1
|
Romerowicz-Misielak M, Kozioł K, Nowak S, Wojnarowska-Nowak R, Łuc K. Aminooxyacetic acid up-regulates the Cry1 and Bmal1 clock gene in a sirtuin 1 dependent manner. In vitro study. Toxicol Appl Pharmacol 2025; 499:117338. [PMID: 40210100 DOI: 10.1016/j.taap.2025.117338] [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: 12/31/2024] [Revised: 03/23/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
The regulation of the cyclic oscillation of the components of the circadian clock is complex in itself. Numerous clock interactions with processes and molecules present in cells further complicate this mechanism. Recently, the anti-aging protein Silencing Information Regulator Two family member, SIRT1, has been linked with the molecular circadian clock. In this study, we investigated the in vitro effect of aminooxyacetic acid on SIRT1 expression in relation to circadian dynamics of Cry1 and Bma11 expressions in serum shocked NIH-3 T3 and HaCaT cells. The study was carried out in the context of the inhibitory activity of aminooxyacetic acid against cystathionine-β-synthase and cystathionine-γ-lyase. We have shown that aminooxyacetic acid effectively inhibits SIRT1 transcription and synthesis, which, given the pleiotropic effects of sirtuin 1 on numerous metabolic pathways, may have other implications. We also found that AOAA contributes to up-regulation of the expression of the Cry1 and Bmal1 genes in cells. This effect does not appear to be related to inhibition of the activity of cystathionine-β-synthase and cystathionine-γ-lyase. At the same time, this does not deny the role of hydrogen sulphide, a product of the activity of these enzymes, in the regulation of the circadian clock.
Collapse
Affiliation(s)
| | - Katarzyna Kozioł
- Collegium Medicum, Faculty of Biotechnology, University of Rzeszow, 35-310 Rzeszow, Poland
| | - Sławomir Nowak
- Collegium Medicum, Faculty of Biotechnology, University of Rzeszow, 35-310 Rzeszow, Poland
| | - Renata Wojnarowska-Nowak
- Institute of Material Engineering, Centre for Microelectronics and Nanotechnology, University of Rzeszow, Poland
| | - Klaudia Łuc
- Collegium Medicum, Faculty of Biotechnology, University of Rzeszow, 35-310 Rzeszow, Poland
| |
Collapse
|
2
|
Bellier F, Walter A, Lecoin L, Chauveau F, Rouach N, Rancillac A. Astrocytes at the heart of sleep: from genes to network dynamics. Cell Mol Life Sci 2025; 82:207. [PMID: 40397158 DOI: 10.1007/s00018-025-05671-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 05/22/2025]
Abstract
Astrocytes have transcended their role from mere structural scaffolds to pivotal regulators of neural circuitry and sleep-wake dynamics. The strategic proximity of their fine processes to blood vessels and synapses positions them as key players in neurobiology, contributing to the tripartite synapse concept. Gap-junction proteins also enable astrocytes to form an extensive network interacting with neuronal assemblies to influence sleep physiology. Recent advances in genetic engineering, neuroimaging and molecular biology have deepened our understanding of astrocytic functions. This review highlights the different mechanisms by which astrocytes regulate sleep, notably through transcriptomic and morphological changes, as well as gliotransmission, whereby intracellular calcium (Ca2+) dynamics plays a significant role in modulating the sleep-wake cycle. In vivo optogenetic stimulation of astrocytes indeed induces ATP release, which is subsequently degraded into adenosine, modulating neuronal excitability in sleep-wake regulatory brain regions. Astrocytes also participate in synaptic plasticity, potentially modulating sleep-associated downscaling, a process essential for memory consolidation and preventing synaptic saturation. Although astrocytic involvement in synaptic maintenance is well supported, the precise molecular mechanisms linking these processes to sleep regulation remain to be elucidated. By highlighting astrocytes' multiple roles in sleep physiology, these insights deepen our understanding of sleep mechanisms and pave the way for improving sleep quality.
Collapse
Affiliation(s)
- Félix Bellier
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology-CIRB, Collège de France, CNRS UMR 7241/Inserm U1050, Université PSL, PSL-NEURO, 11, Place Marcelin Berthelot, 75005, Paris, France
- IRBA (Institut de Recherche Biomédicale Des Armées), Brétigny-sur-Orge, France
| | - Augustin Walter
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology-CIRB, Collège de France, CNRS UMR 7241/Inserm U1050, Université PSL, PSL-NEURO, 11, Place Marcelin Berthelot, 75005, Paris, France
| | - Laure Lecoin
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology-CIRB, Collège de France, CNRS UMR 7241/Inserm U1050, Université PSL, PSL-NEURO, 11, Place Marcelin Berthelot, 75005, Paris, France
| | - Fréderic Chauveau
- IRBA (Institut de Recherche Biomédicale Des Armées), Brétigny-sur-Orge, France
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology-CIRB, Collège de France, CNRS UMR 7241/Inserm U1050, Université PSL, PSL-NEURO, 11, Place Marcelin Berthelot, 75005, Paris, France
| | - Armelle Rancillac
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology-CIRB, Collège de France, CNRS UMR 7241/Inserm U1050, Université PSL, PSL-NEURO, 11, Place Marcelin Berthelot, 75005, Paris, France.
| |
Collapse
|
3
|
Wei T, Cheng Y, Ge J, Zhu M, Chen H, Feng Q. The Pro-Apoptotic Effect of Glucose Restriction in NSCLC via AMPK-Regulated Circadian Clock Gene Bmal1. Cancer Sci 2025. [PMID: 40394734 DOI: 10.1111/cas.70098] [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: 11/14/2024] [Revised: 04/19/2025] [Accepted: 05/01/2025] [Indexed: 05/22/2025] Open
Abstract
The circadian clock is a crucial regulator of mammalian physiology, controlling daily oscillations in key biological processes, such as cell proliferation, apoptosis, and DNA damage repair. Disruption of circadian rhythms has been identified as a significant risk factor for cancer development and progression, yet the specific molecular mechanisms linking circadian dysfunction to cancer remain poorly understood. Recent studies have increasingly focused on the role of diet in modulating circadian rhythms, highlighting the potential for dietary interventions in cancer management. However, how dietary factors like glucose restriction interact with circadian rhythms to influence cancer cell behavior remains an open question. Here, we investigate the mechanisms underlying glucose restriction-induced apoptosis in non-small cell lung cancer (NSCLC) cells, with a focus on the role of circadian clock genes. Analysis of the GEPIA database revealed that the circadian gene Bmal1 is highly expressed in normal tissues and associated with better prognosis in lung adenocarcinoma patients. In NSCLC cells, Bmal1 expression correlated with proapoptotic gene activity. In a tumor xenograft model using severe combined immunodeficiency (SCID) mice, a glucose-restricted (ketogenic) diet significantly delayed tumor growth and increased the expression of Bmal1 and proapoptotic genes. These findings suggest that glucose restriction promotes apoptosis in NSCLC cells through a Bmal1-mediated pathway, providing novel insights into the intersection between circadian regulation and cancer biology. Targeting core circadian clock genes like Bmal1 may represent a promising therapeutic strategy for managing lung cancer, broadening our understanding of how circadian rhythms can be harnessed for cancer prevention and treatment.
Collapse
Affiliation(s)
- Tao Wei
- Department of Nutrition and Food Hygiene, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Key Laboratory of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying Cheng
- Department of Nutrition and Food Hygiene, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Key Laboratory of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jierong Ge
- Department of Nutrition and Food Hygiene, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Key Laboratory of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Manting Zhu
- Department of Nutrition and Food Hygiene, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Key Laboratory of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hong Chen
- Department of Nutrition and Food Hygiene, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Key Laboratory of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qing Feng
- Department of Nutrition and Food Hygiene, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Key Laboratory of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Lago Solis B, Koch R, Nagoshi E. Circadian clock-independent ultradian rhythms in lipid metabolism in the Drosophila fat body. J Biol Chem 2025:110245. [PMID: 40383146 DOI: 10.1016/j.jbc.2025.110245] [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: 12/20/2024] [Revised: 04/19/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025] Open
Abstract
The role of circadian clocks in regulating metabolic processes is well known; however, their impact on metabolic states across species and life stages remains largely unexplored. This study investigates the relationship between circadian rhythms and metabolic regulation in the Drosophila larval fat body, a metabolic hub analogous to the mammalian liver and adipose tissue. Surprisingly, the fat body of period null mutants, which lack a functional circadian clock in all tissues, exhibited 12-hour rhythms in gene expression, particularly those involved in peroxisome function, lipid metabolism, and oxidative stress response. These transcriptomic rhythms were aligned with 12-hour oscillations in peroxisome biogenesis and activity, reactive oxygen species levels, and lipid peroxidation. Furthermore, period mutants exhibited 12-hour rhythms in body fat storage, ultimately leading to a net reduction in body fat levels. Collectively, our results identify clock-independent ultradian rhythms in lipid metabolism that are essential for larval survival and development.
Collapse
Affiliation(s)
- Blanca Lago Solis
- Department of Genetics and Evolution and Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, CH-1205, Geneva, Switzerland
| | - Rafael Koch
- Department of Genetics and Evolution and Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, CH-1205, Geneva, Switzerland
| | - Emi Nagoshi
- Department of Genetics and Evolution and Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, CH-1205, Geneva, Switzerland.
| |
Collapse
|
5
|
Hwang Y, Lee H, Lee MK. Conundrum and chances of diabetes management in the Western Pacific Region: A narrative review. J Diabetes Investig 2025. [PMID: 40371903 DOI: 10.1111/jdi.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/27/2025] [Accepted: 04/11/2025] [Indexed: 05/16/2025] Open
Abstract
The prevalence of diabetes is increasing globally, and glucose management is essential for the treatment of diabetes. Most guidelines recommend early intensive therapy and individualized approaches. Although many countries have implemented various guidelines and educational programs to enhance glucose management, the target achievement rate still remains very low. Studies from several countries and regions have identified various factors that influence blood glucose management, either positively or negatively. These factors have been comprehensively incorporated into guidelines to assist people with diabetes and healthcare professionals in following them and/or developing additional guidelines through further research. We and others have suggested that diverse factors should be considered-including comorbidities, age, complications, life expectancy, and pathophysiologic characteristics, such as ethnic differences in insulin sensitivity and secretion. The Western Pacific (WP) region, comprising countries with significant cultural and racial diversity, necessitates customized programs and community-based management strategies. In this review, we present specific challenges and opportunities for diabetes management identified through a systematic review of the literature from the WP region, along with those common to other regions. To improve healthcare policy and management in the WP region, it is essential to address regional characteristics and the factors that act as either barriers or facilitators to develop strategies for early intensive and individualized therapeutic approaches. Moreover, additional studies on diabetes pathophysiology and management-including pharmacotherapy-are urgently needed.
Collapse
Affiliation(s)
- Yerin Hwang
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Hyunmin Lee
- Department of Social and Preventive Medicine, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Moon-Kyu Lee
- Division of Endocrinology & Metabolism, Department of Internal Medicine, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu, South Korea
| |
Collapse
|
6
|
Khandayataray P, Murthy MK. Exploring the nexus: Sleep disorders, circadian dysregulation, and Alzheimer's disease. Neuroscience 2025; 574:21-41. [PMID: 40189132 DOI: 10.1016/j.neuroscience.2025.03.066] [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: 01/02/2025] [Revised: 03/10/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
We reviewed the connections among Alzheimer's disease (AD), sleep deprivation, and circadian rhythm disorders. Evidence is mounting that disrupted sleep and abnormal circadian rhythms are not merely symptoms of AD, but are also involved in accelerating the disease. Amyloid-beta (Aβ) accumulates, a feature of AD, and worsens with sleep deprivation because glymphatic withdrawal is required to clear toxic proteins from the brain. In addition, disturbances in circadian rhythm can contribute to the induction of neuroinflammation and oxidative stress, thereby accelerating neurodegenerative processes. While these interactions are bidirectional, Alzheimer's pathology further disrupts sleep and circadian function in a vicious cycle that worsens cognitive decline, which is emphasized in the review. The evidence that targeting sleep and circadian mechanisms may serve as therapeutic strategies for AD was strengthened by this study through the analysis of the molecular and physiological pathways. Further work on this nexus could help unravel the neurobiological mechanisms common to the onset of Alzheimer's and disrupted sleep and circadian regulation, which could result in earlier intervention to slow or prevent the onset of the disease.
Collapse
Affiliation(s)
- Pratima Khandayataray
- Department of Biotechnology, Academy of Management and Information Technology, Utkal University, Bhubaneswar, Odisha 752057, India
| | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Punjab 140401, India.
| |
Collapse
|
7
|
Liu C, Liu J, Shao J, Zhao X, Xie L, Shang M, Li Y, Li W. Single-cell and bulk transcriptome sequencing identifies circadian rhythm disruption and cluster-specific clinical insights in colorectal tumorigenesis. Discov Oncol 2025; 16:693. [PMID: 40338428 PMCID: PMC12062483 DOI: 10.1007/s12672-025-02521-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 04/28/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common malignant tumors in the digestive system worldwide, with its mortality ranking second among all cancers. Studies have indicated that disruptions in circadian rhythm (CR) are associated with the occurrence of various cancers; however, the relationship between CR and CRC requires further evidence, and research on the application of CR in CRC is still limited. METHODS In this study, we employed both bulk and single-cell RNA sequencing to explore the dysregulation of CR in patients with CRC. By constructing a CR subtype classifier, we conducted an in-depth analysis of the prognostic significance, the status of the tumor microenvironment, and response to immune checkpoint blockade (ICB) therapy between different CR clusters. Furthermore, we developed a CR scoring system (CRS) using machine learning to predict overall survival and identified several genes as potential targets affecting CRC prognosis. RESULTS Our findings revealed significant alterations in CR genes and status between CRC and normal tissues using bulk and single-cell transcriptome sequencing. Patients with CRC could be categorized into two distinct CR clusters (CR cluster 1 and 2). The prognosis of CR cluster 2, with higher epithelial-mesenchymal transition (EMT) and angiogenesis scores, was significantly worser than that of CR cluster 1. These clusters exhibited distinct levels of tumor-infiltrating lymphocytes. CR cluster 2 with a notably higher proportion of patients with microsatellite-instability-high (MSI-H), potentially benefit from ICB therapy. The proportion of patients belonging to consensus molecular subtype 4 (CMS4) in CR cluster 2 was also notably higher than in CR cluster 1. Additionally, the CRS combined with tumor stage demonstrated superior overall survival prediction efficacy compared to traditional tumor stage. We revealed a potential link between model genes (LSAMP, MS4A2, NAV3, RAB3B, SIX4) and the disruption of CR and patient prognosis. CONCLUSION This study not only provide new insights into the assessment of CR status in CRC patients but also develop a prognosis model based on CR-related genes, offering a new tool for personalized risk assessment in CRC.
Collapse
Affiliation(s)
- Chen Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China
| | - Jingyang Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China
| | - Jing Shao
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China
| | - Xiaoman Zhao
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China
| | - Lin Xie
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China
| | - Mengyao Shang
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China
| | - Ying Li
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China
| | - Weiming Li
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
8
|
Dashti HS, Jansen EC, Zuraikat FM, Dixit S, Brown M, Laposky A, Broussard JL, Butler MP, Creasy SA, Crispim CA, Depner CM, Esser KA, Garaulet M, Hanlon EC, Makarem N, Manoogian ENC, Peterson CM, Scheer FAJL, Wright KP, Goff DC, Pratt CA, Gamble KL, St-Onge MP. Advancing Chrononutrition for Cardiometabolic Health: A 2023 National Heart, Lung, and Blood Institute Workshop Report. J Am Heart Assoc 2025; 14:e039373. [PMID: 40265587 DOI: 10.1161/jaha.124.039373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
The circadian system maintains optimal biological functions at the appropriate time of day, and the disruption of this organization can contribute to the pathogenesis of cardiometabolic disorders. The timing of eating is a prominent external time cue that influences the circadian system. "Chrononutrition" is an emerging dimension of nutrition and active area of research that examines how timing-related aspects of eating and nutrition impact circadian rhythms, biological processes, and disease pathogenesis. There is evidence to support chrononutrition as a form of chronotherapy, such that optimizing the timing of eating may serve as an actionable strategy to improve cardiometabolic health. This report summarizes key information from the National Heart, Lung, and Blood Institute's virtual workshop entitled "Chrononutrition: Elucidating the Role of Circadian Biology and Meal Timing in Cardiometabolic Health," which convened on May 2 to 3, 2023, to review current literature and identify critical knowledge gaps and research opportunities. The speakers presented evidence highlighting the impact on cardiometabolic health of earlier and shorter eating windows and more consistent day-to-day eating patterns. The multidimensionality of chrononutrition was a common theme, as it encompasses multiple facets of eating along with the timing of other behaviors including sleep and physical activity. Advancing the emerging field of chrononutrition will require: (1) standardization of terminology and metrics; (2) scalable and precise tools for real-world settings; (3) consideration of individual differences that may act as effect modifiers; and (4) deeper understanding of social, behavioral, and cultural influences. Ultimately, there is great potential for circadian-based dietary interventions to improve cardiometabolic health.
Collapse
Affiliation(s)
- Hassan S Dashti
- Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Boston MA USA
- Division of Nutrition Harvard Medical School Boston MA USA
- Division of Sleep Medicine Harvard Medical School Boston MA USA
- Broad Institute Cambridge MA USA
| | - Erica C Jansen
- Department of Nutritional Sciences University of Michigan School of Public Health Ann Arbor MI USA
- Department of Neurology University of Michigan Ann Arbor MI USA
| | - Faris M Zuraikat
- Center of Excellence for Sleep and Circadian Research, Department of Medicine Columbia University Irving Medical Center New York NY USA
- Division of General Medicine, Department of Medicine Columbia University Irving Medical Center New York NY USA
- Institute of Human Nutrition, Columbia University Irving Medical Center New York NY USA
| | - Shilpy Dixit
- National Center on Sleep Disorders Research National Heart, Lung, and Blood Institute, National Institutes of Health Bethesda MD USA
| | - Marishka Brown
- National Center on Sleep Disorders Research National Heart, Lung, and Blood Institute, National Institutes of Health Bethesda MD USA
| | - Aaron Laposky
- National Center on Sleep Disorders Research National Heart, Lung, and Blood Institute, National Institutes of Health Bethesda MD USA
| | - Josiane L Broussard
- Department of Health and Exercise Science Colorado State University Fort Collins CO USA
- Ludeman Family Center for Women's Health Research University of Colorado Anschutz Medical Campus Aurora CO USA
- Division of Endocrinology, Metabolism, and Diabetes University of Colorado Anschutz Medical Campus Aurora CO USA
- Department of Integrative Physiology University of Colorado Boulder Boulder CO USA
| | - Matthew P Butler
- Oregon Institute of Occupational Health Sciences Oregon Health and Sciences University Portland OR USA
- Department of Behavioral Neuroscience, School of Medicine Oregon Health and Sciences University Portland OR USA
| | - Seth A Creasy
- Division of Endocrinology, Metabolism, and Diabetes University of Colorado Anschutz Medical Campus Aurora CO USA
- Anschutz Health and Wellness Center University of Colorado Anschutz Medical Campus Aurora CO USA
| | - Cibele A Crispim
- Chrononutrition Research Group, School of Medicine Federal University of Uberlândia Minas Gerais Brazil
| | | | - Karyn A Esser
- Department of Physiology and Aging, College of Medicine University of Florida Gainesville FL USA
| | - Marta Garaulet
- Department of Physiology, Regional Campus of International Excellence University of Murcia Spain
- Biomedical Research Institute of Murcia, IMIB-Arrixaca-UMU, University Clinical Hospital Murcia Spain
- Division of Sleep and Circadian Disorders, Department of Medicine and Neurology Brigham and Women's Hospital Boston MA USA
| | - Erin C Hanlon
- Section of Adult and Pediatric Endocrinology, Department of Medicine University of Chicago IL USA
| | - Nour Makarem
- Department of Epidemiology, Mailman School of Public Health Columbia University Irving Medical Center New York NY USA
| | - Emily N C Manoogian
- Regulatory Biology Department Salk Institute for Biological Sciences La Jolla CA USA
| | - Courtney M Peterson
- Department of Nutrition Sciences University of Alabama at Birmingham Birmingham AL USA
| | - Frank A J L Scheer
- Division of Nutrition Harvard Medical School Boston MA USA
- Division of Sleep Medicine Harvard Medical School Boston MA USA
- Broad Institute Cambridge MA USA
- Division of Sleep and Circadian Disorders, Department of Medicine and Neurology Brigham and Women's Hospital Boston MA USA
| | - Kenneth P Wright
- Division of Endocrinology, Metabolism, and Diabetes University of Colorado Anschutz Medical Campus Aurora CO USA
- Department of Integrative Physiology University of Colorado Boulder Boulder CO USA
| | - David C Goff
- Division of Cardiovascular Sciences National Heart, Lung, and Blood Institute, National Institutes of Health Bethesda MD USA
| | - Charlotte A Pratt
- Division of Cardiovascular Sciences National Heart, Lung, and Blood Institute, National Institutes of Health Bethesda MD USA
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, School of Medicine University of Alabama at Birmingham Birmingham AL USA
- Nutrition Obesity Research Center University of Alabama at Birmingham Birmingham AL USA
| | - Marie-Pierre St-Onge
- Center of Excellence for Sleep and Circadian Research, Department of Medicine Columbia University Irving Medical Center New York NY USA
- Division of General Medicine, Department of Medicine Columbia University Irving Medical Center New York NY USA
| |
Collapse
|
9
|
Feng J, Zhu L, He C, Xiang R, Liu J, Cai J, Wang D. Lactate induces oxidative stress by HIF1α stabilization and circadian clock disturbance in mammary gland of dairy cows. J Anim Sci Biotechnol 2025; 16:62. [PMID: 40307878 PMCID: PMC12044779 DOI: 10.1186/s40104-025-01181-1] [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: 09/26/2024] [Accepted: 02/16/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Lactate is a classical byproduct of glucose metabolism, and the main lactate production pathway depends on glycolysis. Lactate stabilized HIF1α by inhibiting PHD activity, leading to hypoxic stress response and exacerbating glycolysis in multiple tissues. However, the redox induction mechanism of lactate in mammary gland has not been understood yet. Herein, we describe a lactate-responsive HIF1α/circadian control mechanism in oxidative stress in the mammary glands of dairy cows. RESULTS The in vivo study showed that dairy cows with high lactate concentrations are associated with reduced milk yield and more ROS accumulation in mammary gland. Western blot results in MAC-T cells showed positive correlation between lactate concentrations, expression of HIF1α and oxidative stress indicators, but not circadian core components. To test how lactate-mediated HIF1α dysfunction leads to cell protection process, we investigated altered expression of circadian core related genes following HIF1α stabilization. We found that stabilized HIF1α by lactate inhibited stimulated expression of circadian core components due to the similarity of HRE and E-box transcription elements. Furthermore, we found that lactate treatment strengthened the binding of HIF1α with BMAL1, HMOX1 and FOXO3 in MAC-T cells. Moreover, HIF1α knockdown altered expression of circadian rhythm related genes and reduced oxidative stress state. CONCLUSION In summary, our study highlights the central role of competitive transcriptional element occupancy in lactate-mediated oxidative stress of mammary gland, which is caused by HIF1α stabilization and circadian rhythm dysfunction. Our findings introduce a novel nutritional strategy with potential applications in dairy farming for optimizing milk production and maintaining mammary gland health.
Collapse
Affiliation(s)
- Juan Feng
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Lei Zhu
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Cunman He
- College of Life Science, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Ruidong Xiang
- Agriculture Victoria Research, AgriBio Centre for AgriBioscience, Bundoora, VIC, 3052, Australia
- School of Applied Systems Biology, La Trobe University, Bundoora, VIC, 3052, Australia
| | - Jianxin Liu
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Jie Cai
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China.
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China.
| | - Diming Wang
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China.
| |
Collapse
|
10
|
Li H, Seugnet L. Decoding the nexus: branched-chain amino acids and their connection with sleep, circadian rhythms, and cardiometabolic health. Neural Regen Res 2025; 20:1350-1363. [PMID: 39075896 PMCID: PMC11624887 DOI: 10.4103/nrr.nrr-d-23-02020] [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: 12/13/2023] [Revised: 04/18/2024] [Accepted: 05/12/2024] [Indexed: 07/31/2024] Open
Abstract
The sleep-wake cycle stands as an integrative process essential for sustaining optimal brain function and, either directly or indirectly, overall body health, encompassing metabolic and cardiovascular well-being. Given the heightened metabolic activity of the brain, there exists a considerable demand for nutrients in comparison to other organs. Among these, the branched-chain amino acids, comprising leucine, isoleucine, and valine, display distinctive significance, from their contribution to protein structure to their involvement in overall metabolism, especially in cerebral processes. Among the first amino acids that are released into circulation post-food intake, branched-chain amino acids assume a pivotal role in the regulation of protein synthesis, modulating insulin secretion and the amino acid sensing pathway of target of rapamycin. Branched-chain amino acids are key players in influencing the brain's uptake of monoamine precursors, competing for a shared transporter. Beyond their involvement in protein synthesis, these amino acids contribute to the metabolic cycles of γ-aminobutyric acid and glutamate, as well as energy metabolism. Notably, they impact GABAergic neurons and the excitation/inhibition balance. The rhythmicity of branched-chain amino acids in plasma concentrations, observed over a 24-hour cycle and conserved in rodent models, is under circadian clock control. The mechanisms underlying those rhythms and the physiological consequences of their disruption are not fully understood. Disturbed sleep, obesity, diabetes, and cardiovascular diseases can elevate branched-chain amino acid concentrations or modify their oscillatory dynamics. The mechanisms driving these effects are currently the focal point of ongoing research efforts, since normalizing branched-chain amino acid levels has the ability to alleviate the severity of these pathologies. In this context, the Drosophila model, though underutilized, holds promise in shedding new light on these mechanisms. Initial findings indicate its potential to introduce novel concepts, particularly in elucidating the intricate connections between the circadian clock, sleep/wake, and metabolism. Consequently, the use and transport of branched-chain amino acids emerge as critical components and orchestrators in the web of interactions across multiple organs throughout the sleep/wake cycle. They could represent one of the so far elusive mechanisms connecting sleep patterns to metabolic and cardiovascular health, paving the way for potential therapeutic interventions.
Collapse
Affiliation(s)
- Hui Li
- Department of Neurology, Xijing Hospital, Xi’an, Shaanxi Province, China
| | - Laurent Seugnet
- Centre de Recherche en Neurosciences de Lyon, Integrated Physiology of the Brain Arousal Systems (WAKING), Université Claude Bernard Lyon 1, INSERM U1028, CNRS UMR 5292, Bron, France
| |
Collapse
|
11
|
Pu H, Bailey LC, Bauer LG, Voronkov M, Baxter M, Huber KVM, Khorasanizadeh S, Ray D, Rastinejad F. Pharmacological targeting of BMAL1 modulates circadian and immune pathways. Nat Chem Biol 2025; 21:736-745. [PMID: 40133642 PMCID: PMC12037410 DOI: 10.1038/s41589-025-01863-x] [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: 03/24/2024] [Accepted: 02/14/2025] [Indexed: 03/27/2025]
Abstract
The basic helix-loop-helix PER-ARNT-SIM (bHLH-PAS) proteins BMAL1 and CLOCK heterodimerize to form the master transcription factor governing rhythmic gene expression. Owing to connections between circadian regulation and numerous physiological pathways, targeting the BMAL1-CLOCK complex pharmacologically is an attractive entry point for intervening in circadian-related processes. In this study, we developed a small molecule, Core Circadian Modulator (CCM), that targets the cavity in the PASB domain of BMAL1, causing it to expand, leading to conformational changes in the PASB domain and altering the functions of BMAL1 as a transcription factor. Biochemical, structural and cellular investigations validate the high level of selectivity of CCM in engaging BMAL1, enabling direct access to BMAL1-CLOCK cellular activities. CCM induces dose-dependent alterations in PER2-Luc oscillations and orchestrates the downregulation of inflammatory and phagocytic pathways in macrophages. These findings collectively reveal that the BMAL1 protein architecture is inherently configured to enable the binding of chemical ligands for functional modulation.
Collapse
Affiliation(s)
- Hua Pu
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
| | - Laura C Bailey
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| | - Ludwig G Bauer
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Maria Voronkov
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| | - Matthew Baxter
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| | - Kilian V M Huber
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Sepideh Khorasanizadeh
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
| | - David Ray
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| | - Fraydoon Rastinejad
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, UK.
| |
Collapse
|
12
|
Song QX, Suadicani SO, Negoro H, Jiang HH, Jabr R, Fry C, Xue W, Damaser MS. Disruption of circadian rhythm as a potential pathogenesis of nocturia. Nat Rev Urol 2025; 22:276-293. [PMID: 39543359 DOI: 10.1038/s41585-024-00961-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 11/17/2024]
Abstract
Increasing evidence suggested the multifactorial nature of nocturia, but the true pathogenesis of this condition still remains to be elucidated. Contemporary clinical medications are mostly symptom based, aimed at either reducing nocturnal urine volume or targeting autonomic receptors within the bladder to facilitate urine storage. The day-night switch of the micturition pattern is controlled by circadian clocks located both in the central nervous system and in the peripheral organs. Arousal threshold and secretion of melatonin and vasopressin increase at night-time to achieve high-quality sleep and minimize nocturnal urine production. In response to the increased vasopressin, the kidney reduces the glomerular filtration rate and facilitates the reabsorption of water. Synchronously, in the bladder, circadian oscillation of crucial molecules occurs to reduce afferent sensory input and maintain sufficient bladder capacity during the night sleep period. Thus, nocturia might occur as a result of desynchronization in one or more of these circadian regulatory mechanisms. Disrupted rhythmicity of the central nervous system, kidney and bladder (known as the brain-kidney-bladder circadian axis) contributes to the pathogenesis of nocturia. Novel insights into the chronobiological nature of nocturia will be crucial to promote a revolutionary shift towards effective therapeutics targeting the realignment of the circadian rhythm.
Collapse
Affiliation(s)
- Qi-Xiang Song
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sylvia O Suadicani
- Department of Urology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hiromitsu Negoro
- Department of Urology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hai-Hong Jiang
- Department of Urology and Andrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rita Jabr
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Christopher Fry
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Wei Xue
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Margot S Damaser
- Department of Biomedical Engineering, Lerner Research Institute and Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA.
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.
| |
Collapse
|
13
|
Deng Y, Zhang Y, Xiao J, Cao Y, Ho CT, Lu M. Allicin Improves Diet-Induced Nonalcoholic Steatohepatitis and Gut Microbiota Dysbiosis in Mice via the Involvement of the Circadian Clock Gene Rev-erbα. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9019-9032. [PMID: 40168418 DOI: 10.1021/acs.jafc.4c12566] [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: 04/03/2025]
Abstract
Nonalcoholic Steatohepatitis (NASH) is a progressive liver disease characterized by inflammation and liver damage. Allicin, a bioactive compound derived from garlic, has demonstrated anti-inflammatory and antioxidant properties. This study explores the effects of allicin on NASH and gut microbiota dysbiosis induced by a high-fat, high-fructose diet (HFFD) in mice. Allicin supplementation significantly alleviated hepatic inflammation, improved glucose metabolism, and modulated the circadian rhythm gene Rev-erbα, which plays a critical role in regulating inflammation. The anti-inflammatory effects of allicin were diminished in Si-Rev-erbα-treated HepG2 cells, highlighting the importance of circadian regulation in mediating these effects. Allicin's anti-inflammatory effects were associated with increased levels of short-chain fatty acids (SCFAs) and the restoration of diurnal oscillations in proinflammatory cytokines and gut microbiota composition, particularly in genera, such as Akkermansia, Bacteroidetes, and Lactobacillus. These findings suggest that allicin could be a promising therapeutic approach for managing NASH, liver dysfunction, and related metabolic disorders through the modulation of circadian rhythms and the gut microbiome.
Collapse
Affiliation(s)
- Yupei Deng
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yiyi Zhang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Muwen Lu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
14
|
Nagao Y, Taguchi A, Ohta Y. Circadian Rhythm Dysregulation in Inflammatory Bowel Disease: Mechanisms and Chronotherapeutic Approaches. Int J Mol Sci 2025; 26:3724. [PMID: 40332348 PMCID: PMC12028002 DOI: 10.3390/ijms26083724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/06/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Inflammatory bowel disease (IBD), comprising ulcerative colitis (UC) and Crohn's disease (CD), is characterized by chronic intestinal inflammation. Recent research has highlighted the significant interplay between IBD pathogenesis and circadian rhythms. This review synthesizes current evidence regarding circadian regulation in IBD, covering three main areas: (1) circadian rhythms in intestinal physiology, (2) circadian disruption patterns in IBD patients, and (3) the role of clock genes in IBD pathogenesis. We discuss how these findings may inform novel chronotherapeutic approaches for IBD treatment. Future research directions that could facilitate translation of chronobiological insights into clinical applications are also explored.
Collapse
Affiliation(s)
- Yuko Nagao
- Health Science Center, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8511, Japan
| | - Akihiko Taguchi
- Department of Endocrinology, Metabolism, Hematological Science and Therapeutics, Graduate School of Medicine, Yamaguchi University, 1-1-1, Minami Kogushi, Ube 755-8505, Japan;
| | - Yasuharu Ohta
- Department of Endocrinology, Metabolism, Hematological Science and Therapeutics, Graduate School of Medicine, Yamaguchi University, 1-1-1, Minami Kogushi, Ube 755-8505, Japan;
| |
Collapse
|
15
|
Feeney SP, McCarthy JM, Petruconis CR, Tudor JC. Sleep loss is a metabolic disorder. Sci Signal 2025; 18:eadp9358. [PMID: 40198749 DOI: 10.1126/scisignal.adp9358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 03/12/2025] [Indexed: 04/10/2025]
Abstract
Sleep loss dysregulates cellular metabolism and energy homeostasis. Highly metabolically active cells, such as neurons, enter a catabolic state during periods of sleep loss, which consequently disrupts physiological functioning. Specific to the central nervous system, sleep loss results in impaired synaptogenesis and long-term memory, effects that are also characteristic of neurodegenerative diseases. In this review, we describe how sleep deprivation increases resting energy expenditure, leading to the development of a negative energy balance-a state with insufficient metabolic resources to support energy expenditure-in highly active cells like neurons. This disruption of energetic homeostasis alters the balance of metabolites, including adenosine, lactate, and lipid peroxides, such that energetically costly processes, such as synapse formation, are attenuated. During sleep loss, metabolically active cells shunt energetic resources away from those processes that are not acutely essential, like memory formation, to support cell survival. Ultimately, these findings characterize sleep loss as a metabolic disorder.
Collapse
Affiliation(s)
- Sierra P Feeney
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Jordan M McCarthy
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Cecilia R Petruconis
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Jennifer C Tudor
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| |
Collapse
|
16
|
Ren X, Wang W, Li W, Sun L, Liu T, Zhou H, Han T, Sun C, Lu X, Tian W. Circadian rest-activity rhythms and multimorbidity and mortality risks among menopausal women: a trajectory analysis of a UK Biobank cohort. BMC Public Health 2025; 25:1304. [PMID: 40197377 PMCID: PMC11974044 DOI: 10.1186/s12889-025-22536-3] [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: 04/30/2024] [Accepted: 03/28/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Menopausal women undergo substantial physiological changes that can impact their overall health. OBJECTIVES We examined relationships between circadian rest-activity rhythms (CRARs) and multimorbidity progression in this population. METHODS We used UK Biobank data, involving 10,138 participants, who were initially free of chronic conditions. We primarily focused on the relative amplitude (RA) of CRARs, tracking incident first chronic conditions (FCC), multimorbidity, and all-cause mortality. Multimorbidity was indicated by the presence of any 2/35 chronic conditions during the follow-up period. We used a multi-state model to assess the RA impact on the multimorbidity progression trajectory, encompassing transition from health to an FCC, to consequent multimorbidity, and ultimately to mortality, in parallel with sensitivity analyses to ensure results stability and reliability. RESULTS During a mean 8.13-year follow-up period, we identified 855 incident multimorbidity cases and recorded 88 deaths. In a multi-state model, a lower RA was associated with an increased risk of transition from health to FCC onset [hazard ratio (HR): 1.18, 95% confidence interval (CI): 1.07-1.31] and also from an FCC to multimorbidity development (HR: 1.34, 95% CI: 1.12-1.61), even after adjusting for several confounding factors. CONCLUSIONS Among menopausal women, circadian rhythm disturbance increased the risk of transitioning from health to a single chronic condition, as well as transitioning from a single chronic condition to multimorbidity.
Collapse
Affiliation(s)
- Xiyun Ren
- Department of Epidemiology, School of Public Health, Harbin Medical University, Heilongjiang Province, Harbin, 150081, P.R. China
| | - Wentong Wang
- Department of Epidemiology, School of Public Health, Harbin Medical University, Heilongjiang Province, Harbin, 150081, P.R. China
| | - Wei Li
- Department of Epidemiology, School of Public Health, Harbin Medical University, Heilongjiang Province, Harbin, 150081, P.R. China
| | - Lishuang Sun
- Department of Epidemiology, School of Public Health, Harbin Medical University, Heilongjiang Province, Harbin, 150081, P.R. China
| | - Tianyu Liu
- Department of Epidemiology, School of Public Health, Harbin Medical University, Heilongjiang Province, Harbin, 150081, P.R. China
| | - Haibo Zhou
- Department of Epidemiology, School of Public Health, Harbin Medical University, Heilongjiang Province, Harbin, 150081, P.R. China
| | - Tianshu Han
- Key Laboratory of Precision Nutrition and Health, Ministry of Education. Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Heilongjiang Province, Harbin, 150081, P.R. China
| | - Changhao Sun
- Key Laboratory of Precision Nutrition and Health, Ministry of Education. Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Heilongjiang Province, Harbin, 150081, P.R. China
| | - Xiangfeng Lu
- Key Laboratory of Cardiovascular Epidemiology &Department of Epidemiologynational Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, P.R. China.
| | - Wenjing Tian
- Department of Epidemiology, School of Public Health, Harbin Medical University, Heilongjiang Province, Harbin, 150081, P.R. China.
| |
Collapse
|
17
|
Preza S, Zheng B, Gao Z, Liu M, Biju A, Alvarez-Dominguez JR. DEC1 Regulates Human β Cell Functional Maturation and Circadian Rhythm. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.647023. [PMID: 40236051 PMCID: PMC11996484 DOI: 10.1101/2025.04.03.647023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Stem cell-derived islet (SC-islet) organoids offer hope for cell replacement therapy in diabetes, but their immature function remains a challenge. Mature islet function requires the β-cell circadian clock, yet how the clock regulates maturation is unclear. Here, we show that a circadian transcription factor specific to maturing SC-β cells, DEC1, regulates insulin responsiveness to glucose. SC-islet organoids form normally from DEC1 -ablated human pluripotent stem cells, but their insulin release capacity and glucose threshold fail to increase during in vitro culture and upon transplant. This deficit reflects downregulation of maturity-linked effectors of glucose utilization and insulin exocytosis, blunting glycolytic and oxidative metabolism, and is rescued by increasing metabolic flux. Moreover, DEC1 is needed to boost SC-islet maturity by synchronizing circadian glucose-responsive insulin secretion rhythms and clock machinery. Thus, DEC1 links circadian control to human β-cell maturation, highlighting its vitality to foster fully functional SC-islets.
Collapse
|
18
|
Montalvo Landivar AP, Gao Z, Liu M, Gruskin ZL, Leduc A, Preza S, Xie Y, Rozo AV, Ahn JH, Straubhaar JR, Doliba N, Stoffers DA, Slavov N, Alvarez-Dominguez JR. An adult clock regulator links circadian rhythms to pancreatic β-cell maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.08.11.552890. [PMID: 37609178 PMCID: PMC10441398 DOI: 10.1101/2023.08.11.552890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The circadian clock attunes metabolism to daily energy cycles, but how it regulates maturation of metabolic tissues is poorly understood. Here we show that DEC1, a clock transcription factor induced in adult islet β cells, coordinates their glucose responsiveness by synchronizing energetic and secretory rhythms. DEC1 binds and regulates maturity-linked genes to integrate insulin exocytosis with energy metabolism, and β-cell Dec1 ablation disrupts their transcription synchrony. Dec1-disrupted mice develop lifelong glucose intolerance and insulin deficiency, despite normal islet formation and intact Clock/Bmal1 genes. Metabolic dysfunction upon β-cell Dec1 loss stems from poor coupling of insulin secretion to glucose metabolism, reminiscent of fetal/neonatal immaturity. We link stunted maturation to a deficit in circadian bioenergetics, prompted by compromised glucose utilization, mitochondrial dynamics, and respiratory metabolism, which is rescued by increased metabolic flux. Thus, DEC1 links circadian clockwork to β-cell metabolic maturation, revealing a hierarchy for how the clock programs metabolic tissue specialization.
Collapse
Affiliation(s)
- Ana P. Montalvo Landivar
- Department of Neurosurgery, Brigham and Women’s Hospital; Boston, MA 02115, USA
- These authors contributed equally to this work
| | - Zihan Gao
- Chronobiology and Sleep Institute, and Institute for Regenerative Medicine, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania; Philadelphia, PA 19104, USA
- These authors contributed equally to this work
| | - Mai Liu
- Chronobiology and Sleep Institute, and Institute for Regenerative Medicine, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania; Philadelphia, PA 19104, USA
- These authors contributed equally to this work
| | - Zoe L. Gruskin
- Chronobiology and Sleep Institute, and Institute for Regenerative Medicine, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
| | - Andrew Leduc
- Departments of Bioengineering and Biology, Single-Cell Proteomics Center and Barnett Institute, Northeastern University; Boston, MA 02115, USA
| | - Sam Preza
- Chronobiology and Sleep Institute, and Institute for Regenerative Medicine, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania; Philadelphia, PA 19104, USA
| | - Yu Xie
- Chronobiology and Sleep Institute, and Institute for Regenerative Medicine, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania; Philadelphia, PA 19104, USA
| | - Andrea V. Rozo
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
| | - June H. Ahn
- Chronobiology and Sleep Institute, and Institute for Regenerative Medicine, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
| | - Juerg R. Straubhaar
- Bioinformatics Center, Massachusetts Eye and Ear Infirmary; Boston, MA 02114, USA
| | - Nicolai Doliba
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
| | - Doris A. Stoffers
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
| | - Nikolai Slavov
- Departments of Bioengineering and Biology, Single-Cell Proteomics Center and Barnett Institute, Northeastern University; Boston, MA 02115, USA
| | - Juan R. Alvarez-Dominguez
- Chronobiology and Sleep Institute, and Institute for Regenerative Medicine, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104, USA
- Lead contact
| |
Collapse
|
19
|
Cela O, Scrima R, Rosiello M, Pacelli C, Piccoli C, Tamma M, Agriesti F, Mazzoccoli G, Capitanio N. Circadian clockwork controls the balance between mitochondrial turnover and dynamics: What is life … without time marking? BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2025; 1866:149542. [PMID: 39880150 DOI: 10.1016/j.bbabio.2025.149542] [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: 06/12/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Circadian rhythms driven by biological clocks regulate physiological processes in all living organisms by anticipating daily geophysical changes, thus enhancing environmental adaptation. Time-resolved serial multi-omic analyses in vivo, ex vivo, and in synchronized cell cultures have revealed rhythmic changes in the transcriptome, proteome, and metabolome, involving up to 50 % of the mammalian genome. Mitochondrial oxidative metabolism is central to cellular bioenergetics, and many nuclear genes encoding mitochondrial proteins exhibit both circadian and ultradian oscillatory expression. However, studies on mitochondrial DNA (mtDNA) gene expression remain incomplete. Using a well-established in vitro synchronization protocol, we investigated the time-resolved expression of mtDNA genes coding for respiratory chain complex subunits, revealing a rhythmic profile dependent on BMAL1, the master circadian clock transcription factor. Additionally, the expression of genes coding for key mitochondrial biogenesis transcription factors, PGC1a, NRF1, and TFAM, showed BMAL1-dependent circadian oscillations. Notably, LC3-II, involved in mitophagy, displayed a similar in-phase circadian expression, thereby maintaining stable respiratory chain complex levels. Moreover, we found that simultaneous mitochondrial biogenesis and degradation occur in a coordinated manner with cycles in organelle dynamics, leading to rhythmic changes in mitochondrial fission and fusion. This study provides new insights into circadian clock regulation of mitochondrial turnover, emphasizing the importance of temporal regulation in cellular metabolism. Understanding these mechanisms opens potential therapeutic avenues for targeting mitochondrial dysfunctions and related metabolic disorders.
Collapse
Affiliation(s)
- Olga Cela
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Rosella Scrima
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Michela Rosiello
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Mirko Tamma
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Francesca Agriesti
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Gianluigi Mazzoccoli
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Laboratory, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Italy
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| |
Collapse
|
20
|
Urquiza-García U, Molina N, Halliday KJ, Millar AJ. Abundant clock proteins point to missing molecular regulation in the plant circadian clock. Mol Syst Biol 2025; 21:361-389. [PMID: 39979593 PMCID: PMC11965494 DOI: 10.1038/s44320-025-00086-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/20/2024] [Accepted: 01/03/2025] [Indexed: 02/22/2025] Open
Abstract
Understanding the biochemistry behind whole-organism traits such as flowering time is a longstanding challenge, where mathematical models are critical. Very few models of plant gene circuits use the absolute units required for comparison to biochemical data. We refactor two detailed models of the plant circadian clock from relative to absolute units. Using absolute RNA quantification, a simple model predicted abundant clock protein levels in Arabidopsis thaliana, up to 100,000 proteins per cell. NanoLUC reporter protein fusions validated the predicted levels of clock proteins in vivo. Recalibrating the detailed models to these protein levels estimated their DNA-binding dissociation constants (Kd). We estimate the same Kd from multiple results in vitro, extending the method to any promoter sequence. The detailed models simulated the Kd range estimated from LUX DNA-binding in vitro but departed from the data for CCA1 binding, pointing to further circadian mechanisms. Our analytical and experimental methods should transfer to understand other plant gene regulatory networks, potentially including the natural sequence variation that contributes to evolutionary adaptation.
Collapse
Affiliation(s)
- Uriel Urquiza-García
- Centre for Engineering Biology and School of Biological Sciences, C. H. Waddington Building, University of Edinburgh, King's Buildings, Edinburgh, EH9 3BF, UK
- Institute of Synthetic Biology, University of Düsseldorf, Düsseldorf, Germany
- CEPLAS-Cluster of Excellence on Plant Sciences, Düsseldorf, Germany
| | - Nacho Molina
- Centre for Engineering Biology and School of Biological Sciences, C. H. Waddington Building, University of Edinburgh, King's Buildings, Edinburgh, EH9 3BF, UK
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) CNRS UMR 7104, INSERM U964, Université de Strasbourg, 1 Rue Laurent Fries, 67404, Illkirch, France
| | - Karen J Halliday
- School of Biological Sciences, Daniel Rutherford Building, University of Edinburgh, King's Buildings, Edinburgh, EH9 3BF, UK
| | - Andrew J Millar
- Centre for Engineering Biology and School of Biological Sciences, C. H. Waddington Building, University of Edinburgh, King's Buildings, Edinburgh, EH9 3BF, UK.
| |
Collapse
|
21
|
Tice AL, Lee C, Hickner RC, Steiner JL. Scheduled Exercise Partially Offsets Alcohol-Induced Clock Dysfunction in Skeletal Muscle and Liver of Female Mice. J Biol Rhythms 2025; 40:208-228. [PMID: 39924857 DOI: 10.1177/07487304241312461] [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: 02/11/2025]
Abstract
Binge and chronic alcohol intake impair skeletal muscle and liver circadian clocks. Scheduled exercise is suggested to protect against circadian misalignment, like that induced by alcohol. It was tested whether scheduled, voluntary daily wheel running would protect the gastrocnemius and liver clocks against alcohol-induced perturbations. Female C57BL6/Hsd mice were assigned to 1 of 4 groups: control-sedentary (CON SED, n = 26), control-exercise (CON EX, n = 28), alcohol-sedentary (ETOH SED, n = 27), or alcohol-exercise (ETOH EX, n = 25). Exercise mice were granted access to running wheels for 2 h/day (ZT13-15) while ETOH mice consumed alcohol-containing liquid diet for 6 weeks. Tissues were collected every 4 h starting at ZT12 from 4-5 mice/group and were used for RNA/cDNA/RT-PCR (gastrocnemius and liver) and Western blotting (gastrocnemius). A second cohort of mice were weaned off alcohol, given regular chow, and continued daily exercise (2 h/day) for ~2 weeks. Then, all mice (EX and SED) were given 24-h wheel access for 1 week to assess cyclic running behaviors during abstinence. While alcohol differentially disrupted muscle and liver clocks in sedentary mice, differences between exercised groups were minimized. BMAL1 protein expression increased in the nuclear-enriched fraction in the gastrocnemius of both exercise groups compared to both sedentary groups. In the second cohort, wheel running was increased in ETOH EX compared to ETOH SED in the dark cycle. In the light cycle, ETOH mice ran less than CON mice, and EX mice ran less than SED mice despite all mice receiving chow diet and no EtOH. Overall, scheduled wheel running partially offset the alcohol-induced perturbations in the muscle and liver clock while ETOH and EX both influenced the timing of subsequent activity after the dietary intervention ended.
Collapse
Affiliation(s)
- Abigail L Tice
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida
| | - Choogon Lee
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida
| | - Robert C Hickner
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida
- Institute of Sports Sciences and Medicine, Florida State University, Tallahassee, Florida
| | - Jennifer L Steiner
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, Florida
- Institute of Sports Sciences and Medicine, Florida State University, Tallahassee, Florida
| |
Collapse
|
22
|
Chaikin CA, Thakkar AV, Steffeck AWT, Pfrender EM, Hung K, Zhu P, Waldeck NJ, Nozawa R, Song W, Futtner CR, Quattrocelli M, Bass J, Ben-Sahra I, Peek CB. Control of circadian muscle glucose metabolism through the BMAL1-HIF axis in obesity. Proc Natl Acad Sci U S A 2025; 122:e2424046122. [PMID: 40127275 PMCID: PMC12002348 DOI: 10.1073/pnas.2424046122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
Disruptions of circadian rhythms are widespread in modern society and lead to accelerated and worsened symptoms of metabolic syndrome. In healthy mice, the circadian clock factor BMAL1 is required for skeletal muscle function and metabolism. However, the importance of muscle BMAL1 in the development of metabolic diseases, such as diet-induced obesity (DIO), remains unclear. Here, we demonstrate that skeletal muscle-specific BMAL1-deficient mice exhibit worsened glucose tolerance upon high-fat diet feeding, despite no evidence of increased weight gain. Metabolite profiling from Bmal1-deficient muscles revealed impaired glucose utilization specifically at early steps in glycolysis that dictate the switch between anabolic and catabolic glucose fate. We provide evidence that this is due to abnormal control of the nutrient stress-responsive hypoxia-inducible factor (HIF) pathway. Genetic HIF1α stabilization in muscle Bmal1-deficient mice restores glucose tolerance and expression of 217/736 dysregulated genes during DIO, including glycolytic enzymes. Together, these data indicate that during DIO, skeletal muscle BMAL1 is an important regulator of HIF-driven glycolysis and metabolic flexibility, which influences the development of high-fat-diet-induced glucose intolerance.
Collapse
Affiliation(s)
- Claire A. Chaikin
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Abhishek V. Thakkar
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Adam W. T. Steffeck
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Eric M. Pfrender
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Kaitlyn Hung
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Pei Zhu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Nathan J. Waldeck
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Rino Nozawa
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Weimin Song
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Christopher R. Futtner
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Mattia Quattrocelli
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH45229
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Clara B. Peek
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| |
Collapse
|
23
|
Bai Z, Liang J, Nie Y, Wang S, Chang D. The mediating role of the TyG index in the relationship between circadian syndrome and cancer among middle-aged and elderly Chinese. BMC Cancer 2025; 25:431. [PMID: 40065285 PMCID: PMC11895363 DOI: 10.1186/s12885-025-13816-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Circadian Syndrome (CircS) is a significant marker of metabolic imbalance and has been linked to various chronic diseases. However, its relationship with cancer risk remains underexplored. This research aims to explore the relationship between CircS and cancer, while also assessing the possible mediating role of the triglyceride glucose (TyG) index. METHODS Baseline data from the 2011 China Health and Retirement Longitudinal Study (CHARLS) and follow-up data from 2015 were analyzed, including participants' sociodemographic characteristics, health behaviors, and metabolic indicators. Linear regression, mediation analysis, and logistic regression were employed to explore relationships between CircS, cancer risk, and the TyG index, with a dose-response analysis conducted on TyG index and cancer risk. RESULTS Among 7,864 middle-aged and elderly participants, CircS was significantly and positively associated with cancer risk (r = 0.17, P < 0.001). The TyG index showed a significant correlation with both CircS (r = 0.52, P < 0.001) and cancer (r = 0.15, P < 0.001). Mediation modeling indicated that the TyG index partially mediated the association between CircS and cancer, accounting for 23% of this relationship. Additionally, a significant nonlinear dose-response relationship was observed between the TyG index and cancer risk (Pnonlinear = 0.0024). CONCLUSION Circadian syndrome is associated with increased cancer risk, with the TyG index partially mediating this relationship.
Collapse
Affiliation(s)
- Zilong Bai
- Department of Surgical Oncology, The First Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710061, Shanxi, China
| | - Jiale Liang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710061, Shanxi, China
| | - Yuanhua Nie
- Department of Surgical Oncology, The First Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710061, Shanxi, China
| | - Shilong Wang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710061, Shanxi, China
| | - Dongmin Chang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710061, Shanxi, China.
| |
Collapse
|
24
|
Ragsdale SM, Radovich JM, Coiduras II, McCall WV, Grant SC, Lee C, Wilber A. Dual orexin receptor antagonists as promising therapeutics for Alzheimer's disease. NPJ BIOLOGICAL TIMING AND SLEEP 2025; 2:11. [PMID: 40066297 PMCID: PMC11890173 DOI: 10.1038/s44323-025-00025-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 01/23/2025] [Indexed: 03/15/2025]
Abstract
We examine the relationship between sleep, glymphatics and Alzheimer's disease (AD), and recent work questioning glymphatic clearance during sleep. We highlight a need for understanding glymphatic and/or other mechanism of clearance during sleep, and review glymphatic flow measurement methods. Further, we explore dual orexin receptor antagonists (DORAs) potential to mitigate AD sleep disturbances and enhance clearance. Further research could elucidate a linkage between DORAs, improved sleep and reducing AD pathophysiology.
Collapse
Affiliation(s)
- S. M. Ragsdale
- Department of Psychology; Program in Neuroscience; Florida State University, Tallahassee, FL USA
| | - J. M. Radovich
- Department of Chemical & Biochemical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL USA
- CIMAR, National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL USA
| | - I. I. Coiduras
- Department of Psychology; Program in Neuroscience; Florida State University, Tallahassee, FL USA
| | - W. V. McCall
- Department of Psychiatry and Health Behavior; Medical College of Georgia; Augusta University, Augusta, GA USA
| | - S. C. Grant
- Department of Chemical & Biochemical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL USA
- CIMAR, National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL USA
| | - C. Lee
- Department of Biomedical Sciences; Program in Neuroscience; College of Medicine, Florida State University, Tallahassee, FL USA
| | - A. Wilber
- Department of Psychology; Program in Neuroscience; Florida State University, Tallahassee, FL USA
| |
Collapse
|
25
|
Zhu P, Pfrender EM, Steffeck AWT, Reczek CR, Zhou Y, Thakkar AV, Gupta NR, Kupai A, Willbanks A, Lieber RL, Roy I, Chandel NS, Peek CB. Immunomodulatory role of the stem cell circadian clock in muscle repair. SCIENCE ADVANCES 2025; 11:eadq8538. [PMID: 40043110 PMCID: PMC11881903 DOI: 10.1126/sciadv.adq8538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 01/30/2025] [Indexed: 03/09/2025]
Abstract
Circadian rhythms orchestrate physiological processes such as metabolism, immune function, and tissue regeneration, aligning them with the optimal time of day (TOD). This study identifies an interplay between the circadian clock within muscle stem cells (SCs) and their capacity to modulate the immune microenvironment during muscle regeneration. We reveal that the SC clock triggers TOD-dependent inflammatory gene transcription after injury, particularly genes related to neutrophil activity and chemotaxis. These responses are driven by cytosolic regeneration of the signaling metabolite nicotinamide adenine dinucleotide (oxidized form) (NAD+), as enhancing cytosolic NAD+ regeneration in SCs is sufficient to induce inflammatory responses that influence muscle regeneration. Mononuclear single-cell sequencing of the regenerating muscle niche further implicates the cytokine CCL2 in mediating SC-neutrophil cross-talk in a TOD-dependent manner. Our findings highlight the intersection between SC metabolic shifts and immune responses within the muscle microenvironment, dictated by circadian rhythms, and underscore the potential for targeting circadian and metabolic pathways to enhance tissue regeneration.
Collapse
Affiliation(s)
- Pei Zhu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Eric M. Pfrender
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Adam W. T. Steffeck
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Colleen R. Reczek
- Department of Medicine, Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yalu Zhou
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Feinberg Cardiovascular and Renal Research Institute, Chicago, IL, USA
| | - Abhishek Vijay Thakkar
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Neha R. Gupta
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ariana Kupai
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Amber Willbanks
- Shirley Ryan AbilityLab (formerly known as Rehabilitation Institute of Chicago), Chicago, IL, USA
| | - Richard L. Lieber
- Shirley Ryan AbilityLab (formerly known as Rehabilitation Institute of Chicago), Chicago, IL, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, USA
- Hines VA Hospital, Maywood, IL, USA
| | - Ishan Roy
- Shirley Ryan AbilityLab (formerly known as Rehabilitation Institute of Chicago), Chicago, IL, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Navdeep S. Chandel
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Clara B. Peek
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
26
|
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
|
27
|
Deota S, Pendergast JS, Kolthur-Seetharam U, Esser KA, Gachon F, Asher G, Dibner C, Benitah SA, Escobar C, Muoio DM, Zhang EE, Hotamışlıgil GS, Bass J, Takahashi JS, Rabinowitz JD, Lamia KA, de Cabo R, Kajimura S, Longo VD, Xu Y, Lazar MA, Verdin E, Zierath JR, Auwerx J, Drucker DJ, Panda S. The time is now: accounting for time-of-day effects to improve reproducibility and translation of metabolism research. Nat Metab 2025; 7:454-468. [PMID: 40097742 DOI: 10.1038/s42255-025-01237-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
The constant expansion of the field of metabolic research has led to more nuanced and sophisticated understanding of the complex mechanisms that underlie metabolic functions and diseases. Collaborations with scientists of various fields such as neuroscience, immunology and drug discovery have further enhanced the ability to probe the role of metabolism in physiological processes. However, many behaviours, endocrine and biochemical processes, and the expression of genes, proteins and metabolites have daily ~24-h biological rhythms and thus peak only at specific times of the day. This daily variation can lead to incorrect interpretations, lack of reproducibility across laboratories and challenges in translating preclinical studies to humans. In this Review, we discuss the biological, environmental and experimental factors affecting circadian rhythms in rodents, which can in turn alter their metabolic pathways and the outcomes of experiments. We recommend that these variables be duly considered and suggest best practices for designing, analysing and reporting metabolic experiments in a circadian context.
Collapse
Affiliation(s)
- Shaunak Deota
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Ullas Kolthur-Seetharam
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
- Tata Institute of Fundamental Research, Hyderabad, India
| | - Karyn A Esser
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - Frédéric Gachon
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Gad Asher
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Charna Dibner
- Department of Surgery and Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute for Science and Technology, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Carolina Escobar
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Deborah M Muoio
- Departments of Medicine and Pharmacology & Cancer Biology, Duke Molecular Physiology Institute, Durham, NC, USA
| | | | - Gökhan S Hotamışlıgil
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Katja A Lamia
- Department of Molecular and Cellular Biology and Department of Molecular Medicine, the Scripps Research Institute, La Jolla, CA, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Shingo Kajimura
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, USA
| | - Valter D Longo
- Longevity Institute, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- AIRC Institute of Molecular Oncology, Italian Foundation for Cancer Research Institute of Molecular Oncology, Milan, Italy
| | - Ying Xu
- CAM-SU Genomic Resource Center, Soochow University, Suzhou, China
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity and Metabolism and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Daniel J Drucker
- The Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and the Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
28
|
Geng F, Zhao N, Ren Q. Circadian rhythm, microglia-mediated neuroinflammation, and Alzheimer's disease. Neurosci Biobehav Rev 2025; 170:106044. [PMID: 39914702 DOI: 10.1016/j.neubiorev.2025.106044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/16/2024] [Accepted: 02/03/2025] [Indexed: 02/09/2025]
Abstract
Microglia, the brain's resident macrophages, are key mediators of neuroinflammation, responding to immune pathogens and toxins. They play a crucial role in clearing cellular debris, regulating synaptic plasticity, and phagocytosing amyloid-β (Aβ) plaques in Alzheimer's disease (AD). Recent studies indicate that microglia not only exhibit intrinsic circadian rhythms but are also regulated by circadian clock genes, influencing specific functions such as phagocytosis and the modulation of neuroinflammation. Disruption of the circadian rhythm is closely associated with AD pathology. In this review, we will provide an overview of how circadian rhythms regulate microglia-mediated neuroinflammation in the progression of AD, focusing on the pathway from the central nervous system (CNS) and the peripheral immune system. We also discuss potential therapeutic targets, including hormone modulation, lifestyle interventions, and anti-inflammatory therapies, aimed at maintaining brain health in AD. This will shed light on the involvement of circadian rhythm in AD and explore new avenues for AD treatment.
Collapse
Affiliation(s)
- Fan Geng
- Department of Neurology, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing 210009, China
| | - Na Zhao
- Department of Neurology, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing 210009, China
| | - Qingguo Ren
- Department of Neurology, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
29
|
Stevenson EL, Mehalow AK, Loros JJ, Kelliher CM, Dunlap JC. A Compensated Clock: Temperature and Nutritional Compensation Mechanisms Across Circadian Systems. Bioessays 2025; 47:e202400211. [PMID: 39696884 DOI: 10.1002/bies.202400211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/07/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024]
Abstract
Circadian rhythms are ∼24-h biological oscillations that enable organisms to anticipate daily environmental cycles, so that they may designate appropriate day/night functions that align with these changes. The molecular clock in animals and fungi consists of a transcription-translation feedback loop, the plant clock is comprised of multiple interlocking feedback-loops, and the cyanobacterial clock is driven by a phosphorylation cycle involving three main proteins. Despite the divergent core clock mechanisms across these systems, all circadian clocks are able to buffer period length against changes in the ambient growth environment, such as temperature and nutrients. This defining capability, termed compensation, is critical to proper timekeeping, yet the underlying mechanism(s) remain elusive. Here we examine the known players in, and the current models for, compensation across five circadian systems. While compensation models across these systems are not yet unified, common themes exist across them, including regulation via temperature-dependent changes in post-translational modifications.
Collapse
Affiliation(s)
- Elizabeth-Lauren Stevenson
- Department of Molecular & Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Adrienne K Mehalow
- Department of Molecular & Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Jennifer J Loros
- Department of Biochemistry & Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Christina M Kelliher
- Department of Molecular & Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
- Department of Biology, University of Massachusetts Boston, Boston, Massachusetts, USA
| | - Jay C Dunlap
- Department of Molecular & Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
30
|
Bou Serhal J, Fayyad-Kazan M, Kabrita CS. Understanding the mechanistic interlink between circadian misalignment and heart disease in night shift workers: Therapeutic role of behavioral interventions. Sleep Breath 2025; 29:109. [PMID: 39964617 DOI: 10.1007/s11325-025-03260-z] [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: 12/13/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND Rotating and night shift work, especially in older workers, is a growing health concern of modern societies due to the associated high morbidity and mortality rates from cardiovascular disease (CVD). The resulting circadian misalignment disrupts neuroendocrine pathways that regulate cardiovascular physiology, risking myocardial tissue damage and heart dysfunction. AIMS Considering the gaps in the literature as to how atypical work behaviors may disrupt the temporal link between the central and myocardial oscillators at the level of the proteome and transcriptome, the primary goal of this review is to assess the molecular mechanisms linking disrupted biological rhythms to heart health, with a focus on core clock genes like BMAL1 and cardiac troponin I (cTnI) as a myocardial biomarker. MAJOR FINDINGS Circadian misalignment can lead to cognitive decline, metabolic dysfunction, and immune disruption, all of which elevate CVD risk. BMAL1 has a key role in maintaining cardiovascular integrity, with its dysfunction associated with hypertension, arrhythmias, and myocardial injury. Additionally, disrupted sleep patterns influence the expression of clock genes, potentially leading to altered heart function and elevated levels of cardiac biomarkers like troponin. CONCLUSION Circadian misalignment poses significant CVD risks, particularly for older workers. Future research should investigate how the expression of central and peripheral clock genes, as well as cardiac biomarkers is affected by shift work, especially in older individuals. Behavioral interventions such as chronotherapy, light therapy, and scheduled evening sleep may help mitigate these risks, but more studies are needed to assess their long-term effectiveness.
Collapse
Affiliation(s)
- Jad Bou Serhal
- Faculty of Arts and Sciences, Department of Psychology, American University of Beirut (AUB), Beirut, Lebanon
| | - Mohammad Fayyad-Kazan
- College of Arts and Sciences, Department of Natural and Applied Sciences, American University of Iraq-Baghdad (AUIB), Baghdad, Iraq
| | - Colette S Kabrita
- College of Arts and Sciences, Department of Natural and Applied Sciences, American University of Iraq-Baghdad (AUIB), Baghdad, Iraq.
- Faculty of Natural and Applied Sciences, Department of Sciences, Notre Dame University-Louaize (NDU), P.O. Box 72 Zouk Mikael, Zouk Mosbeh, Lebanon.
| |
Collapse
|
31
|
Jaschke NP, Wang A. Integrated control of leukocyte compartments as a feature of adaptive physiology. Immunity 2025; 58:279-294. [PMID: 39909034 DOI: 10.1016/j.immuni.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 12/10/2024] [Accepted: 01/16/2025] [Indexed: 02/07/2025]
Abstract
As a highly diverse and mobile organ, the immune system is uniquely equipped to participate in tissue responses in a tunable manner, depending on the number, type, and nature of cells deployed to the respective organ. Most acute organismal stressors that threaten survival-predation, infection, poisoning, and others-induce pronounced redistribution of immune cells across tissue compartments. Here, we review the current understanding of leukocyte compartmentalization under homeostatic and noxious conditions. We argue that leukocyte shuttling between compartments is a function of local tissue demands, which are linked to the organ's contribution to adaptive physiology at steady state and upon challenge. We highlight the neuroendocrine signals that relay and organize this trafficking behavior and outline mechanisms underlying the functional diversification of leukocyte responses. In this context, we discuss important areas of future inquiry and the implications of this scientific space for clinical medicine in the era of targeted immunomodulation.
Collapse
Affiliation(s)
- Nikolai P Jaschke
- Department of Internal Medicine (Rheumatology, Allergy & Immunology) and Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
| | - Andrew Wang
- Department of Internal Medicine (Rheumatology, Allergy & Immunology) and Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
32
|
Le Collen L, Froguel P, Bonnefond A. Towards the recognition of oligogenic forms of type 2 diabetes. Trends Endocrinol Metab 2025; 36:109-117. [PMID: 38955653 DOI: 10.1016/j.tem.2024.06.006] [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: 04/14/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 07/04/2024]
Abstract
The demarcation between monogenic and polygenic type 2 diabetes (T2D) is less distinct than previously believed. Notably, recent research has highlighted a new entity, that we suggest calling oligogenic forms of T2D, serving as a genetic link between these two forms. In this opinion article, we have reviewed scientific advances that suggest categorizing genes involved in oligogenic T2D. Research focused on polygenic T2D has faced challenges in deepening our comprehension of the pathophysiology of T2D due to the inability to directly establish causal links between a signal and the molecular mechanisms underlying the disease. However, the study of oligogenic forms of T2D has illuminated distinct causal connections between genes and disease risk, thereby indicating potential new drug targets.
Collapse
Affiliation(s)
- Lauriane Le Collen
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, Lille University Hospital, Lille, France; University of Lille, Lille, France; Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, Nutrition, and Metabolism, University Hospital of Nancy, Nancy, France
| | - Philippe Froguel
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, Lille University Hospital, Lille, France; University of Lille, Lille, France; Department of Metabolism, Imperial College London, Hammersmith Hospital, London, UK
| | - Amélie Bonnefond
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, Lille University Hospital, Lille, France; University of Lille, Lille, France; Department of Metabolism, Imperial College London, Hammersmith Hospital, London, UK.
| |
Collapse
|
33
|
Miller MA. Time for bed: diet, sleep and obesity in children and adults. Proc Nutr Soc 2025; 84:45-52. [PMID: 38012858 DOI: 10.1017/s0029665123004846] [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: 11/29/2023]
Abstract
Sufficient sleep is necessary for optimal health, daytime performance and wellbeing and the amount required is age-dependent and decreases across the lifespan. Sleep duration is usually affected by age and several different cultural, social, psychological, behavioural, pathophysiological and environmental factors. This review considers how much sleep children and adults need, why this is important, what the consequences are of insufficient sleep and how we can improve sleep. A lack of the recommended amount of sleep for a given age group has been shown to be associated with detrimental effects on health including effects on metabolism, endocrine function, immune function and haemostatic pathways. Obesity has increased worldwide in the last few decades and the WHO has now declared it a global epidemic. A lack of sleep is associated with an increased risk of obesity in children and adults, which may lead to future poor health outcomes. Data from studies in both children and adults suggest that the relationship between sleep and obesity may be mediated by several different mechanisms including alterations in appetite and satiety, sleep timing, circadian rhythm and energy balance. Moreover, there is evidence to suggest that improvements in sleep, in both children and adults, can be beneficial for weight management and diet and certain foods might be important to promote sleep. In conclusion this review demonstrates that there is a wide body of evidence to suggest that sleep and obesity are causally related and recommends that further research is required to inform policy, and societal change.
Collapse
|
34
|
Kim H, Lee KH, Shin J, Seo WW, Jeon JE, Lee HY, You JH, Kim SY, Kim SJ, Lee YJ. High sleep reactivity in shift workers is associated with increased sleep disturbance, mood problems, and reduced quality of life. Sleep Med 2025; 126:275-281. [PMID: 39733666 DOI: 10.1016/j.sleep.2024.12.027] [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: 09/07/2024] [Revised: 11/28/2024] [Accepted: 12/18/2024] [Indexed: 12/31/2024]
Abstract
INTRODUCTION Shift work disrupts circadian rhythms, causing sleep and mood problems. Sleep reactivity-the sensitivity of sleep to stress-may affect how shift workers cope with these disruptions. This study investigated the relationship between sleep reactivity and shift work, exploring associations between sleep reactivity and sleep disturbance, mood symptoms, and quality of life in shift workers. METHODS In a cross-sectional design, 132 participants (79 shift workers and 53 controls) were assessed using the Ford Insomnia Response to Stress Test (FIRST), Insomnia Severity Index (ISI), Pittsburgh Sleep Quality Index (PSQI), Epworth Sleepiness Scale (ESS), Beck Depression Inventory (BDI), Beck Anxiety Inventory (BAI), and World Health Organization Quality of Life (WHOQOL). We compared the self-reported measurements between shift workers and controls. Two-way ANOVA was performed to explore the interaction effects between shift work and sleep reactivity on sleep, mood parameters, and quality of life. Multiple linear regression analysis was conducted to identify factors associated with sleep, mood, and quality of life among shift workers. RESULTS Shift workers scored higher on ISI and BDI compared to controls. Two-way ANOVA revealed an interaction effect between shift work and sleep reactivity on WHOQOL. Regression analysis indicated that high sleep reactivity was associated with higher ISI, BDI, BAI, and lower WHOQOL among shift workers. CONCLUSION Sleep reactivity significantly affected shift worker's quality of life. Our findings indicate that high sleep reactivity in shift workers was associated with increased sleep disturbance, mood problems, and decreased quality of life, implying that sleep reactivity may predict shift work tolerance.
Collapse
Affiliation(s)
- Hanseul Kim
- Department of Psychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyung Hwa Lee
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jiyoon Shin
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Seoul National University Hospital, Seoul, Republic of Korea; Department of Psychiatry, Uijeongbu Eulji Medical Center, Eulji University, Uijeongbu, Republic of Korea; Department of Psychiatry and Center for Sleep and Chronobiology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Won Woo Seo
- Yonsei Soul Mental Health Clinic, Suwon, Republic of Korea; Department of Psychiatry and Center for Sleep and Chronobiology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jeong Eun Jeon
- Department of Psychiatry and Center for Sleep and Chronobiology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ha Young Lee
- Department of Psychiatry and Center for Sleep and Chronobiology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jin Hyeok You
- Department of Psychiatry and Center for Sleep and Chronobiology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sun-Young Kim
- Department of Psychiatry, Ewha Womans University Hospital, Ewha Womans University College of Medicine, Ewha Womans University Seoul Hospital, Seoul, Republic of Korea
| | - Seog Ju Kim
- Department of Psychiatry, Samsung Medical Center, Seoul, Republic of Korea.
| | - Yu Jin Lee
- Department of Psychiatry and Center for Sleep and Chronobiology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
35
|
Larrondo LF. Circadian rhythms: pervasive, and often times evasive. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230477. [PMID: 39842475 DOI: 10.1098/rstb.2023.0477] [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: 04/23/2024] [Revised: 08/23/2024] [Accepted: 08/30/2024] [Indexed: 01/24/2025] Open
Abstract
Most circadian texts begin by stating that clocks are pervasive throughout the tree of life. Indeed, clock mechanisms have been described from cyanobacteria to humans, representing a notable example of convergent evolution: yet, there are several phyla in animals, protists or within fungi and bacteria, in which homologs of some-or all-known clock components seem to be absent, posing inevitable questions about the evolution of circadian systems. Moreover, as we move away from model organisms, there are several taxa in which core clock elements can be identified at the genomic levels. However, the functional description of those putative clocks has been hard to achieve, as rhythmicity is not observed unless defined abiotic or nutritional cues are provided. The mechanisms 'conditioning' the functionality of clocks remain uncertain, emphasizing the need to delve further into non-model circadian systems. As the absence of evidence is not evidence of absence, the lack of known core-clock homologs or of observable rhythms in a given organism, cannot be an a priori criterion to discard the presence of a functional clock, as rhythmicity may be limited to yet untested experimental conditions or phenotypes. This article seeks to reflect on these topics, highlighting some of the pressing questions awaiting to be addressed.This article is part of the Theo Murphy meeting issue 'Circadian rhythms in infection and immunity'.
Collapse
Affiliation(s)
- Luis F Larrondo
- ANID-Millennium Science Initiative Program-Millennium Institute for Integrative Biology (iBio), Santiago 8331150, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| |
Collapse
|
36
|
Castro-Mata PC, Cueto-Manzano AM, Vizmanos B, González-Ortiz A, Betancourt-Núñez A, Martín-Del-Campo F. Chrononutrition in Chronic Kidney Disease. Nutrients 2025; 17:389. [PMID: 39940247 PMCID: PMC11820925 DOI: 10.3390/nu17030389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 02/14/2025] Open
Abstract
Chrononutrition, the study of the interaction between biological rhythms and nutrition, has emerged as a promising field for addressing metabolic health. However, its role in chronic kidney disease (CKD) remains underexplored. CKD patients often experience circadian disruptions due to renal, metabolic, treatment-related, and lifestyle factors, which may influence their nutritional status and clinical outcomes. Objective: to synthesize and analyze the existing evidence on chrononutrition in CKD patients, identify knowledge gaps, and propose directions for future research across different stages of CKD. Initially, this review contextualizes circadian physiology, alignment, and chronodisruption to explore such factors in CKD patients, focusing on chrononutrition variables already studied in the general population. We discuss how dietary timing and habit adjustments could influence CKD clinical outcomes, offering insights into circadian impacts on disease management. This new approach could optimize patient care, encouraging further research, particularly in the development of personalized strategies for different stages of the disease.
Collapse
Affiliation(s)
- Pilar C Castro-Mata
- Medical Research Unit of Renal Diseases, Specialties Hospital, Western National Medical Center, Mexican Institute of Social Security (IMSS), Guadalajara 44320, Mexico
- PhD Program in Translational Nutrition Sciences, Department of Human Reproduction, Child Growth and Development, University Center of Health Sciences (CUCS), Guadalajara 44340, Mexico
| | - Alfonso M Cueto-Manzano
- Medical Research Unit of Renal Diseases, Specialties Hospital, Western National Medical Center, Mexican Institute of Social Security (IMSS), Guadalajara 44320, Mexico
| | - Barbara Vizmanos
- PhD Program in Translational Nutrition Sciences, Department of Human Reproduction, Child Growth and Development, University Center of Health Sciences (CUCS), Guadalajara 44340, Mexico
| | - Ailema González-Ortiz
- Translational Research Center, National Institute of Pediatrics, Mexico City 04530, Mexico
| | - Alejandra Betancourt-Núñez
- PhD Program in Translational Nutrition Sciences, Department of Human Reproduction, Child Growth and Development, University Center of Health Sciences (CUCS), Guadalajara 44340, Mexico
| | - Fabiola Martín-Del-Campo
- Medical Research Unit of Renal Diseases, Specialties Hospital, Western National Medical Center, Mexican Institute of Social Security (IMSS), Guadalajara 44320, Mexico
- PhD Program in Translational Nutrition Sciences, Department of Human Reproduction, Child Growth and Development, University Center of Health Sciences (CUCS), Guadalajara 44340, Mexico
| |
Collapse
|
37
|
Yang MY, Lin HYH, Chen YYM, Hu ML, Chen IY, Yang CH. Chronic low-dose REV-ERBs agonist SR9009 mitigates constant light-induced weight gain and insulin resistance via adipogenesis modulation. Biomed J 2025:100830. [PMID: 39800061 DOI: 10.1016/j.bj.2025.100830] [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: 11/08/2024] [Revised: 12/24/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Obesity and circadian rhythm disruption are significant global health concerns, contributing to an increased risk of metabolic disorders. Both adipose tissue and circadian rhythms play critical roles in maintaining energy homeostasis, and their dysfunction is closely linked to obesity. This study aimed to assess the effects of chronic low-dose SR9009, a REV-ERB ligand, on circadian disruption induced by constant light exposure in mice. MATERIAL AND METHODS Mice were exposed to constant light for eight weeks (LL mice), resulting in increased body weight, insulin resistance, white fat mass, and altered circadian clock gene expression. Low-dose SR9009 (10 mg/kg daily) was administered chronically to assess its impact on these metabolic disruptions. RESULTS LL mice treated with SR9009 for eight weeks showed reduced weight gain, insulin resistance, and white fat mass but no significant impact on overall energy homeostasis. SR9009 suppressed Bmal1 expression and restored Rev-erbα and Rev-erbβ expression in white and brown adipose tissue (WAT and BAT). In vitro studies using 3T3-L1 cells indicated that SR9009 inhibited adipogenesis, leading to further investigation in vivo. SR9009 restored ChREBP1a and Srebp-1c expression in BAT but did not affect inflammatory cytokine or adipokine gene expression, nor did it restore Fasn, Pparγ, and Prom1 expression in both WAT and BAT. CONCLUSIONS These findings suggest that SR9009 may be a potential therapeutic approach for preventing weight gain and insulin resistance caused by circadian disruptions, likely through adipogenesis inhibition, though its effects on other metabolic pathways remain limited at low doses.
Collapse
Affiliation(s)
- Ming-Yu Yang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hugo Y-H Lin
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Ywan M Chen
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Ming-Luen Hu
- Division of Hepatogestroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - I-Ya Chen
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chao-Hui Yang
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan; School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
38
|
Matak AM, Mu Y, Mohati SM, Makdissi S, Di Cara F. Circadian rhythm and immunity: decoding chrono-immunology using the model organism Drosophila melanogaster. Genome 2025; 68:1-18. [PMID: 40168693 DOI: 10.1139/gen-2025-0003] [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: 04/03/2025]
Abstract
Circadian rhythms are important cellular pathways first described for their essential role in helping organisms adjust to the 24 h day-night cycle and synchronize physiological and behavioral functions. Most organisms have evolved a circadian central clock to anticipate daily environmental changes in light, temperature, and mate availability. It is now understood that multiple clocks exist in organisms to regulate the functions of specific organs. Epidemiological studies in humans reported that disruption of the circadian rhythms caused by sleep deprivation is linked to the onset of immune-related conditions, suggesting the importance of circadian regulation of immunity. Mechanistic studies to define how circadian clocks and immune responses interact have profound implications for human health. However, elucidating the clocks and their tissue-specific functions has been challenging in mammals. Many studies using simple model organisms such as Drosophila melanogaster have been pioneering in discovering that the clock controls innate immune responses and immune challenges can impact circadian rhythms and/or their outcomes. In this review, we will report genetic studies using the humble fruit fly that identified the existence of reciprocal interactions between the circadian pathway and innate immune signaling, contributing to elucidate mechanisms in the growing field of chrono-immunology.
Collapse
Affiliation(s)
- Arash Mohammadi Matak
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3K 6R8, Canada
| | - Yizhu Mu
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3K 6R8, Canada
| | - Seyedeh Mahdiye Mohati
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3K 6R8, Canada
| | - Stephanie Makdissi
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3K 6R8, Canada
| | - Francesca Di Cara
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3K 6R8, Canada
| |
Collapse
|
39
|
Zhu X, Zhang Z, Zhang J, Xiao Y, Wang H, Wang M, Jiang M, Xu Y. Single-cell and Bulk Transcriptomic Analyses Reveal a Stemness and Circadian Rhythm Disturbance-related Signature Predicting Clinical Outcome and Immunotherapy Response in Hepatocellular Carcinoma. Curr Gene Ther 2025; 25:178-193. [PMID: 38847249 DOI: 10.2174/0115665232298240240529131358] [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: 01/22/2024] [Revised: 04/30/2024] [Accepted: 05/19/2024] [Indexed: 11/30/2024]
Abstract
AIMS Investigating the impact of stemness-related circadian rhythm disruption (SCRD) on hepatocellular carcinoma (HCC) prognosis and its potential as a predictor for immunotherapy response. BACKGROUND Circadian disruption has been linked to tumor progression through its effect on the stemness of cancer cells. OBJECTIVE Develop a novel signature for SCRD to accurately predict clinical outcomes and immune therapy response in patients with HCC. METHODS The stemness degree of patients with HCC was assessed based on the stemness index (mRNAsi). The co-expression circadian genes significantly correlated with mRNAsi were identified and defined as stemness- and circadian-related genes (SCRGs). The SCRD scores of samples and cells were calculated based on the SCRGs. Differentially expressed genes with a prognostic value between distinct SCRD groups were identified in bulk and single-cell datasets to develop an SCRD signature. RESULTS A higher SCRD score indicates a worse patient survival rate. Analysis of the tumor microenvironment revealed a significant correlation between SCRD and infiltrating immune cells. Heterogeneous expression patterns, functional states, genomic variants, and cell-cell interactions between two SCRD populations were revealed by transcriptomic, genomic, and interaction analyses. The robust SCRD signature for predicting immunotherapy response and prognosis in patients with HCC was developed and validated in multiple independent cohorts. CONCLUSIONS In summary, distinct tumor immune microenvironment patterns were confirmed under SCRD in bulk and single-cell transcriptomic, and SCRD signature associated with clinical outcomes and immunotherapy response was developed and validated in HCC.
Collapse
Affiliation(s)
- Xiaojing Zhu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zixin Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jiaxing Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yanqi Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hao Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Mingwei Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Minghui Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
40
|
Soliz-Rueda JR, Cuesta-Marti C, O'Mahony SM, Clarke G, Schellekens H, Muguerza B. Gut microbiota and eating behaviour in circadian syndrome. Trends Endocrinol Metab 2025; 36:15-28. [PMID: 39095231 DOI: 10.1016/j.tem.2024.07.008] [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: 05/20/2024] [Revised: 07/04/2024] [Accepted: 07/12/2024] [Indexed: 08/04/2024]
Abstract
Eating behaviour and circadian rhythms are closely related. The type, timing, and quantity of food consumed, and host circadian rhythms, directly influence the intestinal microbiota, which in turn impacts host circadian rhythms and regulates food intake beyond homeostatic eating. This Opinion discusses the impact of food intake and circadian disruptions induced by an obesogenic environment on gut-brain axis signalling. We also explore potential mechanisms underlying the effects of altered gut microbiota on food intake behaviour and circadian rhythmicity. Understanding the crosstalk between gut microbiota, circadian rhythms, and unhealthy eating behaviour is crucial to addressing the obesity epidemic, which remains one of the biggest societal challenges of our time.
Collapse
Affiliation(s)
- Jorge R Soliz-Rueda
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira I Virgili, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain; Center of Environmental, Food and Toxicological Technology (TecnATox), Tarragona, Spain
| | - Cristina Cuesta-Marti
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, Cork, Ireland
| | - Siobhain M O'Mahony
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Harriët Schellekens
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, Cork, Ireland.
| | - Begoña Muguerza
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira I Virgili, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain; Center of Environmental, Food and Toxicological Technology (TecnATox), Tarragona, Spain
| |
Collapse
|
41
|
Gentile F, Emdin M, Passino C, Montuoro S, Tognini P, Floras JS, O'Neill J, Giannoni A. The chronobiology of human heart failure: clinical implications and therapeutic opportunities. Heart Fail Rev 2025; 30:103-116. [PMID: 39392534 DOI: 10.1007/s10741-024-10447-1] [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] [Accepted: 09/29/2024] [Indexed: 10/12/2024]
Abstract
Circadian variation in cardiovascular and metabolic dynamics arises from interactions between intrinsic rhythms and extrinsic cues. By anticipating and accommodating adaptation to awakening and activity, their synthesis maintains homeostasis and maximizes efficiency, flexibility, and resilience. The dyssynchrony of cardiovascular load and energetic capacity arising from attenuation or loss of such rhythms is strongly associated with incident heart failure (HF). Once established, molecular, neurohormonal, and metabolic rhythms are frequently misaligned with each other and with extrinsic cycles, contributing to HF progression and adverse outcomes. Realignment of biological rhythms via lifestyle interventions, chronotherapy, and time-tailored autonomic modulation represents an appealing potential strategy for improving HF-related morbidity and mortality.
Collapse
Affiliation(s)
- Francesco Gentile
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy
- Division of Cardiology and Cardiovascular Medicine, Fondazione Monasterio, Pisa, Italy
| | - Michele Emdin
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy
- Division of Cardiology and Cardiovascular Medicine, Fondazione Monasterio, Pisa, Italy
| | - Claudio Passino
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy
- Division of Cardiology and Cardiovascular Medicine, Fondazione Monasterio, Pisa, Italy
| | - Sabrina Montuoro
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Paola Tognini
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy
| | - John S Floras
- University Health Network and Sinai Health Division of Cardiology, Toronto, ON, Canada
| | - John O'Neill
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Alberto Giannoni
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy.
- Division of Cardiology and Cardiovascular Medicine, Fondazione Monasterio, Pisa, Italy.
| |
Collapse
|
42
|
Wang Y, Zhao X, Ren J, Xue S, Liu Y, Zhang L, Fan M, Su C, Cheng JX. The effect of light therapy on insomnia: A systematic review and meta-analysis. Sleep Breath 2024; 29:66. [PMID: 39733392 DOI: 10.1007/s11325-024-03204-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/20/2024] [Accepted: 09/23/2024] [Indexed: 12/31/2024]
Abstract
PURPOSE Light is a crucial factor influencing sleep arousal patterns. This meta-analysis investigates the efficacy of light therapy (LT) for insomnia treatment. METHODS Five electronic databases were independently searched by two reviewers until August 2024. The literature screening focused specifically on populations with insomnia complaints treated by LT. Weighted Mean Difference (WMD) and 95% Confidence Interval (CI) were used as statistical tools, while the Cochrane Risk of Bias tool version 2 (RoB 2) was employed to assess the quality of evidence. A total of 10 randomized controlled trials (RCTs) were analyzed. RESULTS The results demonstrated that LT showed statistically significant improvements in subjective sleep quality, as evidenced by a reduction in Pittsburgh sleep quality scale (PSQI) scores by -2.89 (95% CI = -4.80 to -0.97) and Insomnia severity index (ISI) scores by -2.16 (95% CI = -4.23 to -0.08) post-intervention. Additionally, actigraphy revealed a statistically significant increase in total sleep time (TST) by 16.78 min (95% CI = 0.67 to 32.89) and a decrease in wake after sleep onset (WASO) by -12.91 min (95% CI = -25.62 to -0.20) by LT. CONCLUSION The preliminary results of the study suggest that LT has some efficacy in improving sleep quality in insomnia. However, it needs to be validated in future clinical trials with larger samples.
Collapse
Affiliation(s)
- Yi Wang
- Sleep Medicine Center, Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, China
| | - Xianchao Zhao
- Sleep Medicine Center, Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, China
| | - Jiafeng Ren
- Sleep Medicine Center, Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, China
| | - Shengwen Xue
- Sleep Medicine Center, Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, China
| | - Yuhang Liu
- Sleep Medicine Center, Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, China
| | - Liping Zhang
- Sleep Medicine Center, Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, China
| | - Mengmeng Fan
- Sleep Medicine Center, Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, China
| | - Changjun Su
- Sleep Medicine Center, Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, China.
| | - Jin-Xiang Cheng
- Sleep Medicine Center, Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, China.
| |
Collapse
|
43
|
Zeng G, Liu X, Zheng Z, Zhao J, Zhuo W, Bai Z, Lin E, Cai S, Cai C, Li P, Zou B, Li J. Knockdown of RASD1 improves MASLD progression by inhibiting the PI3K/AKT/mTOR pathway. Lipids Health Dis 2024; 23:424. [PMID: 39731125 DOI: 10.1186/s12944-024-02419-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/22/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND There is still no reliable therapeutic targets and effective pharmacotherapy for metabolic dysfunction-associated steatotic liver disease (MASLD). RASD1 is short for Ras-related dexamethasone-induced 1, a pivotal factor in various metabolism processes of Human. However, the role of RASD1 remains poorly illustrated in MASLD. Therefore, we designed a study to elucidate how RASD1 could impact on MASLD as well as the mechanisms involved. METHODS The expression level of RASD1 was validated in MASLD. Lipid metabolism and its underlying mechanism were investigated in hepatocytes and mice with either overexpression or knockdown of RASD1. RESULTS Hepatic RASD1 expression was upregulated in MASLD. Lipid deposition was significantly reduced in RASD1-knockdown hepatocytes and mice, accompanied by a marked downregulation of key genes in the signaling pathway of de novo lipogenesis. Conversely, RASD1 overexpression in hepatocytes had the opposite effect. Mechanistically, RASD1 regulated lipid metabolism in MASLD through the PI3K/AKT/mTOR signaling pathway. CONCLUSIONS We discovered a novel role of RASD1 in MASLD by regulating lipogenesis via the PI3K/AKT/mTOR pathway, thereby identifying a potential treatment target for MASLD.
Collapse
Affiliation(s)
- Guifang Zeng
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China.
| | - Xialei Liu
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Zhouying Zheng
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Jiali Zhao
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Wenfeng Zhuo
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Zirui Bai
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - En Lin
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Shanglin Cai
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Chaonong Cai
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Peiping Li
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China.
| | - Baojia Zou
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China.
| | - Jian Li
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China.
| |
Collapse
|
44
|
Hussain Y, Dar MI, Pan X. Circadian Influences on Brain Lipid Metabolism and Neurodegenerative Diseases. Metabolites 2024; 14:723. [PMID: 39728504 DOI: 10.3390/metabo14120723] [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: 11/18/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
Circadian rhythms are intrinsic, 24 h cycles that regulate key physiological, mental, and behavioral processes, including sleep-wake cycles, hormone secretion, and metabolism. These rhythms are controlled by the brain's suprachiasmatic nucleus, which synchronizes with environmental signals, such as light and temperature, and consequently maintains alignment with the day-night cycle. Molecular feedback loops, driven by core circadian "clock genes", such as Clock, Bmal1, Per, and Cry, are essential for rhythmic gene expression; disruptions in these feedback loops are associated with various health issues. Dysregulated lipid metabolism in the brain has been implicated in the pathogenesis of neurological disorders by contributing to oxidative stress, neuroinflammation, and synaptic dysfunction, as observed in conditions such as Alzheimer's and Parkinson's diseases. Disruptions in circadian gene expression have been shown to perturb lipid regulatory mechanisms in the brain, thereby triggering neuroinflammatory responses and oxidative damage. This review synthesizes current insights into the interconnections between circadian rhythms and lipid metabolism, with a focus on their roles in neurological health and disease. It further examines how the desynchronization of circadian genes affects lipid metabolism and explores the potential mechanisms through which disrupted circadian signaling might contribute to the pathophysiology of neurodegenerative disorders.
Collapse
Affiliation(s)
- Yusuf Hussain
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Langone Hospital-Long Island, Mineola, NY 11501, USA
| | - Mohammad Irfan Dar
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Langone Hospital-Long Island, Mineola, NY 11501, USA
| | - Xiaoyue Pan
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Langone Hospital-Long Island, Mineola, NY 11501, USA
| |
Collapse
|
45
|
Young MJ, Heanue S, Kanki M, Moneghetti KJ. Circadian disruption and its impact on the cardiovascular system. Trends Endocrinol Metab 2024:S1043-2760(24)00316-3. [PMID: 39706759 DOI: 10.1016/j.tem.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/11/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
Circadian rhythms are highly conserved biorhythms of ~24 h that govern many fundamental biological processes, including cardiovascular (CV) homeostasis. Disrupting the timing of cellular oscillators promotes cellular stress, and induction of pathogenic pathways underpins the pathogenesis of many CV diseases (CVDs). Thus, shift work, late eating, sleep disturbances, and other disruptors can result in an elevated risk of heart disease and increased incidence of adverse CV events. Here, we discuss the importance of circadian rhythms for CV homeostasis, recent developments in understanding the impact of disrupted circadian rhythms on CV health and disease progression, and how understanding the interactions between circadian and CV physiology is crucial for improving interventions to mitigate CVD, especially in populations impacted by disrupted circadian rhythms.
Collapse
Affiliation(s)
- Morag J Young
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia.
| | - Seamus Heanue
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Medicine, Central Clinical School, Monash University, Clayton, VIC, Australia
| | - Monica Kanki
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Kegan J Moneghetti
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
46
|
Li T, Yan Q, Nan J, Huang X, Wang R, Zhang Y, Zhao X, Wang Q. LncRNA-MSTRG.19083.1 Targets NTRK2 as a miR-429-y Sponge to Regulate Circadian Rhythm via the cAMP Pathway in Yak Testis and Cryptorchidism. Int J Mol Sci 2024; 25:13553. [PMID: 39769319 PMCID: PMC11678581 DOI: 10.3390/ijms252413553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Long noncoding RNAs (LncRNAs) play essential roles in numerous biological processes in mammals, such as reproductive physiology and endocrinology. Cryptorchidism is a common male reproductive disease. Circadian rhythms are actively expressed in the reproductive system. In this study, a total of 191 LncRNAs were obtained from yak testes and cryptorchids. Then, we identified NTRK2's relationship to circadian rhythm and behavioral processes. Meanwhile, the ceRNA (LncRNA-MSTRG.19083.1/miR-429-y/NTRK2) network was constructed, and its influence on circadian rhythm was revealed. The results showed that NTRK2 and LncRNA-MSTRG.19083.1 were significantly upregulated, and miR-429-y was obviously decreased in cryptorchid tissue; NTRK2 protein was mainly distributed in the Leydig cells of the testis. In addition, the upregulation of the expression level of miR-429-y resulted in the significant downregulation of LncRNA and NTRK2 levels, while the mRNA and protein levels of CREB, CLOCK, and BMAL1 were significantly upregulated; the knockdown of miR-429-y resulted in the opposite changes. Our findings suggested that LncRNA-MSTRG.19083.1 competitively binds to miR-429-y to target NTRK2 to regulate circadian rhythm through the cAMP pathway. Taken together, the results of our study provide a comprehensive understanding of how the LncRNA-miRNA-mRNA networks operate when yak cryptorchidism occurs. Knowledge of circadian-rhythm-associated mRNAs and LncRNAs could be useful for better understanding the relationship between circadian rhythm and reproduction.
Collapse
Affiliation(s)
- Tianan Li
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China; (T.L.); (Q.Y.); (J.N.); (X.H.); (R.W.); (Y.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Qiu Yan
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China; (T.L.); (Q.Y.); (J.N.); (X.H.); (R.W.); (Y.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Jinghong Nan
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China; (T.L.); (Q.Y.); (J.N.); (X.H.); (R.W.); (Y.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Xue Huang
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China; (T.L.); (Q.Y.); (J.N.); (X.H.); (R.W.); (Y.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Ruiqing Wang
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China; (T.L.); (Q.Y.); (J.N.); (X.H.); (R.W.); (Y.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China; (T.L.); (Q.Y.); (J.N.); (X.H.); (R.W.); (Y.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Xingxu Zhao
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China; (T.L.); (Q.Y.); (J.N.); (X.H.); (R.W.); (Y.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Qi Wang
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China; (T.L.); (Q.Y.); (J.N.); (X.H.); (R.W.); (Y.Z.)
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| |
Collapse
|
47
|
El-Tanani M, Rabbani SA, Ali AA, Alfaouri IGA, Al Nsairat H, Al-Ani IH, Aljabali AA, Rizzo M, Patoulias D, Khan MA, Parvez S, El-Tanani Y. Circadian rhythms and cancer: implications for timing in therapy. Discov Oncol 2024; 15:767. [PMID: 39692981 PMCID: PMC11655929 DOI: 10.1007/s12672-024-01643-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/27/2024] [Indexed: 12/19/2024] Open
Abstract
Circadian rhythms, intrinsic cycles spanning approximately 24 h, regulate numerous physiological processes, including sleep-wake cycles, hormone release, and metabolism. These rhythms are orchestrated by the circadian clock, primarily located in the suprachiasmatic nucleus (SCN) of the hypothalamus. Disruptions in circadian rhythms, whether due to genetic mutations, environmental factors, or lifestyle choices, can significantly impact health, contributing to disorders such as sleep disturbances, metabolic syndrome, and cardiovascular diseases. Additionally, there is a profound link between the disruption of circadian rhythms and development of various cancer, the influence on disease incidence and progression. This incurred regulation by circadian clock on pathways has its implication in tumorigenesis, such as cell cycle control, DNA damage response, apoptosis, and metabolism. Furthermore, the circadian timing system modulates the efficacy and toxicity of cancer treatments. In cancer treatment, the use of chronotherapy to optimize the timing of medical treatments, involves administering chemotherapy, radiation, or other therapeutic interventions at specific intervals to enhance efficacy and minimize side effects. This approach capitalizes on the circadian variations in cellular processes, including DNA repair, cell cycle progression, and drug metabolism. Preclinical and clinical studies have demonstrated that chronotherapy can significantly improve the therapeutic index of chemotherapeutic agents like cisplatin and 5-fluorouracil by enhancing anticancer activity and reducing toxicity. Further research is needed to elucidate the mechanisms underlying circadian regulation of cancer and to develop robust chronotherapeutic protocols tailored to individual patients' circadian profiles, potentially transforming cancer care into more effective and personalized treatment strategies.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- RAK College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.
- Translational and Medical Research Centre (TMRC), Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.
| | - Syed Arman Rabbani
- RAK College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
- Translational and Medical Research Centre (TMRC), Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Areeg Anwer Ali
- RAK College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
- Translational and Medical Research Centre (TMRC), Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Ibrahim Ghaleb Ali Alfaouri
- Translational and Medical Research Centre (TMRC), Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
- RAK College of Nursing, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Hamdi Al Nsairat
- Pharmacological and Diagnostic Research Center, Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Israa Hamid Al-Ani
- Pharmacological and Diagnostic Research Center, Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Pharmacy, Yarmouk University, Irbid, Jordan
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Childcare, Internal Medicine and Medical Specialties, School of Medicine, University of Palermo, Palermo, Italy
| | - Dimitrios Patoulias
- Second Department of Cardiology, Aristotle University of Thessaloniki, Hippokration General Hospital, Athens, Greece
- Outpatient Department of Cardiometabolic Medicine, Second Department of Cardiology, Aristotle University of Thessaloniki, Hippokration General Hospital, Athens, Greece
| | - Mohammad Ahmed Khan
- School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Suhel Parvez
- School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | | |
Collapse
|
48
|
Chen X, Lin E, Haghighatian MM, Shepard LW, Hattar S, Kuruvilla R, Zhao H. Light modulates glucose and lipid homeostasis via the sympathetic nervous system. SCIENCE ADVANCES 2024; 10:eadp3284. [PMID: 39661675 PMCID: PMC11633741 DOI: 10.1126/sciadv.adp3284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024]
Abstract
Light is an important environmental factor for vision and for diverse physiological and psychological functions. Light can also modulate glucose metabolism. Here, we show that in mice, light is critical for glucose and lipid homeostasis by regulating the sympathetic nervous system, independent of circadian disruption. Light deprivation from birth elicits insulin hypersecretion, glucagon hyposecretion, lower gluconeogenesis, and reduced lipolysis by 6 to 8 weeks in male, but not female, mice. These metabolic defects are consistent with blunted sympathetic activity, and indeed, sympathetic responses to a cold stimulus are substantially attenuated in dark-reared mice. Further, long-term dark rearing leads to body weight gain, insulin resistance, and glucose intolerance. Notably, metabolic dysfunction can be partially alleviated by 5 weeks exposure to a regular light-dark cycle. These studies provide insight into circadian-independent mechanisms by which light directly influences whole-body physiology and better understanding of metabolic disorders linked to aberrant environmental light conditions.
Collapse
Affiliation(s)
- Xiangning Chen
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Eugene Lin
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | | - Samer Hattar
- Section on Light and Circadian Rhythms, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
49
|
Horiguchi M, Yoshihara K, Watanabe K, Tsurudome Y, Mizukami Y, Ushijima K. Circadian Rhythms of Clock Genes After Transplantation of Mesenchymal Stem Cells with Type 2 Diabetes Mellitus. Int J Mol Sci 2024; 25:13145. [PMID: 39684854 DOI: 10.3390/ijms252313145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Regenerative therapy involving stem cell transplantation has become an option for the radical treatment of diabetes mellitus. Disruption in the clock genes of stem cells affects the homeostasis of transplanted tissues. We examined the circadian rhythm of genes in transplanted adipose-derived mesenchymal stem cells derived from a patient with type 2 diabetes mellitus (T2DM-ADSC). The clock genes (PER2, CLOCK1, CRY1, and ARNTL[BMAL1]) exhibited similar daily fluctuations in phase and amplitude between a group transplanted with adipose-derived mesenchymal stem cells derived from a healthy individual (N-ADSC) and a group transplanted with T2DM-ADSC. The findings demonstrated that clock genes in stem cells are synchronized with those in living organisms. Next-generation sequencing was then employed to categorize genes that exhibited variation in expression between N-ADSC and T2DM-ADSC. MTATP8P1 and NDUFA7_2 gene expression was significantly reduced at two time points (ZT6 and ZT18), and daily fluctuations were lost. The present study reports, for the first time, that the circadian rhythms of MTATP8P1 and NDUFA7_2, genes involved in mitochondrial processes, are altered in T2DM-ADSC.
Collapse
Affiliation(s)
- Michiko Horiguchi
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan
| | - Kenichi Yoshihara
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan
| | - Kenji Watanabe
- Institute of Gene Research, Yamaguchi University Science Research Center, Yamaguchi 753-0841, Japan
| | - Yuya Tsurudome
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan
| | - Yoichi Mizukami
- Institute of Gene Research, Yamaguchi University Science Research Center, Yamaguchi 753-0841, Japan
| | - Kentaro Ushijima
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan
| |
Collapse
|
50
|
Das S, Khan R, Banerjee S, Ray S, Ray S. Alterations in Circadian Rhythms, Sleep, and Physical Activity in COVID-19: Mechanisms, Interventions, and Lessons for the Future. Mol Neurobiol 2024; 61:10115-10137. [PMID: 38702566 DOI: 10.1007/s12035-024-04178-5] [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: 10/20/2023] [Accepted: 04/04/2024] [Indexed: 05/06/2024]
Abstract
Although the world is acquitting from the throes of COVID-19 and returning to the regularity of life, its effects on physical and mental health are prominently evident in the post-pandemic era. The pandemic subjected us to inadequate sleep and physical activities, stress, irregular eating patterns, and work hours beyond the regular rest-activity cycle. Thus, perturbing the synchrony of the regular circadian clock functions led to chronic psychiatric and neurological disorders and poor immunological response in several COVID-19 survivors. Understanding the links between the host immune system and viral replication machinery from a clock-infection biology perspective promises novel avenues of intervention. Behavioral improvements in our daily lifestyle can reduce the severity and expedite the convalescent stage of COVID-19 by maintaining consistent eating, sleep, and physical activity schedules. Including dietary supplements and nutraceuticals with prophylactic value aids in combating COVID-19, as their deficiency can lead to a higher risk of infection, vulnerability, and severity of COVID-19. Thus, besides developing therapeutic measures, perpetual healthy practices could also contribute to combating the upcoming pandemics. This review highlights the impact of the COVID-19 pandemic on biological rhythms, sleep-wake cycles, physical activities, and eating patterns and how those disruptions possibly contribute to the response, severity, and outcome of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Sandip Das
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284, Telangana, India
| | - Rajni Khan
- National Institute of Pharmaceutical Education and Research (NIPER) - Hajipur, Vaishali, Hajipur, 844102, Bihar, India
| | - Srishti Banerjee
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284, Telangana, India
| | - Shashikant Ray
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, 845401, India.
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Sandipan Ray
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284, Telangana, India.
| |
Collapse
|