Zilong Bian
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xifeng Wu
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Key Laboratory of Intelligent Preventive Medicine, Hangzhou, 310058, China.
- National Institute for Data Science in Health and Medicine, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
- School of Medicine and Health Science, George Washington University, Washington, DC, USA.
| |
Collapse
|
19
|
Zhang Y, Ding H, Huo Z, Chen L. A novel Bayesian hierarchical model for detecting differential circadian pattern in transcriptomic applications. Brief Bioinform 2025; 26:bbaf139. [PMID: 40234105 PMCID: PMC11998590 DOI: 10.1093/bib/bbaf139] [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: 12/10/2024] [Revised: 01/28/2025] [Accepted: 03/01/2025] [Indexed: 04/17/2025] Open
Abstract
Circadian rhythm plays a critical role in regulating various physiological processes, and disruptions in these rhythms have been linked to a wide range of diseases. Identifying molecular biomarkers showing differential circadian (DC) patterns between biological conditions or disease status is important for disease prevention, diagnosis, and treatment. However, circadian pattern is characterized by three key components: amplitude, phase, and MESOR, which poses a great challenge for DC analysis. Existing statistical methods focus on detecting differential shape (amplitude and phase) but often overlook MESOR difference. Additionally, these methods lack flexibility to incorporate external knowledge such as differential circadian information from similar clinical and biological context to improve the current DC analysis. To address these limitation, we introduce a novel Bayesian hierarchical model, BayesDCirc, designed for detecting differential circadian patterns in a two-group experimental design, which offer the advantage of testing MESOR difference and incorporating external knowledge. Benefiting from explicitly testing MESOR within the Bayesian modeling framework, BayesDCirc demonstrates superior FDR control over existing methods, with further performance improvement by leveraging external knowledge of DC genes. Applied to two real datasets, BayesDCirc successfully identify key circadian genes, particularly with external knowledge incorporated. The R package "BayesDCirc" for the method is publicly available on GitHub at https://github.com/lichen-lab/BayesDCirc.
Collapse
Affiliation(s)
- Yutao Zhang
- Department of Biostatistics, University of Florida, Gainesville, FL, 32603, United States
| | - Haocheng Ding
- Department of Biostatistics, Augusta University, Augusta, GA, 30912, United States
| | - Zhiguang Huo
- Department of Biostatistics, University of Florida, Gainesville, FL, 32603, United States
| | - Li Chen
- Department of Biostatistics, University of Florida, Gainesville, FL, 32603, United States
| |
Collapse
|
20
|
Sengupta S, Smith DF, Koritala BSC. Circadian Rhythms, Immune Regulation, and the Risk for Sepsis: Circadian Rhythms and Neonatal Care. Clin Perinatol 2025; 52:185-197. [PMID: 39892952 PMCID: PMC11788575 DOI: 10.1016/j.clp.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Circadian rhythms provide an anticipatory mechanism for organisms to adapt to environmental changes. Host response to infections is under robust circadian control. Most of the existing literature focuses on adults in epidemiologic and animal studies. Neonatal and early infancy represent critical windows in the consolidation of circadian rhythms. This review summarizes our understanding of the molecular clock, especially its relevance to immunity and adult sepsis. Further, using our knowledge of circadian biology in caring for a newborn host with emerging circadian rhythms represents a unique challenge and an opportunity for improving our approach and outcomes in neonatal sepsis.
Collapse
Affiliation(s)
- Shaon Sengupta
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, The Children's Hospital of Philadelphia, 3615 Curie Boulevard, Abramson Research Building, 1102C, Philadelphia, PA 19104, USA.
| | - David F Smith
- Division of Pediatric Otolaryngology-Head and Neck Surgery, Department of Surgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Bala S C Koritala
- Division of Pediatric Otolaryngology-Head and Neck Surgery, Department of Surgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| |
Collapse
|
21
|
Yao Y, Pauls S, Foley D, Yoshikawa T, Honma S, Honma KI, McVeigh E, Foley NC, Silver R. Suprachiasmatic nucleus-wide estimation of oscillatory temporal dynamics. PLoS Comput Biol 2025; 21:e1012855. [PMID: 40048409 PMCID: PMC11918361 DOI: 10.1371/journal.pcbi.1012855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 03/18/2025] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
The suprachiasmatic nucleus (SCN), locus of a circadian clock, is a small nucleus of approximately 20,000 neurons that oscillate with a period of about 24 hours. While individual neurons produce circadian oscillations even when dispersed in culture, the coherence and robustness of oscillation of the SCN as a whole is dependent on its circuitry. Surprisingly, the individual neurons of the intact SCN do not all oscillate in phase with each other. To understand the oscillatory dynamics across the intact nucleus, we develop a model of the relation of the phase of neurons to their PER2 expression at a particular subjective time (CT1900) using time series data from SCN slice preparations. Next, we use the model, which produces a surprisingly good fit in the SCN slice data, to estimate oscillator phase at a single time point (CT1900) in snapshot data from PER2 expression measurements in intact, unsliced SCN-wide tissue. To monitor temporal changes in phase in time series data, we use PER2::LUC imaging in an ex vivo SCN slice preparation. To study phase in the intact SCN at a fixed time point we use data generated by PER2 staining and a tissue clearing protocol. Because PER2 expression, as measured in the time series slices and the snapshot intact SCN are not directly comparable, the model estimated from time series slices to the snapshot intact SCN data requires a calibrating constant. The results indicate that our model provides a surprisingly good fit to the SCN slice data and is therefore a meaningful method for estimating phase in the intact SCN snapshot data, permitting the study of virtual interventions such as virtual tissue slicing. We next compare oscillation in circuits in the SCN-wide tissue to those that have been disrupted by virtual slicing using a Kuramoto model to simulate the dynamics. The results support prior evidence that the damage done by coronal slicing has the most disruptive impact on SCN oscillation, while horizontal slicing has the least damage. The results point to the importance of connectivity along the caudal-to-rostral axis and indicate that SCN circuit organization depends on the caudal-to-rostral flow of information. In summary, the construction of this model is a major finding of the paper. Our modeling allows us to perform the previously impossible analysis of oscillatory dynamics in static data in an intact SCN captured at a single time point.
Collapse
Affiliation(s)
- Yifan Yao
- Department of Psychology, Columbia University, New York, New York, United States of America
| | - Scott Pauls
- Department of Mathematics, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Duncan Foley
- Department of Economics, Emeritus, The New School for Social Research, New York, New York, United States of America
| | - Tomoko Yoshikawa
- Organization for International Education and Exchange University of Toyama, Toyama, Japan
| | - Sato Honma
- Research and Education Center for Brain Science, Hokkaido University, Sapporo, Japan
- Center for Sleep and Circadian Rhythm Disorders, Sapporo Hanazono Hospital, Sapporo, Japan
| | - Ken-Ichi Honma
- Research and Education Center for Brain Science, Hokkaido University, Sapporo, Japan
- Center for Sleep and Circadian Rhythm Disorders, Sapporo Hanazono Hospital, Sapporo, Japan
| | - Ellie McVeigh
- Department of Psychology, Columbia University, New York, New York, United States of America
| | - Nicolas C. Foley
- Department of Psychiatry, Columbia University Medical Center, New York, New York, United States of America
- New York State Psychiatric Institute, New York, New York, United States of America
| | - Rae Silver
- Department of Psychology, Columbia University, New York, New York, United States of America
- Department of Neuroscience and Behavior, Barnard College, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Medical School, New York, New York, United States of America
- Zuckerman Institute Affiliate, Columbia University, New York, New York, United States of America
| |
Collapse
|
22
|
Smyllie NJ, Hastings MH, Patton AP. Neuron-Astrocyte Interactions and Circadian Timekeeping in Mammals. Neuroscientist 2025; 31:65-79. [PMID: 38602223 PMCID: PMC7616557 DOI: 10.1177/10738584241245307] [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: 04/12/2024]
Abstract
Almost every facet of our behavior and physiology varies predictably over the course of day and night, anticipating and adapting us to their associated opportunities and challenges. These rhythms are driven by endogenous biological clocks that, when deprived of environmental cues, can continue to oscillate within a period of approximately 1 day, hence circa-dian. Normally, retinal signals synchronize them to the cycle of light and darkness, but disruption of circadian organization, a common feature of modern lifestyles, carries considerable costs to health. Circadian timekeeping pivots around a cell-autonomous molecular clock, widely expressed across tissues. These cellular timers are in turn synchronized by the principal circadian clock of the brain: the hypothalamic suprachiasmatic nucleus (SCN). Intercellular signals make the SCN network a very powerful pacemaker. Previously, neurons were considered the sole SCN timekeepers, with glial cells playing supportive roles. New discoveries have revealed, however, that astrocytes are active partners in SCN network timekeeping, with their cell-autonomous clock regulating extracellular glutamate and GABA concentrations to control circadian cycles of SCN neuronal activity. Here, we introduce circadian timekeeping at the cellular and SCN network levels before focusing on the contributions of astrocytes and their mutual interaction with neurons in circadian control in the brain.
Collapse
Affiliation(s)
- Nicola J. Smyllie
- Medical Research Council Laboratory of Molecular Biology, Cambridge, U.K
| | | | - Andrew P. Patton
- Medical Research Council Laboratory of Molecular Biology, Cambridge, U.K
| |
Collapse
|
23
|
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
|
24
|
Mihut A, O'Neill JS, Partch CL, Crosby P. PERspectives on circadian cell biology. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230483. [PMID: 39842483 PMCID: PMC11753889 DOI: 10.1098/rstb.2023.0483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/18/2024] [Accepted: 05/24/2024] [Indexed: 01/24/2025] Open
Abstract
Daily rhythms in the activities of PERIOD proteins are critical to the temporal regulation of mammalian physiology. While the molecular partners and genetic circuits that allow PERIOD to effect auto-repression and regulate transcriptional programmes are increasingly well understood, comprehension of the time-resolved mechanisms that allow PERIOD to conduct this daily dance is incomplete. Here, we consider the character and controversies of this central mammalian clock protein with a focus on its intrinsically disordered nature.This article is part of the Theo Murphy meeting issue 'Circadian rhythms in infection and immunity'.
Collapse
Affiliation(s)
- Andrei Mihut
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, CambridgeCB2 0QH, UK
| | - John S. O'Neill
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, CambridgeCB2 0QH, UK
| | - Carrie L. Partch
- Department of Chemistry and Biochemistry, University of California, 1156 High Street, Santa Cruz, CA95064, USA
| | - Priya Crosby
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, EdinburghEH9 3BF, UK
| |
Collapse
|
25
|
Zhao B, Nepovimova E, Wu Q. The role of circadian rhythm regulator PERs in oxidative stress, immunity, and cancer development. Cell Commun Signal 2025; 23:30. [PMID: 39825442 PMCID: PMC11740368 DOI: 10.1186/s12964-025-02040-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/11/2025] [Indexed: 01/20/2025] Open
Abstract
The complex interaction between circadian rhythms and physiological functions is essential for maintaining human health. At the heart of this interaction lies the PERIOD proteins (PERs), pivotal to the circadian clock, influencing the timing of physiological and behavioral processes and impacting oxidative stress, immune functionality, and tumorigenesis. PER1 orchestrates the cooperation of the enzyme GPX1, modulating mitochondrial dynamics in sync with daily rhythms and oxidative stress, thus regulating the mechanisms managing energy substrates. PERs in innate immune cells modulate the temporal patterns of NF-κB and TNF-α activities, as well as the response to LPS-induced toxic shock, initiating inflammatory responses that escalate into chronic inflammatory conditions. Crucially, PERs modulate cancer cell behaviors including proliferation, apoptosis, and migration by influencing the levels of cell cycle proteins and stimulating the expression of oncogenes c-Myc and MDM2. PER2/3, as antagonists in cancer stem cell biology, play important roles in differentiating cancer stem cells and in maintaining their stemness. Importantly, the expression of Pers serve as a significant factor for early cancer diagnosis and prognosis. This review delves into the link between circadian rhythm regulator PERs, disruptions in circadian rhythm, and oncogenesis. We examine the evidence that highlights how dysfunctions in PERs activities initiate cancer development, aid tumor growth, and modify cancer cell metabolism through pathways involved in oxidative stress and immune system. Comprehending these connections opens new pathways for the development of circadian rhythm-based therapeutic strategies, with the aims of boosting immune responses and enhancing cancer treatments.
Collapse
Affiliation(s)
- Baimei Zhao
- College of Life Science, Yangtze University, Jingzhou, 434025, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové , 500 03, Czech Republic
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, 434025, China.
| |
Collapse
|
26
|
Sharma SA, Oladejo SO, Kuang Z. Chemical interplay between gut microbiota and epigenetics: Implications in circadian biology. Cell Chem Biol 2025; 32:61-82. [PMID: 38776923 PMCID: PMC11569273 DOI: 10.1016/j.chembiol.2024.04.016] [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: 12/15/2023] [Revised: 03/22/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Circadian rhythms are intrinsic molecular mechanisms that synchronize biological functions with the day/night cycle. The mammalian gut is colonized by a myriad of microbes, collectively named the gut microbiota. The microbiota impacts host physiology via metabolites and structural components. A key mechanism is the modulation of host epigenetic pathways, especially histone modifications. An increasing number of studies indicate the role of the microbiota in regulating host circadian rhythms. However, the mechanisms remain largely unknown. Here, we summarize studies on microbial regulation of host circadian rhythms and epigenetic pathways, highlight recent findings on how the microbiota employs host epigenetic machinery to regulate circadian rhythms, and discuss its impacts on host physiology, particularly immune and metabolic functions. We further describe current challenges and resources that could facilitate research on microbiota-epigenetic-circadian rhythm interactions to advance our knowledge of circadian disorders and possible therapeutic avenues.
Collapse
Affiliation(s)
- Samskrathi Aravinda Sharma
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Sarah Olanrewaju Oladejo
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Zheng Kuang
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA.
| |
Collapse
|
27
|
Mao W, Ge X, Chen Q, Li JD. Epigenetic Mechanisms in the Transcriptional Regulation of Circadian Rhythm in Mammals. BIOLOGY 2025; 14:42. [PMID: 39857273 PMCID: PMC11762092 DOI: 10.3390/biology14010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025]
Abstract
Almost all organisms, from the simplest bacteria to advanced mammals, havea near 24 h circadian rhythm. Circadian rhythms are highly conserved across different life forms and are regulated by circadian genes as well as by related transcription factors. Transcription factors are fundamental to circadian rhythms, influencing gene expression, behavior in plants and animals, and human diseases. This review examines the foundational research on transcriptional regulation of circadian rhythms, emphasizing histone modifications, chromatin remodeling, and Pol II pausing control. These studies have enhanced our understanding of transcriptional regulation within biological circadian rhythms and the importance of circadian biology in human health. Finally, we summarize the progress and challenges in these three areas of regulation to move the field forward.
Collapse
Affiliation(s)
- Wei Mao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou 310000, China; (W.M.); (X.G.)
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China
| | - Xingnan Ge
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou 310000, China; (W.M.); (X.G.)
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China
| | - Qianping Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou 310000, China; (W.M.); (X.G.)
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China
| | - Jia-Da Li
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| |
Collapse
|
28
|
Procopio SB, Esser KA. Clockwork conditioning: Aligning the skeletal muscle clock with time-of-day exercise for cardiometabolic health. J Mol Cell Cardiol 2025; 198:36-44. [PMID: 39615287 PMCID: PMC11780665 DOI: 10.1016/j.yjmcc.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/04/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025]
Abstract
Circadian rhythms have evolved to synchronize gene expression, physiology, and behavior with time-of-day changes in the external environment. In every mammalian cell exists a core clock mechanism that consists of a transcriptional-translational feedback loop that drives rhythmic gene expression. Circadian disruption, as observed in shift workers and genetic mouse models, contributes to the onset and progression of cardiometabolic disorders. The central clock, located in the hypothalamus, is uniquely sensitive to external light cues, while the peripheral clocks are responsive to non-photic stimuli such as feeding and activity in addition to signals from the central clock. Recent research has illustrated the sensitivity of the skeletal muscle circadian clock to exercise timing, offering a promising avenue for therapeutic intervention in cardiometabolic health. Here we provide an in-depth examination of the molecular mechanisms underlying skeletal muscle clock function and its impact on cardiometabolic pathways, including glucose and lipid metabolism, as well as inflammation. To highlight the role of exercise as a time-cue for the skeletal muscle clock, we discuss evidence of exercise-induced shifts in the skeletal muscle clock and the differential response to exercise performed at different times of the day. Furthermore, we present data in support of time-of-day exercise as a potential therapeutic strategy for mitigating cardiometabolic disease burden. By exploring the relationship between the skeletal muscle clock, exercise timing, and cardiometabolic health, we identify new areas for future research and offer valuable insights into novel therapeutic approaches aimed at improving cardiometabolic disease outcomes.
Collapse
Affiliation(s)
- Spencer B Procopio
- Department of Physiology and Aging, University of Florida, Gainesville, FL, United States
| | - Karyn A Esser
- Department of Physiology and Aging, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
29
|
Wang Z, Wang S, Bi Y, Boiti A, Zhang S, Vallone D, Lan X, Foulkes NS, Zhao H. Light-regulated microRNAs shape dynamic gene expression in the zebrafish circadian clock. PLoS Genet 2025; 21:e1011545. [PMID: 39777894 PMCID: PMC11750094 DOI: 10.1371/journal.pgen.1011545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/21/2025] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
A key property of the circadian clock is that it is reset by light to remain synchronized with the day-night cycle. An attractive model to explore light input to the circadian clock in vertebrates is the zebrafish. Circadian clocks in zebrafish peripheral tissues and even zebrafish-derived cell lines are entrainable by direct light exposure thus providing unique insight into the function and evolution of light regulatory pathways. Our previous work has revealed that light-induced gene transcription is a key step in the entrainment of the circadian clock as well as enabling the more general adaptation of zebrafish cells to sunlight exposure. However, considerable evidence points to post-transcriptional regulatory mechanisms, notably microRNAs (miRNAs), playing an essential role in shaping dynamic changes in mRNA levels. Therefore, does light directly impact the function of miRNAs? Are there light-regulated miRNAs and if so, which classes of mRNA do they target? To address these questions, we performed a complete sequencing analysis of light-induced changes in the zebrafish transcriptome, encompassing small non-coding RNAs as well as mRNAs. Importantly, we identified sets of light-regulated miRNAs, with many regulatory targets representing light-inducible mRNAs including circadian clock genes and genes involved in redox homeostasis. We subsequently focused on the light-responsive miR-204-3-3p and miR-430a-3p which are predicted to regulate the expression of cryptochrome genes (cry1a and cry1b). Luciferase reporter assays validated the target binding of miR-204-3-3p and miR-430a-3p to the 3'UTRs of cry1a and cry1b, respectively. Furthermore, treatment with mimics and inhibitors of these two miRNAs significantly affected the dynamic expression of their target genes but also other core clock components (clock1a, bmal1b, per1b, per2, per3), as well as the rhythmic locomotor activity of zebrafish larvae. Thus, our identification of light-responsive miRNAs reveals new intricacy in the multi-level regulation of the circadian clockwork by light.
Collapse
Affiliation(s)
- Zuo Wang
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, China
| | - Shuang Wang
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, China
| | - Yi Bi
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, China
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Alessandra Boiti
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany
| | - Shengxiang Zhang
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, China
| | - Daniela Vallone
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Nicholas S. Foulkes
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany
| | - Haiyu Zhao
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, China
| |
Collapse
|
30
|
Burckard O, Teboul M, Delaunay F, Chaves M. Benchmark for quantitative characterization of circadian clock cycles. Biosystems 2025; 247:105363. [PMID: 39551427 DOI: 10.1016/j.biosystems.2024.105363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/03/2024] [Indexed: 11/19/2024]
Abstract
Understanding circadian clock mechanisms is fundamental in order to counteract the harmful effects of clock malfunctioning and associated diseases. Biochemical, genetic and systems biology approaches have provided invaluable information on the mechanisms of the circadian clock, from which many mathematical models have been developed to understand the dynamics and quantitative properties of the circadian oscillator. To better analyze and compare quantitatively all these circadian cycles, we propose a method based on a previously proposed circadian cycle segmentation into stages. We notably identify a sequence of eight stages that characterize the progress of the circadian cycle. Next, we apply our approach to an experimental dataset and to five different models, all built with ordinary differential equations. Our method permits to assess the agreement of mathematical model cycles with biological properties or to detect some inconsistencies. As another application of our method, we provide insights on how this segmentation into stages can help to analyze the effect of a clock gene loss of function on the dynamic of a genetic oscillator. The strength of our method is to provide a benchmark for characterization, comparison and improvement of new mathematical models of circadian oscillators in a wide variety of model systems.
Collapse
Affiliation(s)
- Odile Burckard
- Centre Inria d'Université Côte d'Azur, INRAE, CNRS, Macbes team, Sophia Antipolis, France.
| | | | | | - Madalena Chaves
- Centre Inria d'Université Côte d'Azur, INRAE, CNRS, Macbes team, Sophia Antipolis, France
| |
Collapse
|
31
|
Crouchet E, Dachraoui M, Jühling F, Roehlen N, Oudot MA, Durand SC, Ponsolles C, Gadenne C, Meiss-Heydmann L, Moehlin J, Martin R, Brignon N, Del Zompo F, Teraoka Y, Aikata H, Abe-Chayama H, Chayama K, Saviano A, Heide D, Onea M, Geyer L, Wolf T, Felli E, Pessaux P, Heikenwälder M, Chambon P, Schuster C, Lupberger J, Mukherji A, Baumert TF. Targeting the liver clock improves fibrosis by restoring TGF-β signaling. J Hepatol 2025; 82:120-133. [PMID: 39173955 DOI: 10.1016/j.jhep.2024.07.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND & AIMS Liver fibrosis is the major driver of hepatocellular carcinoma and liver disease-related death. Approved antifibrotic therapies are absent and compounds in development have limited efficacy. Increased TGF-β signaling drives collagen deposition by hepatic stellate cells (HSCs)/myofibroblasts. Here, we aimed to dissect the role of the circadian clock (CC) in controlling TGF-β signaling and liver fibrosis. METHODS Using CC-mutant mice, enriched HSCs and myofibroblasts obtained from healthy and fibrotic mice in different CC phases and loss-of-function studies in human hepatocytes and myofibroblasts, we investigated the relationship between CC and TGF-β signaling. We explored hepatocyte-myofibroblast communication through bioinformatic analyses of single-nuclei transcriptomes and performed validation in cell-based models. Using mouse models for MASH (metabolic dysfunction-associated steatohepatitis)-related fibrosis and spheroids from patients with liver disease, we performed proof-of-concept studies to validate pharmacological targetability and clinical translatability. RESULTS We discovered that the CC oscillator temporally gates TGF-β signaling and this regulation is broken in fibrosis. We demonstrate that HSCs and myofibroblasts contain a functional CC with rhythmic expression of numerous genes, including fibrogenic genes. Perturbation studies in hepatocytes and myofibroblasts revealed a reciprocal relationship between TGF-β activation and CC perturbation, which was confirmed in patient-derived ex vivo and in vivo models. Pharmacological modulation of CC-TGF-β signaling inhibited fibrosis in mouse models in vivo as well as in patient-derived liver spheroids. CONCLUSION The CC regulates TGF-β signaling, and the breakdown of this control is associated with liver fibrosis in patients. Pharmacological proof-of-concept studies across different models have uncovered the CC as a novel therapeutic target for liver fibrosis - a growing unmet medical need. IMPACT AND IMPLICATIONS Liver fibrosis due to metabolic diseases is a global health challenge. Many liver functions are rhythmic throughout the day, being controlled by the circadian clock (CC). Here we demonstrate that regulation of the CC is perturbed upon chronic liver injury and this perturbation contributes to fibrotic disease. By showing that a compound targeting the CC improves liver fibrosis in patient-derived models, this study provides a novel therapeutic candidate strategy to treat fibrosis in patients. Additional studies will be needed for clinical translation. Since the findings uncover a previously undiscovered profibrotic mechanism and therapeutic target, the study is of interest for scientists investigating liver disease, clinical hepatologists and drug developers.
Collapse
Affiliation(s)
- Emilie Crouchet
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
| | - Mayssa Dachraoui
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
| | - Frank Jühling
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
| | - Natascha Roehlen
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France; Department of Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Diseases, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marine A Oudot
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
| | - Sarah C Durand
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
| | - Clara Ponsolles
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
| | - Cloé Gadenne
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
| | - Laura Meiss-Heydmann
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
| | - Julien Moehlin
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
| | - Romain Martin
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France; ÆPIC Animal Facility Platform, University of Strasbourg, Inserm UMR_S1110, Strasbourg, France
| | - Nicolas Brignon
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France; ÆPIC Animal Facility Platform, University of Strasbourg, Inserm UMR_S1110, Strasbourg, France
| | - Fabio Del Zompo
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
| | - Yuji Teraoka
- Department of Gastroenterology, NHO Kure Medical Center and Chugoku Cancer Center, Kure, Japan
| | | | - Hiromi Abe-Chayama
- Hiroshima Institute of Life Sciences, Hiroshima, Japan; Center for Medical Specialist Graduate Education and Research, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuaki Chayama
- Hiroshima Institute of Life Sciences, Hiroshima, Japan; RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Antonio Saviano
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France; Pôle des Pathologies Hépatiques et Digestives, Strasbourg University Hospitals, Strasbourg, France
| | - Danijela Heide
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mihaela Onea
- Biological Resource Center, Hautepierre, Department of Pathology, Strasbourg University Hospital, Strasbourg, France
| | - Lucas Geyer
- Biological Resource Center, Hautepierre, Department of Pathology, Strasbourg University Hospital, Strasbourg, France
| | - Thibaut Wolf
- Biological Resource Center, Hautepierre, Department of Pathology, Strasbourg University Hospital, Strasbourg, France
| | - Emanuele Felli
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France; Groupe Hospitalier Saint Vincent, Strasbourg, France
| | - Patrick Pessaux
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France; Pôle des Pathologies Hépatiques et Digestives, Strasbourg University Hospitals, Strasbourg, France
| | - Mathias Heikenwälder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; The M3 Research Center, University Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Otfried-Müller-Straße 37, 72076 Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - Pierre Chambon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, Inserm U964, Illkirch, France; Collège de France, Illkirch, France
| | - Catherine Schuster
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
| | - Joachim Lupberger
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France.
| | - Atish Mukherji
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, Inserm U964, Illkirch, France
| | - Thomas F Baumert
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France; Pôle des Pathologies Hépatiques et Digestives, Strasbourg University Hospitals, Strasbourg, France; Institut Hospitalo-Universitaire (IHU) Strasbourg, France; Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
32
|
Zheng Q, Weekley BH, Vinson DA, Zhao S, Bastle RM, Thompson RE, Stransky S, Ramakrishnan A, Cunningham AM, Dutta S, Chan JC, Di Salvo G, Chen M, Zhang N, Wu J, Fulton SL, Kong L, Wang H, Zhang B, Vostal L, Upad A, Dierdorff L, Shen L, Molina H, Sidoli S, Muir TW, Li H, David Y, Maze I. Bidirectional histone monoaminylation dynamics regulate neural rhythmicity. Nature 2025; 637:974-982. [PMID: 39779849 PMCID: PMC11754111 DOI: 10.1038/s41586-024-08371-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 11/08/2024] [Indexed: 01/11/2025]
Abstract
Histone H3 monoaminylations at Gln5 represent an important family of epigenetic marks in brain that have critical roles in permissive gene expression1-3. We previously demonstrated that serotonylation4-10 and dopaminylation9,11-13 of Gln5 of histone H3 (H3Q5ser and H3Q5dop, respectively) are catalysed by transglutaminase 2 (TG2), and alter both local and global chromatin states. Here we found that TG2 additionally functions as an eraser and exchanger of H3 monoaminylations, including H3Q5 histaminylation (H3Q5his), which displays diurnally rhythmic expression in brain and contributes to circadian gene expression and behaviour. We found that H3Q5his, in contrast to H3Q5ser, inhibits the binding of WDR5, a core member of histone H3 Lys4 (H3K4) methyltransferase complexes, thereby antagonizing methyltransferase activities on H3K4. Taken together, these data elucidate a mechanism through which a single chromatin regulatory enzyme has the ability to sense chemical microenvironments to affect the epigenetic states of cells, the dynamics of which have critical roles in the regulation of neural rhythmicity.
Collapse
Affiliation(s)
- Qingfei Zheng
- Department of Radiation Oncology, College of Medicine and Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, USA
| | - Benjamin H Weekley
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David A Vinson
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shuai Zhao
- State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Protein Sciences, Beijing Frontier Research Center for Biological Structure, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Ryan M Bastle
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Stephanie Stransky
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ashley M Cunningham
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sohini Dutta
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jennifer C Chan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giuseppina Di Salvo
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Min Chen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nan Zhang
- Department of Radiation Oncology, College of Medicine and Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Jinghua Wu
- Department of Radiation Oncology, College of Medicine and Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, USA
| | - Sasha L Fulton
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lingchun Kong
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Haifeng Wang
- State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Protein Sciences, Beijing Frontier Research Center for Biological Structure, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Baichao Zhang
- State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Protein Sciences, Beijing Frontier Research Center for Biological Structure, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Lauren Vostal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA
| | - Akhil Upad
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA
| | - Lauren Dierdorff
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Henrik Molina
- The Rockefeller University Proteomics Resource Center, The Rockefeller University, New York, NY, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY, USA
| | - Tom W Muir
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Haitao Li
- State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Protein Sciences, Beijing Frontier Research Center for Biological Structure, School of Basic Medical Sciences, Tsinghua University, Beijing, China.
- SXMU-TM Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| | - Yael David
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA.
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA.
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.
| | - Ian Maze
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Howard Hughes Medical Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
33
|
Burckard O, Chaves M. Analytic solutions for the circadian oscillator characterize cycle dynamics and its robustness. J Math Biol 2024; 90:5. [PMID: 39673639 DOI: 10.1007/s00285-024-02164-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 09/25/2024] [Accepted: 11/10/2024] [Indexed: 12/16/2024]
Abstract
Circadian clocks form a fundamental mechanism that promotes the correct behavior of many cellular and molecular processes by synchronizing them on a 24 h period. However, the circadian cycles remain difficult to describe mathematically. To overcome this problem, we first propose a segmentation of the circadian cycle into eight stages based on the levels of expression of the core clock components CLOCK:BMAL1, REV-ERB and PER:CRY. This cycle segmentation is next characterized through a piecewise affine model, whose analytical study allows us to propose an Algorithm to generate biologically-consistent circadian oscillators. Our study provides a characterization of the cycle dynamics in terms of four fundamental threshold parameters and one scaling parameter, shows robustness of the circadian system and its period, and identifies critical points for correct cycle progression.
Collapse
Affiliation(s)
- Odile Burckard
- Macbes team, INRAE, CNRS, Centre Inria d'Université Côte d'Azur, Sophia Antipolis, France.
| | - Madalena Chaves
- Macbes team, INRAE, CNRS, Centre Inria d'Université Côte d'Azur, Sophia Antipolis, France
| |
Collapse
|
34
|
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
|
35
|
Morena da Silva F, Esser KA, Murach KA, Greene NP. Inflammation o'clock: interactions of circadian rhythms with inflammation-induced skeletal muscle atrophy. J Physiol 2024; 602:6587-6607. [PMID: 37563881 PMCID: PMC10858298 DOI: 10.1113/jp284808] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
Circadian rhythms are ∼24 h cycles evident in behaviour, physiology and metabolism. The molecular mechanism directing circadian rhythms is the circadian clock, which is composed of an interactive network of transcription-translation feedback loops. The core clock genes include Bmal1, Clock, Rev-erbα/β, Per and Cry. In addition to keeping time, the core clock regulates a daily programme of gene expression that is important for overall cell homeostasis. The circadian clock mechanism is present in all cells, including skeletal muscle fibres, and disruption of the muscle clock is associated with changes in muscle phenotype and function. Skeletal muscle atrophy is largely associated with a lower quality of life, frailty and reduced lifespan. Physiological and genetic modification of the core clock mechanism yields immune dysfunction, alters inflammatory factor expression and secretion and is associated with skeletal muscle atrophy in multiple conditions, such as ageing and cancer cachexia. Here, we summarize the possible interplay between the circadian clock modulation of immune cells, systemic inflammatory status and skeletal muscle atrophy in chronic inflammatory conditions. Although there is a clear disruption of circadian clocks in various models of atrophy, the mechanism behind such alterations remains unknown. Understanding the modulatory potential of muscle and immune circadian clocks in inflammation and skeletal muscle health is essential for the development of therapeutic strategies to protect skeletal muscle mass and function of patients with chronic inflammation.
Collapse
Affiliation(s)
- Francielly Morena da Silva
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Karyn A Esser
- Department of Physiology and Ageing, College of Medicine, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, FL, USA
| | - Kevin A Murach
- Molecular Muscle Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Nicholas P Greene
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| |
Collapse
|
36
|
Ye Y, Liu C, Wu R, Kang D, Gao H, Lv H, Feng Z, Shi Y, Liu Z, Chen L. Circadian clock component PER2 negatively regulates CD4 + T cell IFN-γ production in ulcerative colitis. Mucosal Immunol 2024; 17:1161-1173. [PMID: 39097147 DOI: 10.1016/j.mucimm.2024.07.010] [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: 12/14/2023] [Revised: 06/24/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Period circadian clock 2 (PER2) is involved in the pathogenesis of various inflammatory and autoimmune diseases. However, there are gaps in our understanding of the role of PER2 in regulating CD4+ T cells beyond its time-keeping function in ulcerative colitis (UC) pathogenesis. Our findings revealed PER2 was predominantly expressed in CD4+ T cells, while it was significantly decreased in the inflamed mucosa and peripheral blood CD4+ T cells of UC patients compared with that in Crohn's disease (CD) patients and healthy controls (HC). Notably, PER2 expression was significantly recovered in UC patients in remission (R-UC) compared to that in active UC patients (A-UC) but not in CD patients. It was negatively correlated with the Ulcerative Colitis Endoscopic Index of Severity (UCEIS), Crohn's Disease Activity Index (CDAI), Simple Endoscopic Score for Crohn's disease (SES-CD), and C-reactive protein (CRP), respectively. Overexpression of PER2 markedly inhibited IFN-γ production in UC CD4+ T cells. RNA-seq analysis showed that overexpression of PER2 could repress the expression of a disintegrin and metalloproteinase 12 (ADAM12), a costimulatory molecule that determines Th1 cell fate. Mechanistically, cleavage under targets and tagmentation (CUT&Tag) analysis revealed that PER2 down-regulated ADAM12 expression by reducing its binding activity, thereby suppressing IFN-γ production in UC CD4+ T cells. Additionally, our data further demonstrated that ADAM12 was upregulated in CD4+ T cells and inflamed mucosa of A-UC patients compared to HC. Our study reveals a critical role of PER2 in regulating CD4+ T cell differentiation and highlights its potential as a therapeutic target for UC treatment.
Collapse
Affiliation(s)
- Yulan Ye
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Gastroenterology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou 215008, China
| | - Changqin Liu
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Ruijin Wu
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Dengfeng Kang
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Han Gao
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Huiying Lv
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Zhongsheng Feng
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Yanhong Shi
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Zhanju Liu
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China.
| | - Liang Chen
- Center for IBD Research and Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China.
| |
Collapse
|
37
|
de Assis LVM, Kramer A. Circadian de(regulation) in physiology: implications for disease and treatment. Genes Dev 2024; 38:933-951. [PMID: 39419580 PMCID: PMC11610937 DOI: 10.1101/gad.352180.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Time plays a crucial role in the regulation of physiological processes. Without a temporal control system, animals would be unprepared for cyclic environmental changes, negatively impacting their survival. Experimental studies have demonstrated the essential role of the circadian system in the temporal coordination of physiological processes. Translating these findings to humans has been challenging. Increasing evidence suggests that modern lifestyle factors such as diet, sedentarism, light exposure, and social jet lag can stress the human circadian system, contributing to misalignment; i.e., loss of phase coherence across tissues. An increasing body of evidence supports the negative impact of circadian disruption on several human health parameters. This review aims to provide a comprehensive overview of how circadian disruption influences various physiological processes, its long-term health consequences, and its association with various diseases. To illustrate the relevant consequences of circadian disruption, we focused on describing the many physiological consequences faced by shift workers, a population known to experience high levels of circadian disruption. We also discuss the emerging field of circadian medicine, its founding principles, and its potential impact on human health.
Collapse
Affiliation(s)
| | - Achim Kramer
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, Laboratory of Chronobiology, Berlin Institute of Health, 10117 Berlin, Germany
| |
Collapse
|
38
|
Torres M, Kirchner M, Marks CG, Mertins P, Kramer A. Proteomic insights into circadian transcription regulation: novel E-box interactors revealed by proximity labeling. Genes Dev 2024; 38:1020-1032. [PMID: 39562139 PMCID: PMC11610934 DOI: 10.1101/gad.351836.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024]
Abstract
Circadian clocks (∼24 h) are responsible for daily physiological, metabolic, and behavioral changes. Central to these oscillations is the regulation of gene transcription. Previous research has identified clock protein complexes that interact with the transcriptional machinery to orchestrate circadian transcription, but technological constraints have limited the identification of de novo proteins. Here we use a novel genomic locus-specific quantitative proteomics approach to provide a new perspective on time of day-dependent protein binding at a critical chromatin locus involved in circadian transcription: the E-box. Using proximity labeling proteomics at the E-box of the clock-controlled Dbp gene in mouse fibroblasts, we identified 69 proteins at this locus at the time of BMAL1 binding. This method successfully enriched BMAL1 as well as HDAC3 and HISTONE H2A.V/Z, known circadian regulators. New E-box proteins include the MINK1 kinase and the transporters XPO7 and APPL1, whose depletion in human U-2 OS cells results in disrupted circadian rhythms, suggesting a role in the circadian transcriptional machinery. Overall, our approach uncovers novel circadian modulators and provides a new strategy to obtain a complete temporal picture of circadian transcriptional regulation.
Collapse
Affiliation(s)
- Manon Torres
- Laboratory of Chronobiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Marieluise Kirchner
- Core Unit Proteomics, Berlin Institute of Health at Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Caroline G Marks
- Laboratory of Chronobiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Philipp Mertins
- Core Unit Proteomics, Berlin Institute of Health at Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Achim Kramer
- Laboratory of Chronobiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany;
| |
Collapse
|
39
|
Duong TVQ, Yaw AM, Zhou G, Sina N, Cherukuri AS, Nguyen D, Cataldo K, Ly N, Sen A, Sempere L, Detrie C, Seiler R, Olomu IN, Cortese R, Long R, Hoffmann HM. Interaction between time-of-day and oxytocin efficacy in mice and humans with and without gestational diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.16.622460. [PMID: 39605626 PMCID: PMC11601330 DOI: 10.1101/2024.11.16.622460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Management of labor in women with diabetes is challenging due to the high risk of peri- and postpartum complications. To avoid cesarean section and assist with labor progression, Pitocin, a synthetic oxytocin, is frequently used to induce and augment labor. However, the efficacy of Pitocin is often compromised in diabetic pregnancies, leading to increased cesarian delivery. As diabetes deregulates the body's circadian timekeeping system and the time-of-day of the first Pitocin administration contributes to labor duration, our objective was to determine how the time of day and the circadian clock gene, Bmal1, gates oxytocin efficacy. Our studies in mice show that, compared to the rest phase of the day (lights on), the uterotonic efficacy of oxytocin is significantly increased during the active phase (lights off). Using in vitro studies, a myometrium-specific Bmal1 conditional knockout mouse model, and a mouse model of food-induced gestational diabetes, we find that Bmal1 is crucial for maintaining oxytocin receptor expression and response in the myometrium in mice. These findings also translate to humans, where oxytocin-induced human myometrial cell contraction is time-of-day dependent, and retrospective clinical data suggest that administration of Pitocin in the morning should be considered for pregnant women with gestational diabetes.
Collapse
Affiliation(s)
- Thu Van-Quynh Duong
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
- Department of Obstetrics and Gynecology, McLaren Greater Lansing Hospital, Lansing, MI, USA, 48910
| | - Alexandra M. Yaw
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
| | - Guoli Zhou
- Center for Statistical Training & Consulting (CSTAT), Michigan State University, East Lansing, MI, USA
| | - Niharika Sina
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
| | - Aneesh Sai Cherukuri
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
| | - Duong Nguyen
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
| | - Kylie Cataldo
- Department of Pediatrics; Department of Obstetrics, Gynecology, and Women’s Health. University of Missouri. Columbia, MO, 65212, USA
| | - Nicollette Ly
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
| | - Aritro Sen
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
| | - Lorenzo Sempere
- Precision Health Program; Department of Radiology, Michigan State University, East Lansing, MI, USA
| | - Cara Detrie
- Department of Obstetrics and Gynecology, McLaren Greater Lansing Hospital, Lansing, MI, USA, 48910
| | - Robert Seiler
- Department of Obstetrics and Gynecology, McLaren Greater Lansing Hospital, Lansing, MI, USA, 48910
| | - I. Nicholas Olomu
- Department of Pediatrics & Human Development, Division of Neonatal-Perinatal Medicine, Michigan State University, East Lansing MI, 48824
| | - Rene Cortese
- Department of Pediatrics; Department of Obstetrics, Gynecology, and Women’s Health. University of Missouri. Columbia, MO, 65212, USA
| | - Robert Long
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hanne M. Hoffmann
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA, 48824
| |
Collapse
|
40
|
Yamazaki S, Valekunja UK, Chen-Roetling J, Reddy AB. Heat Shock Factor 1 Governs Sleep-Wake Cycles Across Species. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623879. [PMID: 39605613 PMCID: PMC11601485 DOI: 10.1101/2024.11.15.623879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Heat Shock Factor 1 (HSF1) is a critical transcription factor for cellular proteostasis, but its role in sleep regulation remains unexplored. We demonstrate that nuclear HSF1 levels in the mouse brain fluctuate with sleep-wake cycles, increasing during extended wakefulness and decreasing during sleep. Using CUT&RUN and RNA-seq, we identified HSF1-regulated transcriptional changes involved in synaptic organization, expanding its known functions beyond traditional heat shock responses. Both systemic and brain-specific Hsf1 knockout mice exhibit altered sleep homeostasis, including increased delta power after sleep deprivation and upregulation of sleep-related genes. However, these knockouts struggle to maintain sleep due to disrupted synaptic organization. In Drosophila , knockout of HSF1's ortholog results in fragmented sleep patterns, suggesting a conserved role for HSF1 in sleep regulation across species. Our findings reveal a novel molecular mechanism underlying sleep regulation and offer potential therapeutic targets for sleep disturbances.
Collapse
|
41
|
Ding H, Meng L, Zhang Y, Bryant AJ, Xing C, Esser KA, Chen L, Huo Z. A Bayesian Framework for Genome-wide Circadian Rhythmicity Biomarker Detection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620703. [PMID: 39554018 PMCID: PMC11565714 DOI: 10.1101/2024.10.28.620703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Circadian rhythms are endogenous ∼24-hour cycles that significantly influence physiological and behavioral processes. These rhythms are governed by a transcriptional-translational feedback loop of core circadian genes and are essential for maintaining overall health. The study of circadian rhythms has expanded into various omics datasets, necessitating accurate analytical methodology for circadian biomarker detection. Here, we introduce a novel Bayesian framework for the genome-wide detection of circadian rhythms that is capable of incorporating prior biological knowledge and adjusting for multiple testing issue via a false discovery rate approach. Our framework leverages a Bayesian hierarchical model and employs a reverse jump Markov chain Monte Carlo (rjMCMC) technique for model selection. Through extensive simulations, our method, BayesCircRhy, demonstrated superior false discovery rate control over competing methods, robustness against heavier-tailed error distributions, and better performance compared to existing approaches. The method's efficacy was further validated in two RNA-Sequencing data, including a human resitrcted feeding data and a mouse aging data, where it successfully identified known and novel circadian genes. R package "BayesianCircadian" for the method is publicly available on GitHub https://github.com/jxncdhc/BayesianCircadian .
Collapse
|
42
|
Singh A, Anjum B, Naz Q, Raza S, Sinha RA, Ahmad MK, Mehdi AA, Verma N. Night shift-induced circadian disruption: links to initiation of non-alcoholic fatty liver disease/non-alcoholic steatohepatitis and risk of hepatic cancer. HEPATOMA RESEARCH 2024:2394-5079.2024.88. [PMID: 39525867 PMCID: PMC7616786 DOI: 10.20517/2394-5079.2024.88] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The circadian system plays a crucial role in regulating metabolic homeostasis at both systemic and tissue levels by synchronizing the central and peripheral clocks with exogenous time cues, known as zeitgebers (such as the light/dark cycle). Our body's behavioral rhythms, including sleep-wake cycles and feeding-fasting patterns, align with these extrinsic time cues. The body cannot effectively rest and repair itself when circadian rhythms are frequently disrupted. In many shift workers, the internal rhythms fail to fully synchronize with the end and start times of their shifts. Additionally, exposure to artificial light at night (LAN), irregular eating patterns, and sleep deprivation contribute to circadian disruption and misalignment. Shift work and jet lag disrupt the normal circadian rhythm of liver activity, resulting in a condition known as "circadian disruption". This disturbance adversely affects the metabolism and homeostasis of the liver, contributing to excessive fat accumulation and abnormal liver function. Additionally, extended working hours, such as prolonged night shifts, may worsen the progression of non-alcoholic fatty liver disease (NAFLD) toward non-alcoholic steatohepatitis (NASH) and increase disease severity. Studies have demonstrated a positive correlation between night shift work (NSW) and elevated liver enzymes, indicative of hepatic metabolic dysfunction, potentially increasing the risk of hepatocellular carcinoma (HCC) related to NAFLD. This review consolidates research findings on circadian disruption caused by NSW, late chronotype, jet lag, and social jet lag, drawing insights from studies involving both humans and animal models that investigate the effects of these factors on circadian rhythms in liver metabolism.
Collapse
Affiliation(s)
- Anjali Singh
- Department of Physiology, King George’s Medical University, Lucknow226003, India
| | - Baby Anjum
- Department of Neurology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow226014, India
| | - Qulsoom Naz
- Department of Medicine, King George’s Medical University, Lucknow226003, India
| | - Sana Raza
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow226014, India
| | - Rohit A. Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow226014, India
| | | | | | - Narsingh Verma
- Hind Institute of Medical Sciences, Sitapur 261304, India
| |
Collapse
|
43
|
Xu K, Zhang Y, Shi Y, Zhang Y, Zhang C, Wang T, Lv P, Bai Y, Wang S. Circadian rhythm disruption: a potential trigger in Parkinson's disease pathogenesis. Front Cell Neurosci 2024; 18:1464595. [PMID: 39539340 PMCID: PMC11557417 DOI: 10.3389/fncel.2024.1464595] [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: 07/14/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by the gradual loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc), abnormal accumulation of α-synuclein (α-syn), and activation of microglia leading to neuroinflammation. Disturbances in circadian rhythm play a significant role in PD, with most non-motor symptoms associated with disruptions in circadian rhythm. These disturbances can be observed years before motor symptoms appear and are marked by the emergence of non-motor symptoms related to PD, such as rapid eye movement sleep behavior disorder (RBD), restless leg syndrome (RLS), excessive daytime sleepiness (EDS), depression and anxiety, changes in blood pressure, gastrointestinal dysfunction, and urinary problems. Circadian rhythm disruption precedes the onset of motor symptoms and contributes to the progression of PD. In brief, this article outlines the role of circadian rhythm disruption in triggering PD at cellular and molecular levels, as well as its clinical manifestations. It also explores how circadian rhythm research can contribute to preventing the onset and progression of PD from current and future perspectives.
Collapse
Affiliation(s)
- Ke Xu
- The Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yu Zhang
- The Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yue Shi
- The Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yake Zhang
- The Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Chengguang Zhang
- The Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Tianjiao Wang
- The Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Peizhu Lv
- The Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yan Bai
- Institute of Acupuncture and Moxibustion, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Shun Wang
- The Second Clinical Medical College, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
44
|
Zee A, Deng DZ, DiTacchio L, Vollmers C. The Circadian Isoform Landscape of Mouse Livers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618716. [PMID: 39464136 PMCID: PMC11507890 DOI: 10.1101/2024.10.16.618716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The mammalian circadian clock is an autoregulatory feedback process that is responsible for homeostasis in mouse livers. These circadian processes are well understood at the gene-level, however, not well understood at the isoform-level. To investigate circadian oscillations at the isoform-level, we used the nanopore-based R2C2 method to create over 78 million highly-accurate, full-length cDNA reads for 12 RNA samples extracted from mouse livers collected at 2 hour intervals. To generate a circadian mouse liver isoform-level transcriptome, we processed these reads using the Mandalorion tool which identified and quantified 58,612 isoforms, 1806 of which showed circadian oscillations. We performed detailed analysis on the circadian oscillation of these isoforms, their coding sequences, and transcription start sites and compiled easy-to-access resources for other researchers. This study and its results add a new layer of detail to the quantitative analysis of transcriptomes.
Collapse
Affiliation(s)
- Alexander Zee
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California 95064, USA
| | - Dori Z.Q. Deng
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Cruz, Santa Cruz, California 95064, USA
| | - Luciano DiTacchio
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66101, USA
- Current affiliation: Synexin LLC, Carlsbad, CA 92010
| | - Christopher Vollmers
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California 95064, USA
| |
Collapse
|
45
|
Uriu K, Hernandez-Sanchez JP, Kojima S. Impacts of the feedback loop between sense-antisense RNAs in regulating circadian rhythms. NPJ Syst Biol Appl 2024; 10:119. [PMID: 39414861 PMCID: PMC11484753 DOI: 10.1038/s41540-024-00451-4] [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: 06/04/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
Antisense transcripts are a unique group of non-coding RNAs and play regulatory roles in a variety of biological processes, including circadian rhythms. Per2AS is an antisense transcript to the sense core clock gene Period2 (Per2) in mouse and its expression is rhythmic and antiphasic to Per2. To understand the impact of Per2AS-Per2 interaction, we developed a new mathematical model that mechanistically described the mutually repressive relationship between Per2 and Per2AS. This mutual repression can regulate both amplitude and period of circadian oscillation by affecting a negative feedback regulation of Per2. Simulations from this model also fit with experimental observations that could not be fully explained by our previous model. Our revised model can not only serve as a foundation to build more detailed models to better understand the impact of Per2AS-Per2 interaction in the future, but also be used to analyze other sense-antisense RNA pairs that mutually repress each other.
Collapse
Affiliation(s)
- Koichiro Uriu
- School of Life Science and Technology, Institute of Science Tokyo, Meguro, Tokyo, Japan.
- Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa, Ishikawa, Japan.
| | - Juan P Hernandez-Sanchez
- Department of Biological Sciences, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, USA
| | - Shihoko Kojima
- Department of Biological Sciences, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, USA.
- Center for the Mathematics of Biosystems, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
46
|
McManus D, Patton AP, Smyllie NJ, Chin JW, Hastings MH. PERfect Day: reversible and dose-dependent control of circadian time-keeping in the mouse suprachiasmatic nucleus by translational switching of PERIOD2 protein expression. Eur J Neurosci 2024; 60:5537-5552. [PMID: 39300693 PMCID: PMC7617102 DOI: 10.1111/ejn.16537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024]
Abstract
The biological clock of the suprachiasmatic nucleus (SCN) orchestrates circadian (approximately daily) rhythms of behaviour and physiology that underpin health. SCN cell-autonomous time-keeping revolves around a transcriptional/translational feedback loop (TTFL) within which PERIOD (PER1,2) and CRYPTOCHROME (CRY1,2) proteins heterodimerise and suppress trans-activation of their encoding genes (Per1,2; Cry1,2). To explore its contribution to SCN time-keeping, we used adeno-associated virus-mediated translational switching to express PER2 (tsPER2) in organotypic SCN slices carrying bioluminescent TTFL circadian reporters. Translational switching requires provision of the non-canonical amino acid, alkyne lysine (AlkK), for protein expression. Correspondingly, AlkK, but not vehicle, induced constitutive expression of tsPER2 in SCN neurons and reversibly and dose-dependently suppressed pPer1-driven transcription in PER-deficient (Per1,2-null) SCN, illustrating the potency of PER2 in negative regulation within the TTFL. Constitutive expression of tsPER2, however, failed to initiate circadian oscillations in arrhythmic PER-deficient SCN. In rhythmic, PER-competent SCN, AlkK dose-dependently reduced the amplitude of PER2-reported oscillations as inhibition by tsPER2 progressively damped the TTFL. tsPER2 also dose-dependently lengthened the period of the SCN TTFL and neuronal calcium rhythms. Following wash-out of AlkK to remove tsPER2, the SCN regained TTFL amplitude and period. Furthermore, SCN retained their pre-washout phase: the removal of tsPER2 did not phase-shift the TTFL. Given that constitutive tsCRY1 can regulate TTFL amplitude and period, but also reset TTFL phase and initiate rhythms in CRY-deficient SCN, these results reveal overlapping and distinct properties of PER2 and CRY1 within the SCN, and emphasise the utility of translational switching to explore the functions of circadian proteins.
Collapse
Affiliation(s)
- David McManus
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Andrew P Patton
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Nicola J Smyllie
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Jason W Chin
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Michael H Hastings
- Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
47
|
Rashed N, Liu W, Zhou X, Bode AM, Luo X. The role of circadian gene CLOCK in cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119782. [PMID: 38871225 DOI: 10.1016/j.bbamcr.2024.119782] [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: 02/03/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024]
Abstract
Circadian Locomotor Output Cycles Kaput (CLOCK) is one of the circadian clock genes and is considered to be a fundamental regulatory gene in the circadian rhythm, responsible for mediating several biological processes. Therefore, abnormal expression of CLOCK affects its role in the circadian clock and its more general function as a direct regulator of gene expression. This dysfunction can lead to severe pathological effects, including cancer. To better understand the role of CLOCK in cancer, we compiled this review to describe the biological function of CLOCK, and especially highlighted its function in cancer development, progression, tumor microenvironment, cancer cell metabolism, and drug resistance.
Collapse
Affiliation(s)
- Nasot Rashed
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; NHC Key Laboratory of Carcinogenesis, the Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China
| | - Wenbin Liu
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; Department of Pathology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China
| | - Xinran Zhou
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; NHC Key Laboratory of Carcinogenesis, the Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Xiangjian Luo
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; NHC Key Laboratory of Carcinogenesis, the Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China; Key Laboratory of Biological Nanotechnology of National Health Commission, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
48
|
Bieuville M, Dujon A, Raven N, Ujvari B, Pujol P, Eslami‐S Z, Alix Panabières C, Capp J, Thomas F. When Do Tumours Develop? Neoplastic Processes Across Different Timescales: Age, Season and Round the Circadian Clock. Evol Appl 2024; 17:e70024. [PMID: 39444444 PMCID: PMC11496201 DOI: 10.1111/eva.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
While it is recognised that most, if not all, multicellular organisms harbour neoplastic processes within their bodies, the timing of when these undesirable cell proliferations are most likely to occur and progress throughout the organism's lifetime remains only partially documented. Due to the different mechanisms implicated in tumourigenesis, it is highly unlikely that this probability remains constant at all times and stages of life. In this article, we summarise what is known about this variation, considering the roles of age, season and circadian rhythm. While most studies requiring that level of detail be done on humans, we also review available evidence in other animal species. For each of these timescales, we identify mechanisms or biological functions shaping the variation. When possible, we show that evolutionary processes likely played a role, either directly to regulate the cancer risk or indirectly through trade-offs. We find that neoplastic risk varies with age in a more complex way than predicted by early epidemiological models: rather than resulting from mutations alone, tumour development is dictated by tissue- and age-specific processes. Similarly, the seasonal cycle can be associated with risk variation in some species with life-history events such as sexual competition or mating being timed according to the season. Lastly, we show that the circadian cycle influences tumourigenesis in physiological, pathological and therapeutic contexts. We also highlight two biological functions at the core of these variations across our three timescales: immunity and metabolism. Finally, we show that our understanding of the entanglement between tumourigenic processes and biological cycles is constrained by the limited number of species for which we have extensive data. Improving our knowledge of the periods of vulnerability to the onset and/or progression of (malignant) tumours is a key issue that deserves further investigation, as it is key to successful cancer prevention strategies.
Collapse
Affiliation(s)
- Margaux Bieuville
- CREEC (CREES), Unité Mixte de RecherchesIRD 224‐CNRS 5290‐Université de MontpellierMontpellierFrance
- Institute of Organismic and Molecular Evolution (iomE)Johannes Gutenberg‐UniversitätMainzGermany
- Institute for Quantitative and Computational Biosciences (IQCB)Johannes Gutenberg‐UniversitätMainzGermany
| | - Antoine M. Dujon
- School of Life and Environmental SciencesDeakin UniversityWaurn PondsVictoriaAustralia
| | - Nynke Raven
- School of Life and Environmental SciencesDeakin UniversityWaurn PondsVictoriaAustralia
| | - Beata Ujvari
- CREEC (CREES), Unité Mixte de RecherchesIRD 224‐CNRS 5290‐Université de MontpellierMontpellierFrance
- School of Life and Environmental SciencesDeakin UniversityWaurn PondsVictoriaAustralia
| | - Pascal Pujol
- CREEC (CREES), Unité Mixte de RecherchesIRD 224‐CNRS 5290‐Université de MontpellierMontpellierFrance
- Oncogenetic DepartmentUniversity Medical Centre of MontpellierMontpellierFrance
| | - Zahra Eslami‐S
- CREEC (CREES), Unité Mixte de RecherchesIRD 224‐CNRS 5290‐Université de MontpellierMontpellierFrance
- Laboratory of Rare Human Circulating Cells and Liquid Biopsy (LCCRH)University Medical Centre of MontpellierMontpellierFrance
- European Liquid Biopsy Society (ELBS)HamburgGermany
| | - Catherine Alix Panabières
- CREEC (CREES), Unité Mixte de RecherchesIRD 224‐CNRS 5290‐Université de MontpellierMontpellierFrance
- Laboratory of Rare Human Circulating Cells and Liquid Biopsy (LCCRH)University Medical Centre of MontpellierMontpellierFrance
- European Liquid Biopsy Society (ELBS)HamburgGermany
| | - Jean‐Pascal Capp
- Toulouse Biotechnology InstituteUniversity of Toulouse, INSA, CNRS, INRAEToulouseFrance
| | - Frédéric Thomas
- CREEC (CREES), Unité Mixte de RecherchesIRD 224‐CNRS 5290‐Université de MontpellierMontpellierFrance
| |
Collapse
|
49
|
Onuma S, Kawai M. Circadian Regulatory Networks of Glucose Homeostasis and Its Disruption as a Potential Cause of Undernutrition. Endocrinology 2024; 165:bqae126. [PMID: 39276035 DOI: 10.1210/endocr/bqae126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/09/2024] [Accepted: 09/12/2024] [Indexed: 09/16/2024]
Abstract
The circadian clock system, an evolutionarily conserved mechanism, orchestrates diurnal rhythms in biological activities such as behavior and metabolism, aligning them with the earth's 24-hour light/dark cycle. This synchronization enables organisms to anticipate and adapt to predictable environmental changes, including nutrient availability. However, modern lifestyles characterized by irregular eating and sleeping habits disrupt this synchrony, leading to metabolic disorders such as obesity and metabolic syndrome, evidenced by higher obesity rates among shift workers. Conversely, circadian disturbances are also associated with reduced nutrient absorption and an increased risk of malnutrition in populations such as the critically ill or the elderly. The precise mechanisms of these disturbances in leading to either overnutrition or undernutrition is complex and not yet fully understood. Glucose, a crucial energy source, is closely linked to obesity when consumed excessively and to weight loss when intake is reduced, which suggests that circadian regulation of glucose metabolism is a key factor connecting circadian disturbances with nutritional outcomes. In this review, we describe how the biological clock in various tissues regulates glucose metabolism, with a primary focus on studies utilizing animal models. Additionally, we highlight current clinical evidence supporting the association between circadian disturbance and glucose metabolism, arguing that such disruption could predominantly contribute to undernutrition due to impaired efficient utilization of nutrients.
Collapse
Affiliation(s)
- Shinsuke Onuma
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, 594-1101, Osaka, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Masanobu Kawai
- Department of Molecular Genetics and Endocrinology, Research Institute, Osaka Women's and Children's Hospital, 594-1101, Osaka, Japan
- Department of Gastroenterology, Nutrition and Endocrinology, Osaka Women's and Children's Hospital, 594-1101, Osaka, Japan
| |
Collapse
|
50
|
Miao L, Weidemann DE, Ngo K, Unruh BA, Kojima S. A Comparative Study of Algorithms Detecting Differential Rhythmicity in Transcriptomic Data. Bioinform Biol Insights 2024; 18:11779322241281188. [PMID: 39351295 PMCID: PMC11440551 DOI: 10.1177/11779322241281188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 08/19/2024] [Indexed: 10/04/2024] Open
Abstract
Rhythmic transcripts play pivotal roles in driving the daily oscillations of various biological processes. Genetic or environmental disruptions can lead to alterations in the rhythmicity of transcripts, ultimately impacting downstream circadian outputs, including metabolic processes and even behavior. To statistically compare the differences in transcript rhythms between 2 or more conditions, several algorithms have been developed to analyze circadian transcriptomic data, each with distinct features. In this study, we compared the performance of 7 algorithms that were specifically designed to detect differential rhythmicity (DODR, LimoRhyde, CircaCompare, compareRhythms, diffCircadian, dryR, and RepeatedCircadian). We found that even when applying the same statistical threshold, these algorithms yielded varying numbers of differentially rhythmic transcripts, most likely because each algorithm defines rhythmic and differentially rhythmic transcripts differently. Nevertheless, the output for the differential phase and amplitude were identical between dryR and compareRhyhms, and diffCircadian and CircaCompare, while the output from LimoRhyde2 was highly correlated with that from diffCircadian and CircaCompare. Because each algorithm has unique requirements for input data and reports different information as an output, it is crucial to ensure the compatibility of input data with the chosen algorithm and assess whether the algorithm's output fits the user's needs when selecting an algorithm for analysis.
Collapse
Affiliation(s)
- Lin Miao
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, USA
| | - Douglas E Weidemann
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, USA
| | - Katherine Ngo
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, USA
| | - Benjamin A Unruh
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, USA
| | - Shihoko Kojima
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|