1
|
El Jamal N, Brooks TG, Skarke C, FitzGerald GA. Sleep disorders as risk factors for calcific aortic stenosis. Am J Prev Cardiol 2025; 22:100958. [PMID: 40151467 PMCID: PMC11937941 DOI: 10.1016/j.ajpc.2025.100958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/20/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
Background and Aims Circadian disruption and sleep disorders have been shown to increase the risk for many cardiovascular diseases. Their association specifically with valvular heart disease, however, is inconclusive. In this study we test the association between sleep disorders and the future incidence of aortic stenosis using two large electronic health record (EHR) databases datasets (the TriNetX network and the All of Us study). We also explore biochemical data for potential mechanistic insights into that association. Methods We fitted Cox proportional hazards models to quantify the risk of future incidence of AS in patients with sleep disorders. We also explored clinical laboratory test datasets for biochemical signals that might explain the association, running mediation analyses. Results In our fully adjusted Cox models, we find that having any sleep disorder increases the risk for the future incidence of AS (HR: 1.15 95 % CI: 1.13-1.18). Changes in lipid profile mediate a proportion of that association. Conclusion Sleep disorders are associated with an increased risk of AS incidence. That association is independent of classical cardiovascular risk factors even though dyslipidemia plays a large role in mediating this risk.
Collapse
Affiliation(s)
- Nadim El Jamal
- Institute for Translational Medicine and Therapeutics (ITMAT), University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Bldg 421, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Thomas G. Brooks
- Institute for Translational Medicine and Therapeutics (ITMAT), University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Bldg 421, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Carsten Skarke
- Institute for Translational Medicine and Therapeutics (ITMAT), University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Bldg 421, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Garret A. FitzGerald
- Institute for Translational Medicine and Therapeutics (ITMAT), University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Bldg 421, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
2
|
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
|
3
|
Cimino A, Pat F, Oyebamiji O, Pferdehirt L, Pham CTN, Herzog ED, Guilak F. Programmable chronogenetic gene circuits for self-regulated circadian delivery of biologic drugs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643274. [PMID: 40161636 PMCID: PMC11952517 DOI: 10.1101/2025.03.14.643274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Cells of the body rely on the circadian clock to orchestrate daily changes in physiology that impact both homeostatic and pathological conditions, such as the inflammatory autoimmune disease rheumatoid arthritis (RA). In RA, high levels of proinflammatory cytokines peak early in the morning hours, reflected by daily changes in joint stiffness. Chronotherapy (or circadian medicine) seeks to delivery drugs at optimal times to maximize their efficacy. However, chronotherapy remains a largely unexplored approach for disease modifying, antirheumatic treatment, particularly for cell-based therapies. In this study, we developed autonomous chronogenetic gene circuits that produce the biologic drug interleukin-1 receptor antagonist (IL-1Ra) with desired phase and amplitude. We compared expression of IL-1Ra from circuits that contained different circadian promoter elements (E'-boxes, D-boxes, or RREs) and their ability to respond to inflammatory challenges in murine pre-differentiated induced pluripotent stem cells (PDiPSC) or engineered cartilage pellets. We confirmed that each circuit reliably peaked at a distinct circadian time over multiple days. Engineered cells generated significant amounts of IL-1Ra on a circadian basis, which protected them from circadian dysregulation and inflammatory damage. These programmable chronogenetic circuits have the potential to align with an individual's circadian rhythm for optimized, self-regulated daily drug delivery.
Collapse
|
4
|
Zhang Y, Ding H, Huo Z, Chen L. A novel Bayesian hierarchical model for detecting differential circadian pattern in transcriptomic applications. Brief Bioinform 2025; 26:bbaf139. [PMID: 40234105 PMCID: PMC11998590 DOI: 10.1093/bib/bbaf139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/28/2025] [Accepted: 03/01/2025] [Indexed: 04/17/2025] Open
Abstract
Circadian rhythm plays a critical role in regulating various physiological processes, and disruptions in these rhythms have been linked to a wide range of diseases. Identifying molecular biomarkers showing differential circadian (DC) patterns between biological conditions or disease status is important for disease prevention, diagnosis, and treatment. However, circadian pattern is characterized by three key components: amplitude, phase, and MESOR, which poses a great challenge for DC analysis. Existing statistical methods focus on detecting differential shape (amplitude and phase) but often overlook MESOR difference. Additionally, these methods lack flexibility to incorporate external knowledge such as differential circadian information from similar clinical and biological context to improve the current DC analysis. To address these limitation, we introduce a novel Bayesian hierarchical model, BayesDCirc, designed for detecting differential circadian patterns in a two-group experimental design, which offer the advantage of testing MESOR difference and incorporating external knowledge. Benefiting from explicitly testing MESOR within the Bayesian modeling framework, BayesDCirc demonstrates superior FDR control over existing methods, with further performance improvement by leveraging external knowledge of DC genes. Applied to two real datasets, BayesDCirc successfully identify key circadian genes, particularly with external knowledge incorporated. The R package "BayesDCirc" for the method is publicly available on GitHub at https://github.com/lichen-lab/BayesDCirc.
Collapse
Affiliation(s)
- Yutao Zhang
- Department of Biostatistics, University of Florida, Gainesville, FL, 32603, United States
| | - Haocheng Ding
- Department of Biostatistics, Augusta University, Augusta, GA, 30912, United States
| | - Zhiguang Huo
- Department of Biostatistics, University of Florida, Gainesville, FL, 32603, United States
| | - Li Chen
- Department of Biostatistics, University of Florida, Gainesville, FL, 32603, United States
| |
Collapse
|
5
|
Xin M, Bi F, Wang C, Huang Y, Xu Y, Liang S, Cai T, Xu X, Dong L, Li T, Wang X, Fang Y, Xu Z, Wang M, Song X, Zheng Y, Sun W, Li L. The circadian rhythm: A new target of natural products that can protect against diseases of the metabolic system, cardiovascular system, and nervous system. J Adv Res 2025; 69:495-514. [PMID: 38631431 PMCID: PMC11954810 DOI: 10.1016/j.jare.2024.04.005] [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: 12/27/2023] [Revised: 03/17/2024] [Accepted: 04/07/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND The treatment of metabolic system, cardiovascular system, and nervous system diseases remains to be explored. In the internal environment of organisms, the metabolism of substances such as carbohydrates, lipids and proteins (including biohormones and enzymes) exhibit a certain circadian rhythm to maintain the energy supply and material cycle needed for the normal activities of organisms. As a key factor for the health of organisms, the circadian rhythm can be disrupted by pathological conditions, and this disruption accelerates the progression of diseases and results in a vicious cycle. The current treatments targeting the circadian rhythm for the treatment of metabolic system, cardiovascular system, and nervous system diseases have certain limitations, and the identification of safer and more effective circadian rhythm regulators is needed. AIM OF THE REVIEW To systematically assess the possibility of using the biological clock as a natural product target for disease intervention, this work reviews a range of evidence on the potential effectiveness of natural products targeting the circadian rhythm to protect against diseases of the metabolic system, cardiovascular system, and nervous system. This manuscript focuses on how natural products restore normal function by affecting the amplitude of the expression of circadian factors, sleep/wake cycles and the structure of the gut microbiota. KEY SCIENTIFIC CONCEPTS OF THE REVIEW This work proposes that the circadian rhythm, which is regulated by the amplitude of the expression of circadian rhythm-related factors and the sleep/wake cycle, is crucial for diseases of the metabolic system, cardiovascular system and nervous system and is a new target for slowing the progression of diseases through the use of natural products. This manuscript provides a reference for the molecular modeling of natural products that target the circadian rhythm and provides a new perspective for the time-targeted action of drugs.
Collapse
Affiliation(s)
- Meiling Xin
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100000, China
| | - Fangjie Bi
- Heart Center, Zibo Central Hospital, Zibo, Shandong 255000, China
| | - Chao Wang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Yuhong Huang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Yujia Xu
- Department of Echocardiography, Zibo Central Hospital, Zibo, Shandong 255000, China
| | - Shufei Liang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Tianqi Cai
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Xiaoxue Xu
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Ling Dong
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Tianxing Li
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100000, China; Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xueke Wang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100000, China; The Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yini Fang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100000, China; Basic Medical College, Zhejiang Chinese Medical University, Hangzhou 310053 China
| | - Zhengbao Xu
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Meng Wang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Xinhua Song
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China.
| | - Yanfei Zheng
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100000, China.
| | - Wenlong Sun
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China.
| | - Lingru Li
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100000, China.
| |
Collapse
|
6
|
Kutsenko Y, Iñiguez LP, Barreda A, Pardo-Marín L, Toval A, Garrigos D, Martínez-Morga M, Pujante S, Ribeiro Do-Couto B, Tseng KY, Cerón JJ, Garaulet M, Wisniewska MB, Irimia M, Ferran JL. Timing of exercise differentially impacts adipose tissue gain in male adolescent rats. Mol Metab 2025; 93:102100. [PMID: 39832562 PMCID: PMC11810837 DOI: 10.1016/j.molmet.2025.102100] [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: 10/10/2024] [Revised: 01/11/2025] [Accepted: 01/11/2025] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE Circadian rhythms of metabolic, hormonal, and behavioral fluctuations and their alterations can impact health. An important gap in knowledge in the field is whether the time of the day of exercise and the age of onset of exercise exert distinct effects at the level of whole-body adipose tissue and body composition. The goal of the present study was to determine how exercise at different times of the day during adolescence impacts the adipose tissue transcriptome and content in a rodent model. METHODS Rats were subjected to one of four conditions during their adolescence: early active phase control or exercise (EAC or EAE; ZT13), and late active phase control or exercise (LAC or LAE; ZT23). The effects of exercise timing were assessed at the level of subcutaneous and visceral adipose tissue transcriptome, body composition, hypothalamic expression of orexigenic and anorexigenic genes, blood serum markers and 24-hour core body temperature patterns. RESULTS We found that late active phase exercise (ZT23) greatly upregulated pathways of lipid synthesis, glycolysis and NADH shuttles in LAE rats, compared to LAC or EAE. Conversely, LAE rats showed notably lower content of adipose tissue. In addition, LAE rats showed signs of impaired FGF21-adiponectin axis compared to other groups. CONCLUSIONS Finally, LAE rats showed higher post-exercise core body temperature compared to other groups. Our results thus indicate that our exercise protocol induced an unusual effect characterized by enhanced lipid synthesis but reduced adipose tissue content in late active phase but not early active phase exercise during adolescence.
Collapse
Affiliation(s)
- Y Kutsenko
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, 30120, Spain; Institute of Biomedical Research of Murcia, Virgen de la Arrixaca University Hospital, Murcia, 30120, Spain
| | - L P Iñiguez
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, 08003, Spain
| | - A Barreda
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, 30120, Spain; Institute of Biomedical Research of Murcia, Virgen de la Arrixaca University Hospital, Murcia, 30120, Spain
| | - L Pardo-Marín
- Interdisciplinary Laboratory of Clinical Analysis, Interlab UMU, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain
| | - A Toval
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, 30120, Spain; Institute of Biomedical Research of Murcia, Virgen de la Arrixaca University Hospital, Murcia, 30120, Spain; PROFITH "PROmoting FITness and Health Through Physical Activity" Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical Education and Sports, Faculty of Sport Sciences, University of Granada, 18071, Granada, Spain
| | - D Garrigos
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, 30120, Spain; Institute of Biomedical Research of Murcia, Virgen de la Arrixaca University Hospital, Murcia, 30120, Spain
| | - M Martínez-Morga
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, 30120, Spain; Institute of Biomedical Research of Murcia, Virgen de la Arrixaca University Hospital, Murcia, 30120, Spain
| | - S Pujante
- Faculty of Psychology, University of Murcia, Murcia, 30100, Spain
| | - B Ribeiro Do-Couto
- Institute of Biomedical Research of Murcia, Virgen de la Arrixaca University Hospital, Murcia, 30120, Spain; Faculty of Psychology, University of Murcia, Murcia, 30100, Spain
| | - K Y Tseng
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 60612, Chicago, Illinois, USA
| | - J J Cerón
- Interdisciplinary Laboratory of Clinical Analysis, Interlab UMU, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain
| | - M Garaulet
- Department of Physiology, University of Murcia, IMIB-Arrixaca, 30120, Murcia, Spain
| | - M B Wisniewska
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, 02-097, Warsaw, Poland
| | - M Irimia
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, 08003, Spain; Universitat Pompeu Fabra, Barcelona, 08002, Spain; ICREA, Barcelona, 08010, Spain
| | - J L Ferran
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, 30120, Spain; Institute of Biomedical Research of Murcia, Virgen de la Arrixaca University Hospital, Murcia, 30120, Spain.
| |
Collapse
|
7
|
Halder SK, Melkani GC. The Interplay of Genetic Predisposition, Circadian Misalignment, and Metabolic Regulation in Obesity. Curr Obes Rep 2025; 14:21. [PMID: 40024983 PMCID: PMC11872776 DOI: 10.1007/s13679-025-00613-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2025] [Indexed: 03/04/2025]
Abstract
PURPOSE OF REVIEW This review explores the complex interplay between genetic predispositions to obesity, circadian rhythms, metabolic regulation, and sleep. It highlights how genetic factors underlying obesity exacerbate metabolic dysfunction through circadian misalignment and examines promising interventions to mitigate these effects. RECENT FINDINGS Genome-wide association Studies (GWAS) have identified numerous Single Nucleotide Polymorphisms (SNPs) associated with obesity traits, attributing 40-75% heritability to body mass index (BMI). These findings illuminate critical links between genetic obesity, circadian clocks, and metabolic processes. SNPs in clock-related genes influence metabolic pathways, with disruptions in circadian rhythms-driven by poor sleep hygiene or erratic eating patterns-amplifying metabolic dysfunction. Circadian clocks, synchronized with the 24-h light-dark cycle, regulate key metabolic activities, including glucose metabolism, lipid storage, and energy utilization. Genetic mutations or external disruptions, such as irregular sleep or eating habits, can destabilize circadian rhythms, promoting weight gain and metabolic disorders. Circadian misalignment in individuals with genetic predispositions to obesity disrupts the release of key metabolic hormones, such as leptin and insulin, impairing hunger regulation and fat storage. Interventions like time-restricted feeding (TRF) and structured physical activity offer promising strategies to restore circadian harmony, improve metabolic health, and mitigate obesity-related risks.
Collapse
Affiliation(s)
- Sajal Kumar Halder
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Girish C Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- UAB Nathan Shock Center, Birmingham, AL, 35294, USA.
| |
Collapse
|
8
|
Costantini C, Brancorsini S, Grignani F, Romani L, Bellet MM. Circadian metabolic adaptations to infections. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230473. [PMID: 39842481 PMCID: PMC11753887 DOI: 10.1098/rstb.2023.0473] [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: 01/04/2024] [Revised: 04/22/2024] [Accepted: 05/16/2024] [Indexed: 01/24/2025] Open
Abstract
Circadian clocks are biological oscillators that evolved to coordinate rhythms in behaviour and physiology around the 24-hour day. In mammalian tissues, circadian rhythms and metabolism are highly intertwined. The clock machinery controls rhythmic levels of circulating hormones and metabolites, as well as rate-limiting enzymes catalysing biosynthesis or degradation of macromolecules in metabolic tissues, such control being exerted both at the transcriptional and post-transcriptional level. During infections, major metabolic adaptation occurs in mammalian hosts, at the level of both the single immune cell and the whole organism. Under these circumstances, the rhythmic metabolic needs of the host intersect with those of two other players: the pathogen and the microbiota. These three components cooperate or compete to meet their own metabolic demands across the 24 hours. Here, we review findings describing the circadian regulation of the host response to infection, the circadian metabolic adaptations occurring during host-microbiota-pathogen interactions and how such regulation can influence the immune response of the host and, ultimately, its own survival.This article is part of the Theo Murphy meeting issue 'Circadian rhythms in infection and immunity'.
Collapse
Affiliation(s)
- Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| | - Stefano Brancorsini
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| | - Francesco Grignani
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| | - Marina Maria Bellet
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, Perugia06132, Italy
| |
Collapse
|
9
|
Akpoghiran O, Strich AK, Koh K. Effects of sex, mating status, and genetic background on circadian behavior in Drosophila. Front Neurosci 2025; 18:1532868. [PMID: 39844849 PMCID: PMC11750873 DOI: 10.3389/fnins.2024.1532868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Circadian rhythms play a crucial role in regulating behavior, physiology, and health. Sexual dimorphism, a widespread phenomenon across species, influences circadian behaviors. Additionally, post-mating physiological changes in females are known to modulate various behaviors, yet their effects on circadian rhythms remain underexplored. Here, using Drosophila melanogaster, a powerful model for studying circadian mechanisms, we systematically assessed the impact of sex and mating status on circadian behavior. We measured circadian period length and rhythm strength in virgin and mated males and females, including females mated to males lacking Sex Peptide (SP), a key mediator of post-mating changes. Across four wild-type and control strains, we found that males consistently exhibited shorter circadian periods than females, regardless of mating status, suggesting that circadian period length is a robust sexually dimorphic trait. In contrast, rhythm strength was influenced by the interaction between sex and mating status, with female mating generally reducing rhythm strength in the presence of SP signaling. Notably, genetic background significantly modulated these effects on rhythm strength. Our findings demonstrate that while circadian period length is a stable sex-specific trait, rhythm strength is shaped by a complex interplay between sex, mating status, and genetic background. This study advances our understanding of how sex and mating influence circadian rhythms in Drosophila and provides a foundation for future research into sexually dimorphic mechanisms underlying human diseases associated with circadian disruptions.
Collapse
Affiliation(s)
| | | | - Kyunghee Koh
- Department of Neuroscience, Farber Institute for Neurosciences, Synaptic Biology Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
10
|
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
|
11
|
Tam CHT, Wang Y, Wang CC, Yuen LY, Lim CKP, Leng J, Wu L, Ng ACW, Hou Y, Tsoi KY, Wang H, Ozaki R, Li AM, Wang Q, Chan JCN, Ye YC, Tam WH, Yang X, Ma RCW. Identification and Potential Clinical Utility of Common Genetic Variants in Gestational Diabetes among Chinese Pregnant Women. Diabetes Metab J 2025; 49:128-143. [PMID: 39301664 PMCID: PMC11788552 DOI: 10.4093/dmj.2024.0139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/17/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGRUOUND The genetic basis for hyperglycaemia in pregnancy remain unclear. This study aimed to uncover the genetic determinants of gestational diabetes mellitus (GDM) and investigate their applications. METHODS We performed a meta-analysis of genome-wide association studies (GWAS) for GDM in Chinese women (464 cases and 1,217 controls), followed by de novo replications in an independent Chinese cohort (564 cases and 572 controls) and in silico replication in European (12,332 cases and 131,109 controls) and multi-ethnic populations (5,485 cases and 347,856 controls). A polygenic risk score (PRS) was derived based on the identified variants. RESULTS Using the genome-wide scan and candidate gene approaches, we identified four susceptibility loci for GDM. These included three previously reported loci for GDM and type 2 diabetes mellitus (T2DM) at MTNR1B (rs7945617, odds ratio [OR], 1.64; 95% confidence interval [CI], 1.38 to 1.96), CDKAL1 (rs7754840, OR, 1.33; 95% CI, 1.13 to 1.58), and INS-IGF2-KCNQ1 (rs2237897, OR, 1.48; 95% CI, 1.23 to 1.79), as well as a novel genome-wide significant locus near TBR1-SLC4A10 (rs117781972, OR, 2.05; 95% CI, 1.61 to 2.62; Pmeta=7.6×10-9), which has not been previously reported in GWAS for T2DM or glycaemic traits. Moreover, we found that women with a high PRS (top quintile) had over threefold (95% CI, 2.30 to 4.09; Pmeta=3.1×10-14) and 71% (95% CI, 1.08 to 2.71; P=0.0220) higher risk for GDM and abnormal glucose tolerance post-pregnancy, respectively, compared to other individuals. CONCLUSION Our results indicate that the genetic architecture of glucose metabolism exhibits both similarities and differences between the pregnant and non-pregnant states. Integrating genetic information can facilitate identification of pregnant women at a higher risk of developing GDM or later diabetes.
Collapse
Affiliation(s)
- Claudia Ha-ting Tam
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Center in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ying Wang
- Scientific Research Platform of the Second School of Clinical Medicine, Guangdong Medical University, Dongguan, China
| | - Chi Chiu Wang
- Department of Obstetrics and Gynecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Development and Reproduction Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong, China
- Chinese University of Hong Kong-Sichuan University Joint Laboratory in Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Lai Yuk Yuen
- Department of Obstetrics and Gynecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Cadmon King-poo Lim
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Center in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Junhong Leng
- Department of Children’s Health, Tianjin Women and Children’s Health Center, Tianjin, China
| | - Ling Wu
- Department of Obstetrics and Gynecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Alex Chi-wai Ng
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Yong Hou
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Kit Ying Tsoi
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Hui Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Risa Ozaki
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
| | - Albert Martin Li
- Department of Pediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Qingqing Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Juliana Chung-ngor Chan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Center in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yan Chou Ye
- Department of Obstetrics and Gynecology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Wing Hung Tam
- Department of Obstetrics and Gynecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Ronald Ching-wan Ma
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-SJTU Joint Research Center in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
12
|
Akpoghiran O, Strich AK, Koh K. Effects of sex, mating status, and genetic background on circadian behavior in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624853. [PMID: 39605702 PMCID: PMC11601570 DOI: 10.1101/2024.11.22.624853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Circadian rhythms play a crucial role in regulating behavior, physiology, and health. Sexual dimorphism, a widespread phenomenon across species, influences circadian behaviors. Additionally, post-mating physiological changes in females are known to modulate various behaviors, yet their effects on circadian rhythms remain underexplored. Here, using Drosophila melanogaster, a powerful model for studying circadian mechanisms, we systematically assessed the impact of sex and mating status on circadian behavior. We measured circadian period length and rhythm strength in virgin and mated males and females, including females mated to males lacking Sex Peptide (SP), a key mediator of post-mating changes. Across four wild-type and control strains, we found that males consistently exhibited shorter circadian periods than females, regardless of mating status, suggesting that circadian period length is a robust sexually dimorphic trait. In contrast, rhythm strength was influenced by the interaction between sex and mating status, with female mating generally reducing rhythm strength in the presence of SP signaling. Notably, genetic background significantly modulated these effects on rhythm strength. Our findings demonstrate that while circadian period length is a stable sex-specific trait, rhythm strength is shaped by a complex interplay between sex, mating status, and genetic background. This study advances our understanding of how sex and mating influence circadian rhythms in Drosophila and provides a foundation for future research into sexually dimorphic mechanisms underlying human diseases associated with circadian disruptions.
Collapse
Affiliation(s)
- Oghenerukevwe Akpoghiran
- Department of Neuroscience, Farber Institute for Neurosciences, and Synaptic Biology Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Alexandra K. Strich
- Department of Neuroscience, Farber Institute for Neurosciences, and Synaptic Biology Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Kyunghee Koh
- Department of Neuroscience, Farber Institute for Neurosciences, and Synaptic Biology Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
13
|
Carra MC, Balagny P, Bouchard P. Sleep and periodontal health. Periodontol 2000 2024; 96:42-73. [PMID: 39233377 PMCID: PMC11579834 DOI: 10.1111/prd.12611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/18/2024] [Accepted: 08/19/2024] [Indexed: 09/06/2024]
Abstract
Sleep is fundamental for health and well-being. An adequate amount and quality of sleep is a cardinal component of a healthy lifestyle at the basis of the prevention of many non-communicable chronic diseases. Recent evidence suggests that sleep disorders, particularly obstructive sleep apnea, represent an emerging risk factor for periodontal health. This review article provides a critical appraisal of the existing literature concerning the association between sleep duration, sleep quality, sleep disorders in general, and obstructive sleep apnea with periodontal diseases, including gingivitis and periodontitis. The putative mechanisms underlying these associations are described as well as the potential clinical implications for diagnosis and treatment.
Collapse
Affiliation(s)
- Maria Clotilde Carra
- UFR of Odontology, Université Paris CitéParisFrance
- Service of Odontology, Rothschild Hospital (AP‐HP)ParisFrance
- METHODS Team, CRESS, INSERM, INRAe, Université Paris CitéParisFrance
| | - Pauline Balagny
- INSERM, UMS 011 Population‐based Cohorts UnitUniversité Paris Cité, Paris Saclay University, Université de Versailles Saint‐Quentin‐en‐YvelinesParisFrance
- Department of Physiology Functional ExplorationHôpital Bichat (AP‐HP)ParisFrance
| | - Philippe Bouchard
- UFR of Odontology, Université Paris CitéParisFrance
- URP 2496MontrougeFrance
| |
Collapse
|
14
|
Rashed N, Liu W, Zhou X, Bode AM, Luo X. The role of circadian gene CLOCK in cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119782. [PMID: 38871225 DOI: 10.1016/j.bbamcr.2024.119782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024]
Abstract
Circadian Locomotor Output Cycles Kaput (CLOCK) is one of the circadian clock genes and is considered to be a fundamental regulatory gene in the circadian rhythm, responsible for mediating several biological processes. Therefore, abnormal expression of CLOCK affects its role in the circadian clock and its more general function as a direct regulator of gene expression. This dysfunction can lead to severe pathological effects, including cancer. To better understand the role of CLOCK in cancer, we compiled this review to describe the biological function of CLOCK, and especially highlighted its function in cancer development, progression, tumor microenvironment, cancer cell metabolism, and drug resistance.
Collapse
Affiliation(s)
- Nasot Rashed
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; NHC Key Laboratory of Carcinogenesis, the Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China
| | - Wenbin Liu
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; Department of Pathology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China
| | - Xinran Zhou
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; NHC Key Laboratory of Carcinogenesis, the Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Xiangjian Luo
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; NHC Key Laboratory of Carcinogenesis, the Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China; Key Laboratory of Biological Nanotechnology of National Health Commission, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
15
|
Rajan PK, Udoh UAS, Finley R, Pierre SV, Sanabria J. The Biological Clock of Liver Metabolism in Metabolic Dysfunction-Associated Steatohepatitis Progression to Hepatocellular Carcinoma. Biomedicines 2024; 12:1961. [PMID: 39335475 PMCID: PMC11428469 DOI: 10.3390/biomedicines12091961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024] Open
Abstract
Circadian rhythms are endogenous behavioral or physiological cycles that are driven by a daily biological clock that persists in the absence of geophysical or environmental temporal cues. Circadian rhythm-related genes code for clock proteins that rise and fall in rhythmic patterns driving biochemical signals of biological processes from metabolism to physiology and behavior. Clock proteins have a pivotal role in liver metabolism and homeostasis, and their disturbances are implicated in various liver disease processes. Encoded genes play critical roles in the initiation and progression of metabolic dysfunction-associated steatohepatitis (MASH) to hepatocellular carcinoma (HCC) and their proteins may become diagnostic markers as well as therapeutic targets. Understanding molecular and metabolic mechanisms underlying circadian rhythms will aid in therapeutic interventions and may have broader clinical applications. The present review provides an overview of the role of the liver's circadian rhythm in metabolic processes in health and disease, emphasizing MASH progression and the oncogenic associations that lead to HCC.
Collapse
Affiliation(s)
- Pradeep Kumar Rajan
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Utibe-Abasi S Udoh
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Robert Finley
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
| | - Juan Sanabria
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
- Department of Nutrition and Metabolomic Core Facility, School of Medicine, Case Western Reserve University, Cleveland, OH 44100, USA
| |
Collapse
|
16
|
Ribas-Latre A, Fernández-Veledo S, Vendrell J. Time-restricted eating, the clock ticking behind the scenes. Front Pharmacol 2024; 15:1428601. [PMID: 39175542 PMCID: PMC11338815 DOI: 10.3389/fphar.2024.1428601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
Introduction Maintaining metabolic balance relies on accumulating nutrients during feeding periods and their subsequent release during fasting. In obesity and metabolic disorders, strategies aimed at reducing food intake while simulating fasting have garnered significant attention for weight loss. Caloric restriction (CR) diets and intermittent fasting (IF) interventions have emerged as effective approaches to improving cardiometabolic health. Although the comparative metabolic benefits of CR versus IF remain inconclusive, this review focuses on various forms of IF, particularly time-restricted eating (TRE). Methods This study employs a narrative review methodology, systematically collecting, synthesizing, and interpreting the existing literature on TRE and its metabolic effects. A comprehensive and unbiased search of relevant databases was conducted to identify pertinent studies, including pre-clinical animal studies and clinical trials in humans. Keywords such as "Obesity," "Intermittent Fasting," "Time-restricted eating," "Chronotype," and "Circadian rhythms" guided the search. The selected studies were critically appraised based on predefined inclusion and exclusion criteria, allowing for a thorough exploration and synthesis of current knowledge. Results This article synthesizes pre-clinical and clinical studies on TRE and its metabolic effects, providing a comprehensive overview of the current knowledge and identifying gaps for future research. It explores the metabolic outcomes of recent clinical trials employing different TRE protocols in individuals with overweight, obesity, or type II diabetes, emphasizing the significance of individual chronotype, which is often overlooked in practice. In contrast to human studies, animal models underscore the role of the circadian clock in mitigating metabolic disturbances induced by obesity through time-restricted feeding (TRF) interventions. Consequently, we examine pre-clinical evidence supporting the interplay between the circadian clock and TRF interventions. Additionally, we provide insights into the role of the microbiota, which TRE can modulate and its influence on circadian rhythms.
Collapse
Affiliation(s)
- Aleix Ribas-Latre
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Departament de Medicina i Cirugia, Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Sonia Fernández-Veledo
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Departament de Medicina i Cirugia, Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Joan Vendrell
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Departament de Medicina i Cirugia, Universitat Rovira i Virgili (URV), Tarragona, Spain
| |
Collapse
|
17
|
Carra MC, Cistulli PA. Exploring the links between periodontal diseases and obstructive sleep apnoea: An overview for clinicians. Aust Dent J 2024; 69 Suppl 1:S31-S37. [PMID: 39323042 DOI: 10.1111/adj.13040] [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] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
Both periodontal diseases (PDs) and obstructive sleep apnoea (OSA) are highly prevalent disorders with global impact, associated with a large burden at individual patient and health system levels. These disorders often co-exist, but there is growing evidence that the association between the disorders goes beyond an overlap between two highly prevalent diseases that have shared risk factors. Evidence suggests a potential causal relationship, although further research is required to verify this. Regardless of any causal relationship, the co-existence of these disorders is important to recognize since they may act in combination to heighten health risks, particularly cardiovascular risk. Thus, dentists have an important role in screening for OSA in patients presenting with PDs, and similarly, they need to evaluate periodontal health in patients requiring treatment for OSA. Here we provide a narrative review of the association between PDs and OSA to raise awareness among clinicians and promote multidisciplinary collaborations that aim at an evidence-based and effective management of such patients.
Collapse
Affiliation(s)
- M C Carra
- Université Paris Cité, METHODS Team, CRESS, INSERM, INRAe, Paris, France
- Departement of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - P A Cistulli
- Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
- Department of Respiratory and Sleep Medicine, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
18
|
Saint-Maurice PF, Freeman JR, Russ D, Almeida JS, Shams-White MM, Patel S, Wolff-Hughes DL, Watts EL, Loftfield E, Hong HG, Moore SC, Matthews CE. Associations between actigraphy-measured sleep duration, continuity, and timing with mortality in the UK Biobank. Sleep 2024; 47:zsad312. [PMID: 38066693 PMCID: PMC10925955 DOI: 10.1093/sleep/zsad312] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 11/23/2023] [Indexed: 01/12/2024] Open
Abstract
STUDY OBJECTIVES To examine the associations between sleep duration, continuity, timing, and mortality using actigraphy among adults. METHODS Data were from a cohort of 88 282 adults (40-69 years) in UK Biobank that wore a wrist-worn triaxial accelerometer for 7 days. Actigraphy data were processed to generate estimates of sleep duration and other sleep characteristics including wake after sleep onset (WASO), number of 5-minute awakenings, and midpoint for sleep onset/wake-up and the least active 5 hours (L5). Data were linked to mortality outcomes with follow-up to October 31, 2021. We implemented Cox models (hazard ratio, confidence intervals [HR, 95% CI]) to quantify sleep associations with mortality. Models were adjusted for demographics, lifestyle factors, and medical conditions. RESULTS Over an average of 6.8 years 2973 deaths occurred (1700 cancer, 586 CVD deaths). Overall sleep duration was significantly associated with risk for all-cause (p < 0.01), cancer (p < 0.01), and CVD (p = 0.03) mortality. For example, when compared to sleep durations of 7.0 hrs/d, durations of 5 hrs/d were associated with a 29% higher risk for all-cause mortality (HR: 1.29 [1.09, 1.52]). WASO and number of awakenings were not associated with mortality. Individuals with L5 early or late midpoints (<2:30 or ≥ 3:30) had a ~20% higher risk for all-cause mortality, compared to those with intermediate L5 midpoints (3:00-3:29; p ≤ 0.01; e.g. HR ≥ 3:30: 1.19 [1.07, 1.32]). CONCLUSIONS Shorter sleep duration and both early and late sleep timing were associated with a higher mortality risk. These findings reinforce the importance of public health efforts to promote healthy sleep patterns in adults.
Collapse
Affiliation(s)
- Pedro F Saint-Maurice
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | - Joshua R Freeman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel Russ
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jonas S Almeida
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marissa M Shams-White
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shreya Patel
- Department of Epidemiology and Biostatistics, Dornsife School of Public Health, Drexel University, Philadelphia, USA
| | - Dana L Wolff-Hughes
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eleanor L Watts
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Erikka Loftfield
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hyokyoung G Hong
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Steven C Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charles E Matthews
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
19
|
Engin A. Misalignment of Circadian Rhythms in Diet-Induced Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:27-71. [PMID: 39287848 DOI: 10.1007/978-3-031-63657-8_2] [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: 09/19/2024]
Abstract
The biological clocks of the circadian timing system coordinate cellular and physiological processes and synchronize them with daily cycles. While the central clock in the suprachiasmatic nucleus (SCN) is mainly synchronized by the light/dark cycles, the peripheral clocks react to other stimuli, including the feeding/fasting state, nutrients, sleep-wake cycles, and physical activity. During the disruption of circadian rhythms due to genetic mutations or social and occupational obligations, incorrect arrangement between the internal clock system and environmental rhythms leads to the development of obesity. Desynchronization between the central and peripheral clocks by altered timing of food intake and diet composition leads to uncoupling of the peripheral clocks from the central pacemaker and to the development of metabolic disorders. The strong coupling of the SCN to the light-dark cycle creates a situation of misalignment when food is ingested during the "wrong" time of day. Food-anticipatory activity is mediated by a self-sustained circadian timing, and its principal component is a food-entrainable oscillator. Modifying the time of feeding alone greatly affects body weight, whereas ketogenic diet (KD) influences circadian biology, through the modulation of clock gene expression. Night-eating behavior is one of the causes of circadian disruption, and night eaters have compulsive and uncontrolled eating with severe obesity. By contrast, time-restricted eating (TRE) restores circadian rhythms through maintaining an appropriate daily rhythm of the eating-fasting cycle. The hypothalamus has a crucial role in the regulation of energy balance rather than food intake. While circadian locomotor output cycles kaput (CLOCK) expression levels increase with high-fat diet-induced obesity, peroxisome proliferator-activated receptor-alpha (PPARα) increases the transcriptional level of brain and muscle aryl hydrocarbon receptor nuclear translocator (ARNT)-like 1 (BMAL1) in obese subjects. In this context, effective timing of chronotherapies aiming to correct SCN-driven rhythms depends on an accurate assessment of the SCN phase. In fact, in a multi-oscillator system, local rhythmicity and its disruption reflects the disruption of either local clocks or central clocks, thus imposing rhythmicity on those local tissues, whereas misalignment of peripheral oscillators is due to exosome-based intercellular communication.Consequently, disruption of clock genes results in dyslipidemia, insulin resistance, and obesity, while light exposure during the daytime, food intake during the daytime, and sleeping during the biological night promote circadian alignment between the central and peripheral clocks. Thus, shift work is associated with an increased risk of obesity, diabetes, and cardiovascular diseases because of unusual eating times as well as unusual light exposure and disruption of the circadian rhythm.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
20
|
Li C, Yang D, Yang W, Wang Y, Li D, Li Y, Xiao B, Zhang H, Zhao H, Dong H, Zhang J, Chu G, Wang A, Jin Y, Liu Y, Chen H. Hypoxia activation attenuates progesterone synthesis in goat trophoblast cells via NR1D1 inhibition of StAR expression†. Biol Reprod 2023; 109:720-735. [PMID: 37552055 DOI: 10.1093/biolre/ioad094] [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: 01/10/2023] [Revised: 07/03/2023] [Accepted: 08/02/2023] [Indexed: 08/09/2023] Open
Abstract
Trophoblast plays a crucial role in gestation maintenance and embryo implantation, partly due to the synthesis of progesterone. It has been demonstrated that hypoxia regulates invasion, proliferation, and differentiation of trophoblast cells. Additionally, human trophoblasts display rhythmic expression of circadian clock genes. However, it remains unclear if the circadian clock system is present in goat trophoblast cells (GTCs), and its involvement in hypoxia regulation of steroid hormone synthesis remains elusive. In this study, immunofluorescence staining revealed that both BMAL1 and NR1D1 (two circadian clock components) were highly expressed in GTCs. Quantitative real-time PCR analysis showed that several circadian clock genes were rhythmically expressed in forskolin-synchronized GTCs. To mimic hypoxia, GTCs were treated with hypoxia-inducing reagents (CoCl2 or DMOG). Quantitative real-time PCR results demonstrated that hypoxia perturbed the mRNA expression of circadian clock genes and StAR. Notably, the increased expression of NR1D1 and the reduction of StAR expression in hypoxic GTCs were also detected by western blotting. In addition, progesterone secretion exhibited a notable decline in hypoxic GTCs. SR9009, an NR1D1 agonist, significantly decreased StAR expression at both the mRNA and protein levels and markedly inhibited progesterone secretion in GTCs. Moreover, SR8278, an NR1D1 antagonist, partially reversed the inhibitory effect of CoCl2 on mRNA and protein expression levels of StAR and progesterone synthesis in GTCs. Our results demonstrate that hypoxia reduces StAR expression via the activation of NR1D1 signaling in GTCs, thus inhibiting progesterone synthesis. These findings provide new insights into the NR1D1 regulation of progesterone synthesis in GTCs under hypoxic conditions.
Collapse
Affiliation(s)
- Chao Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Dan Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Wanghao Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yiqun Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Dan Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yating Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Bonan Xiao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Haisen Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Hongcong Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Hao Dong
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jing Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Guiyan Chu
- Laboratory of Animal Fat Deposition & Muscle Development, Department of Animal Genetics Breeding and Reproduction, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Aihua Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yaping Jin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yingqiu Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Huatao Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
21
|
Van Gilst D, Puchkina AV, Roelants JA, Kervezee L, Dudink J, Reiss IKM, Van Der Horst GTJ, Vermeulen MJ, Chaves I. Effects of the neonatal intensive care environment on circadian health and development of preterm infants. Front Physiol 2023; 14:1243162. [PMID: 37719464 PMCID: PMC10500197 DOI: 10.3389/fphys.2023.1243162] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/18/2023] [Indexed: 09/19/2023] Open
Abstract
The circadian system in mammals ensures adaptation to the light-dark cycle on Earth and imposes 24-h rhythmicity on metabolic, physiological and behavioral processes. The central circadian pacemaker is located in the brain and is entrained by environmental signals called Zeitgebers. From here, neural, humoral and systemic signals drive rhythms in peripheral clocks in nearly every mammalian tissue. During pregnancy, disruption of the complex interplay between the mother's rhythmic signals and the fetal developing circadian system can lead to long-term health consequences in the offspring. When an infant is born very preterm, it loses the temporal signals received from the mother prematurely and becomes totally dependent on 24/7 care in the Neonatal Intensive Care Unit (NICU), where day/night rhythmicity is usually blurred. In this literature review, we provide an overview of the fetal and neonatal development of the circadian system, and short-term consequences of disruption of this process as occurs in the NICU environment. Moreover, we provide a theoretical and molecular framework of how this disruption could lead to later-life disease. Finally, we discuss studies that aim to improve health outcomes after preterm birth by studying the effects of enhancing rhythmicity in light and noise exposure.
Collapse
Affiliation(s)
- D. Van Gilst
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - A. V. Puchkina
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - J. A. Roelants
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus University Medical Center Rotterdam-Sophia Children’s Hospital, Rotterdam, Netherlands
| | - L. Kervezee
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - J. Dudink
- Department of Neonatology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - I. K. M. Reiss
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus University Medical Center Rotterdam-Sophia Children’s Hospital, Rotterdam, Netherlands
| | - G. T. J. Van Der Horst
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - M. J. Vermeulen
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus University Medical Center Rotterdam-Sophia Children’s Hospital, Rotterdam, Netherlands
| | - I. Chaves
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
22
|
Iweka CA, Seigneur E, Hernandez AL, Paredes SH, Cabrera M, Blacher E, Pasternak CT, Longo FM, de Lecea L, Andreasson KI. Myeloid deficiency of the intrinsic clock protein BMAL1 accelerates cognitive aging by disrupting microglial synaptic pruning. J Neuroinflammation 2023; 20:48. [PMID: 36829230 PMCID: PMC9951430 DOI: 10.1186/s12974-023-02727-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 02/10/2023] [Indexed: 02/26/2023] Open
Abstract
Aging is associated with loss of circadian immune responses and circadian gene transcription in peripheral macrophages. Microglia, the resident macrophages of the brain, also show diurnal rhythmicity in regulating local immune responses and synaptic remodeling. To investigate the interaction between aging and microglial circadian rhythmicity, we examined mice deficient in the core clock transcription factor, BMAL1. Aging Cd11bcre;Bmallox/lox mice demonstrated accelerated cognitive decline in association with suppressed hippocampal long-term potentiation and increases in immature dendritic spines. C1q deposition at synapses and synaptic engulfment were significantly decreased in aging Bmal1-deficient microglia, suggesting that BMAL1 plays a role in regulating synaptic pruning in aging. In addition to accelerated age-associated hippocampal deficits, Cd11bcre;Bmallox/lox mice also showed deficits in the sleep-wake cycle with increased wakefulness across light and dark phases. These results highlight an essential role of microglial BMAL1 in maintenance of synapse homeostasis in the aging brain.
Collapse
Affiliation(s)
- Chinyere Agbaegbu Iweka
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Erica Seigneur
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Amira Latif Hernandez
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | | | - Mica Cabrera
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Eran Blacher
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus Givat-Ram, 9190401, Jerusalem, Israel
| | - Connie Tsai Pasternak
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Katrin I Andreasson
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Stanford Immunology Program, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA.
| |
Collapse
|
23
|
Chiarello DI, Ustáriz J, Marín R, Carrasco-Wong I, Farías M, Giordano A, Gallardo FS, Illanes SE, Gutiérrez J. Cellular mechanisms linking to outdoor and indoor air pollution damage during pregnancy. Front Endocrinol (Lausanne) 2023; 14:1084986. [PMID: 36875486 PMCID: PMC9974835 DOI: 10.3389/fendo.2023.1084986] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
Pregnancies are a critical window period for environmental influences over the mother and the offspring. There is a growing body of evidence associating indoor and outdoor air pollution exposure to adverse pregnancy outcomes such as preterm birth and hypertensive disorders of pregnancy. Particulate matter (PM) could trigger oxi-inflammation and could also reach the placenta leading to placental damage with fetal consequences. The combination of strategies such as risk assessment, advise about risks of environmental exposures to pregnant women, together with nutritional strategies and digital solutions to monitor air quality can be effective in mitigating the effects of air pollution during pregnancy.
Collapse
Affiliation(s)
- Delia I. Chiarello
- Cellular Signaling and Differentiation Laboratory (CSDL), School of Medical Technology, Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| | - Javier Ustáriz
- Department of Chemical and Bioprocess Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Reinaldo Marín
- Center for Biophysics and Biochemistry (CBB), Venezuelan Institute for Scientific Research (IVIC), Caracas, Venezuela
| | - Ivo Carrasco-Wong
- Cellular Signaling and Differentiation Laboratory (CSDL), School of Medical Technology, Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| | - Marcelo Farías
- Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ady Giordano
- Inorganic Chemistry Department, Faculty of Chemistry and of Pharmacy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe S. Gallardo
- Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sebastián E. Illanes
- Reproductive Biology Program, Center for Biomedical Research and Innovation (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Jaime Gutiérrez
- Cellular Signaling and Differentiation Laboratory (CSDL), School of Medical Technology, Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| |
Collapse
|
24
|
Roth JR, Varshney S, de Moraes RCM, Melkani GC. Circadian-mediated regulation of cardiometabolic disorders and aging with time-restricted feeding. Obesity (Silver Spring) 2023; 31 Suppl 1:40-49. [PMID: 36623845 PMCID: PMC10089654 DOI: 10.1002/oby.23664] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 01/11/2023]
Abstract
Circadian rhythms are present throughout biology, from the molecular level to complex behaviors such as eating and sleeping. They are driven by molecular clocks within cells, and different tissues can have unique rhythms. Circadian disruption can trigger obesity and other common metabolic disorders such as aging, diabetes, and cardiovascular disease, and circadian genes control metabolism. At an organismal level, feeding and fasting rhythms are key drivers of circadian rhythms. This underscores the bidirectional relationship between metabolism and circadian rhythms, and many metabolic disorders have circadian disruption or misalignment. Therefore, studying circadian rhythms may offer new avenues for understanding the etiology and management of obesity. This review describes how circadian rhythm dysregulation is linked with cardiometabolic disorders and how the lifestyle intervention of time-restricted feeding (TRF) regulates them. TRF reinforces feeding-fasting rhythms without reducing caloric intake and ameliorates metabolic disorders such as obesity and associated cardiac dysfunction, along with reducing inflammation. TRF optimizes the expression of genes and pathways related to normal metabolic function, linking metabolism with TRF's benefits and demonstrating the molecular link between metabolic disorders and circadian rhythms. Thus, TRF has tremendous therapeutic potential that could be easily adopted to reduce obesity-linked dysfunction and cardiometabolic disorders.
Collapse
Affiliation(s)
- Jonathan R. Roth
- Department of Pathology, Division of Molecular and Cellular Pathology, School of Medicine, The University of Alabama at Birmingham, AL 35294, USA
| | - Shweta Varshney
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ruan Carlos Macedo de Moraes
- Department of Pathology, Division of Molecular and Cellular Pathology, School of Medicine, The University of Alabama at Birmingham, AL 35294, USA
| | - Girish C. Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, School of Medicine, The University of Alabama at Birmingham, AL 35294, USA
| |
Collapse
|
25
|
Kamat PK, Khan MB, Smith C, Siddiqui S, Baban B, Dhandapani K, Hess DC. The time dimension to stroke: Circadian effects on stroke outcomes and mechanisms. Neurochem Int 2023; 162:105457. [PMID: 36442686 PMCID: PMC9839555 DOI: 10.1016/j.neuint.2022.105457] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/14/2022] [Accepted: 11/19/2022] [Indexed: 11/26/2022]
Abstract
The circadian system is widely involved in the various pathological outcomes affected by time dimension changes. In the brain, the master circadian clock, also known as the "pacemaker," is present in the hypothalamus's suprachiasmatic nucleus (SCN). The SCN consists of molecular circadian clocks that operate in each neuron and other brain cells. These circadian mechanisms are controlled by the transcription and translation of specific genes such as the clock circadian regulator (Clock) and brain and muscle ARNT-Like 1 (Bmal1). Period (Per1-3) and cryptochrome (Cry1 and 2) negatively feedback and regulate the clock genes. Variations in the circadian cycle and these clock genes can affect stroke outcomes. Studies suggest that the peak stroke occurs in the morning after patients awaken from sleep, while stroke severity and poor outcomes worsen at midnight. The main risk factor associated with stroke is high blood pressure (hypertension). Blood pressure usually dips by 15-20% during sleep, but many hypertensives do not display this normal dipping pattern and are non-dippers. A sleep blood pressure is the primary determinant of stroke risk. This article discusses the possible mechanism associated with circadian rhythm and stroke outcomes.
Collapse
Affiliation(s)
- Pradip K Kamat
- Departments of Neurology, Medical College of Georgia, Augusta University, USA.
| | | | - Cameron Smith
- Departments of Neurology, Medical College of Georgia, Augusta University, USA
| | - Shahneela Siddiqui
- Departments of Neurology, Medical College of Georgia, Augusta University, USA
| | - Babak Baban
- Departments of Oral Biology, Dental College of Georgia, Augusta University, USA
| | - Krishnan Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, USA
| | - David C Hess
- Departments of Neurology, Medical College of Georgia, Augusta University, USA
| |
Collapse
|
26
|
Fang Y, Jo SK, Park SJ, Yang J, Ko YS, Lee HY, Oh SW, Cho WY, Kim K, Son GH, Kim MG. Role of the Circadian Clock and Effect of Time-Restricted Feeding in Adenine-Induced Chronic Kidney Disease. J Transl Med 2023; 103:100008. [PMID: 36748191 DOI: 10.1016/j.labinv.2022.100008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/02/2022] [Accepted: 08/11/2022] [Indexed: 01/19/2023] Open
Abstract
Most physiological functions exhibit circadian rhythmicity that is partly regulated by the molecular circadian clock. Herein, we investigated the relationship between the circadian clock and chronic kidney disease (CKD). The role of the clock gene in adenine-induced CKD and the mechanisms of interaction were investigated in mice in which Bmal1, the master regulator of the clock gene, was knocked out, and Bmal1 knockout (KO) tubule cells. We also determined whether the renoprotective effect of time-restricted feeding (TRF), a dietary strategy to enhance circadian rhythm, is clock gene-dependent. The mice with CKD showed altered expression of the core clock genes with a loss of diurnal variations in renal functions and key tubular transporter gene expression. Bmal1 KO mice developed more severe fibrosis, and transcriptome profiling followed by gene ontology analysis suggested that genes associated with the cell cycle, inflammation, and fatty acid oxidation pathways were significantly affected in the mutant mice. Tubule-specific deletion of BMAL1 in HK-2 cells by CRISPR/Cas9 led to upregulation of p21 and tumor necrosis α and exacerbated epithelial-mesenchymal transition-related gene expression upon transforming growth factor β stimulation. Finally, TRF in the mice with CKD partially restored the disrupted oscillation of the kidney clock genes, accompanied by improved cell cycle arrest and inflammation, leading to decreased fibrosis. However, the renoprotective effect of TRF was abolished in Bmal1 KO mice, suggesting that TRF is partially dependent on the clock gene. Our data demonstrate that the molecular clock system plays an important role in CKD via cell cycle regulation and inflammation. Understanding the role of the circadian clock in kidney diseases can be a new research field for developing novel therapeutic targets.
Collapse
Affiliation(s)
- Yina Fang
- Division of Nephrology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sang-Kyung Jo
- Division of Nephrology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Soo-Ji Park
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea; Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jihyun Yang
- Division of Nephrology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yoon Sook Ko
- Division of Nephrology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hee Young Lee
- Division of Nephrology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Se Won Oh
- Division of Nephrology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Won Yong Cho
- Division of Nephrology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyoungmi Kim
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea; Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea.
| | - Gi Hoon Son
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea.
| | - Myung-Gyu Kim
- Division of Nephrology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Rosa I, Lages M, Grilo C, Barros R, Guarino MP. mHealth Applications to Monitor Lifestyle Behaviors and Circadian Rhythm in Clinical Settings: Current Perspective and Future Directions. Front Public Health 2022; 10:862065. [PMID: 35923965 PMCID: PMC9339674 DOI: 10.3389/fpubh.2022.862065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
Metabolic diseases are a global rising health burden, mainly due to the deleterious interaction of current lifestyles with the underlying biology of these diseases. Daily habits and behaviors, such as diet, sleep, and physical exercise impact the whole-body circadian system through the synchronization of the peripheral body clocks that contribute to metabolic homeostasis. The disruption of this system may promote the development of metabolic diseases, including obesity and diabetes, emphasizing the importance of assessing and monitoring variables that affect circadian rhythms. Advances in technology are generating innovative resources and tools for health care management and patient monitoring, particularly important for chronic conditions. The use of mobile health technologies, known as mHealth, is increasing and these approaches are contributing to aiding both patients and healthcare professionals in disease management and education. The mHealth solutions allow continuous monitoring of patients, sharing relevant information and data with physicians and other healthcare professionals and accessing education resources to support informed decisions. Thus, if properly used, these tools empower patients and help them to adopt healthier lifestyles. This article aims to give an overview of the influence of circadian rhythms disruption and lifestyle habits in the progression of metabolic diseases while also reviewing some of the mobile applications available to monitor lifestyle behaviors and individual chronobiology. Herein is also described the design and development of the NutriClock system, an mHealth solution developed by our team to monitor these variables.
Collapse
Affiliation(s)
- Iolanda Rosa
- School of Technology and Management, Polytechnic of Leiria, Leiria, Portugal
| | - Marlene Lages
- ciTechCare - Center for Innovative Care and Health Technology, Polytechnic of Leiria, Leiria, Portugal
- Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal
- EPIUnit, Instituto de Saúde Publica, Universidade do Porto, Porto, Portugal
- Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal
| | - Carlos Grilo
- School of Technology and Management, Polytechnic of Leiria, Leiria, Portugal
- Computer Science and Communication Research Center, Polytechnic of Leiria, Leiria, Portugal
| | - Renata Barros
- Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal
- EPIUnit, Instituto de Saúde Publica, Universidade do Porto, Porto, Portugal
- Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal
| | - Maria P. Guarino
- ciTechCare - Center for Innovative Care and Health Technology, Polytechnic of Leiria, Leiria, Portugal
- School of Health Sciences, Polytechnic of Leiria, Leiria, Portugal
- *Correspondence: Maria P. Guarino
| |
Collapse
|
28
|
Moreno JP, Hannay KM, Walch O, Dadabhoy H, Christian J, Puyau M, El-Mubasher A, Bacha F, Grant SR, Park RJ, Cheng P. Estimating circadian phase in elementary school children: leveraging advances in physiologically informed models of circadian entrainment and wearable devices. Sleep 2022; 45:zsac061. [PMID: 35275213 PMCID: PMC9189953 DOI: 10.1093/sleep/zsac061] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/02/2022] [Indexed: 11/12/2022] Open
Abstract
STUDY OBJECTIVES Examine the ability of a physiologically based mathematical model of human circadian rhythms to predict circadian phase, as measured by salivary dim light melatonin onset (DLMO), in children compared to other proxy measurements of circadian phase (bedtime, sleep midpoint, and wake time). METHODS As part of an ongoing clinical trial, a sample of 29 elementary school children (mean age: 7.4 ± .97 years) completed 7 days of wrist actigraphy before a lab visit to assess DLMO. Hourly salivary melatonin samples were collected under dim light conditions (<5 lx). Data from actigraphy were used to generate predictions of circadian phase using both a physiologically based circadian limit cycle oscillator mathematical model (Hannay model), and published regression equations that utilize average sleep onset, midpoint, and offset to predict DLMO. Agreement of proxy predictions with measured DLMO were assessed and compared. RESULTS DLMO predictions using the Hannay model outperformed DLMO predictions based on children's sleep/wake parameters with a Lin's Concordance Correlation Coefficient (LinCCC) of 0.79 compared to 0.41-0.59 for sleep/wake parameters. The mean absolute error was 31 min for the Hannay model compared to 35-38 min for the sleep/wake variables. CONCLUSION Our findings suggest that sleep/wake behaviors were weak proxies of DLMO phase in children, but mathematical models using data collected from wearable data can be used to improve the accuracy of those predictions. Additional research is needed to better adapt these adult models for use in children. CLINICAL TRIAL The i Heart Rhythm Project: Healthy Sleep and Behavioral Rhythms for Obesity Prevention https://clinicaltrials.gov/ct2/show/NCT04445740.
Collapse
Affiliation(s)
- Jennette P Moreno
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Kevin M Hannay
- Department of Mathematics, University of Michigan, Ann Arbor, MI, USA
- Arcascope Inc., Chantilly, VA, USA
| | - Olivia Walch
- Arcascope Inc., Chantilly, VA, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Hafza Dadabhoy
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Jessica Christian
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Maurice Puyau
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Abeer El-Mubasher
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Fida Bacha
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Sarah R Grant
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Rebekah Julie Park
- Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Philip Cheng
- Thomas Roth Sleep Disorders and Research Center, Henry Ford Health System, Detroit, MI, USA
| |
Collapse
|
29
|
Škrlec I, Talapko J, Džijan S, Cesar V, Lazić N, Lepeduš H. The Association between Circadian Clock Gene Polymorphisms and Metabolic Syndrome: A Systematic Review and Meta-Analysis. BIOLOGY 2021; 11:20. [PMID: 35053018 PMCID: PMC8773381 DOI: 10.3390/biology11010020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/11/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023]
Abstract
Metabolic syndrome (MetS) is a combination of cardiovascular risk factors associated with type 2 diabetes, obesity, and cardiovascular diseases. The circadian clock gene polymorphisms are very likely to participate in metabolic syndrome genesis and development. However, research findings of the association between circadian rhythm gene polymorphisms and MetS and its comorbidities are not consistent. In this study, a review of the association of circadian clock gene polymorphisms with overall MetS risk was performed. In addition, a meta-analysis was performed to clarify the association between circadian clock gene polymorphisms and MetS susceptibility based on available data. The PubMed and Scopus databases were searched for studies reporting the association between circadian rhythm gene polymorphisms (ARNTL, BMAL1, CLOCK, CRY, PER, NPAS2, REV-ERBα, REV-ERBβ, and RORα) and MetS, and its comorbidities diabetes, obesity, and hypertension. Thirteen independent studies were analyzed with 17,381 subjects in total. The results revealed that the BMAL1 rs7950226 polymorphism was associated with an increased risk of MetS in the overall population. In contrast, the CLOCK rs1801260 and rs6850524 polymorphisms were not associated with MetS. This study suggests that some circadian rhythm gene polymorphisms might be associated with MetS in different populations and potentially used as predictive biomarkers for MetS.
Collapse
Affiliation(s)
- Ivana Škrlec
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (J.T.); (S.D.); (V.C.); (N.L.); (H.L.)
| | - Jasminka Talapko
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (J.T.); (S.D.); (V.C.); (N.L.); (H.L.)
| | - Snježana Džijan
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (J.T.); (S.D.); (V.C.); (N.L.); (H.L.)
- Genos Ltd., DNA Laboratory, 10000 Zagreb, Croatia
| | - Vera Cesar
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (J.T.); (S.D.); (V.C.); (N.L.); (H.L.)
- Department of Biology, Josip Juraj Strossmayer University of Osijek, Ul. Cara Hadrijana 8/A, 31000 Osijek, Croatia
| | - Nikolina Lazić
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (J.T.); (S.D.); (V.C.); (N.L.); (H.L.)
| | - Hrvoje Lepeduš
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (J.T.); (S.D.); (V.C.); (N.L.); (H.L.)
- Faculty of Humanities and Social Sciences Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
30
|
Qin BW, Zhao L, Lin W. A frequency-amplitude coordinator and its optimal energy consumption for biological oscillators. Nat Commun 2021; 12:5894. [PMID: 34625549 PMCID: PMC8501100 DOI: 10.1038/s41467-021-26182-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/22/2021] [Indexed: 02/08/2023] Open
Abstract
Biorhythm including neuron firing and protein-mRNA interaction are fundamental activities with diffusive effect. Their well-balanced spatiotemporal dynamics are beneficial for healthy sustainability. Therefore, calibrating both anomalous frequency and amplitude of biorhythm prevents physiological dysfunctions or diseases. However, many works were devoted to modulate frequency exclusively whereas amplitude is usually ignored, although both quantities are equally significant for coordinating biological functions and outputs. Especially, a feasible method coordinating the two quantities concurrently and precisely is still lacking. Here, for the first time, we propose a universal approach to design a frequency-amplitude coordinator rigorously via dynamical systems tools. We consider both spatial and temporal information. With a single well-designed coordinator, they can be calibrated to desired levels simultaneously and precisely. The practical usefulness and efficacy of our method are demonstrated in representative neuronal and gene regulatory models. We further reveal its fundamental mechanism and optimal energy consumption providing inspiration for biorhythm regulation in future.
Collapse
Affiliation(s)
- Bo-Wei Qin
- School of Mathematical Sciences, Fudan University, 200433, Shanghai, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China.
| | - Lei Zhao
- School of Mathematical Sciences, Fudan University, 200433, Shanghai, China
- The GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Wei Lin
- School of Mathematical Sciences, Fudan University, 200433, Shanghai, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 200032, Shanghai, China.
- Shanghai Center for Mathematical Sciences, 200438, Shanghai, China.
- Center for Computational Systems Biology of ISTBI, LCNBI, and Research Institute of Intelligent Complex Systems, Fudan University, 200433, Shanghai, China.
| |
Collapse
|
31
|
Osborn LJ, Orabi D, Goudzari M, Sangwan N, Banerjee R, Brown AL, Kadam A, Gromovsky AD, Linga P, Cresci GAM, Mak TD, Willard BB, Claesen J, Brown JM. A Single Human-Relevant Fast Food Meal Rapidly Reorganizes Metabolomic and Transcriptomic Signatures in a Gut Microbiota-Dependent Manner. IMMUNOMETABOLISM 2021; 3:e210029. [PMID: 34804604 PMCID: PMC8601658 DOI: 10.20900/immunometab20210029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND A major contributor to cardiometabolic disease is caloric excess, often a result of consuming low cost, high calorie fast food. Studies have demonstrated the pivotal role of gut microbes contributing to cardiovascular disease in a diet-dependent manner. Given the central contributions of diet and gut microbiota to cardiometabolic disease, we hypothesized that microbial metabolites originating after fast food consumption can elicit acute metabolic responses in the liver. METHODS We gave conventionally raised mice or mice that had their microbiomes depleted with antibiotics a single oral gavage of a liquified fast food meal or liquified control rodent chow meal. After four hours, mice were sacrificed and we used untargeted metabolomics of portal and peripheral blood, 16S rRNA gene sequencing, targeted liver metabolomics, and host liver RNA sequencing to identify novel fast food-derived microbial metabolites and their acute effects on liver function. RESULTS Several candidate microbial metabolites were enriched in portal blood upon fast food feeding, and were essentially absent in antibiotic-treated mice. Strikingly, at four hours post-gavage, fast food consumption resulted in rapid reorganization of the gut microbial community and drastically altered hepatic gene expression. Importantly, diet-driven reshaping of the microbiome and liver transcriptome was dependent on an intact microbial community and not observed in antibiotic ablated animals. CONCLUSIONS Collectively, these data suggest a single fast food meal is sufficient to reshape the gut microbial community in mice, yielding a unique signature of food-derived microbial metabolites. Future studies are in progress to determine the contribution of select metabolites to cardiometabolic disease progression and the translational relevance of these animal studies.
Collapse
Affiliation(s)
- Lucas J. Osborn
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Danny Orabi
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
- Department of General Surgery, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Maryam Goudzari
- Mass Spectrometry Core, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
| | - Naseer Sangwan
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
| | - Rakhee Banerjee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
| | - Amanda L. Brown
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Anagha Kadam
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
| | - Anthony D. Gromovsky
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Pranavi Linga
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
| | - Gail A. M. Cresci
- Department of Inflammation and Immunity, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
| | - Tytus D. Mak
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Belinda B. Willard
- Mass Spectrometry Core, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jan Claesen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - J. Mark Brown
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| |
Collapse
|
32
|
Lu X, Yu S, Chen G, Zheng W, Peng J, Huang X, Chen L. Insight into the roles of melatonin in bone tissue and bone‑related diseases (Review). Int J Mol Med 2021; 47:82. [PMID: 33760138 PMCID: PMC7979260 DOI: 10.3892/ijmm.2021.4915] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 02/17/2021] [Indexed: 12/18/2022] Open
Abstract
Bone‑related diseases comprise a large group of common diseases, including fractures, osteoporosis and osteoarthritis (OA), which affect a large number of individuals, particularly the elderly. The progressive destruction and loss of alveolar bone caused by periodontitis is a specific type of bone loss, which has a high incidence and markedly reduces the quality of life of patients. With the existing methods of prevention and treatment, the incidence and mortality of bone‑related diseases are still gradually increasing, creating a significant financial burden to societies worldwide. To prevent the occurrence of bone‑related diseases, delay their progression or reverse the injuries they cause, new alternative or complementary treatments need to be developed. Melatonin exerts numerous physiological effects, including inducing anti‑inflammatory and antioxidative functions, resetting circadian rhythms and promoting wound healing and tissue regeneration. Melatonin also participates in the health management of bone and cartilage. In the present review, the potential roles of melatonin in the pathogenesis and progression of bone injury, osteoporosis, OA and periodontitis are summarized. Furthermore, the high efficiency and diversity of the physiological regulatory effects of melatonin are highlighted and the potential benefits of the use of melatonin for the clinical prevention and treatment of bone‑related diseases are discussed.
Collapse
Affiliation(s)
- Xiaofeng Lu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Shaoling Yu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Guangjin Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wenhao Zheng
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jinfeng Peng
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiaofei Huang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
33
|
Ma Q, Mo G, Tan Y. Micro RNAs and the biological clock: a target for diseases associated with a loss of circadian regulation. Afr Health Sci 2020; 20:1887-1894. [PMID: 34394254 PMCID: PMC8351835 DOI: 10.4314/ahs.v20i4.46] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Circadian clocks are self-sustaining oscillators that coordinate behavior and physiology over a 24 hour period, achieving time-dependent homeostasis with the external environment. The molecular clocks driving circadian rhythmic changes are based on intertwined transcriptional/translational feedback loops that combine with a range of environmental and metabolic stimuli to generate daily internal programing. Understanding how biological rhythms are generated throughout the body and the reasons for their dysregulation can provide avenues for temporally directed therapeutics. Summary In recent years, microRNAs have been shown to play important roles in the regulation of the circadian clock, particularly in Drosophila, but also in some small animal and human studies. This review will summarize our current understanding of the role of miRNAs during clock regulation, with a particular focus on the control of clock regulated gene expression.
Collapse
Affiliation(s)
- Qianwen Ma
- Gynecology department, Zhenjiang Hospital Affiliated to Nanjing University of Chinese Medicine (Zhenjiang Hospital of Traditional Chinese Medicine), Zhenjiang, China
- Reproductive medicine department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Genlin Mo
- Advanced manufacturing institution, Jiangsu University, Zhenjiang, China
| | - Yong Tan
- Reproductive medicine department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
34
|
Yu Z, Shen J, Li Z, Yao J, Li W, Xue L, Vandenberg LN, Yin D. Obesogenic Effect of Sulfamethoxazole on Drosophila melanogaster with Simultaneous Disturbances on Eclosion Rhythm, Glucolipid Metabolism, and Microbiota. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:5667-5675. [PMID: 32285665 DOI: 10.1021/acs.est.9b07889] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Antibiotics have recently gained attention because they are emerging environmental pollutants with obesogenic properties. In this study, Drosophila melanogaster were exposed to sulfamethoxazole (SMX), a sulfonamide antibiotic, and the effects were measured on circadian rhythm (represented by the eclosion rhythm), lipid metabolism, and microbiota. Circadian rhythm disorder was considered due to its connection with lipid metabolism and microbiota in association with obesity. SMX decreased the proportion of adult flies that eclosed in the morning (AM adults) and increased the proportion of PM adults. Moreover, SMX increased the body weight of PM adults, indicating that SMX exposure caused dysrhythmia in eclosion together with obesity. In measurements of key metabolites and metabolic enzymes, SMX exposure stimulated 3 indices in AM adults and 10 indices in PM adults. In AMP-activated protein kinase and insulin/IGF-1 signaling pathways, SMX upregulated six genes in AM adults and nine genes in PM adults. Finally, microbiota analysis demonstrated that SMX increased the Firmicutes/Bacteroides ratios (F/B) by 79.6- and 5.8-fold compared to concurrent controls in AM and PM adults. Collectively, these results suggest that SMX showed obesogenic effects accompanied with dysrhythmia and disturbances in lipid metabolism and microbiota. Further studies on the intrinsic connection are needed.
Collapse
Affiliation(s)
- Zhenyang Yu
- State Key Laboratory of Pollution Control and Resource Reuse, Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, P. R. China
- Shanghai Institute of Pollution Control and Ecological Security, Shanghai 200092, P. R. China
- Jiaxing Tongji Institute for Environment, Jiaxing, Zhejiang 314051, P. R. China
| | - Jiaying Shen
- State Key Laboratory of Pollution Control and Resource Reuse, Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, P. R. China
| | - Zhuo Li
- State Key Laboratory of Pollution Control and Resource Reuse, Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, P. R. China
| | - Jinmin Yao
- State Key Laboratory of Pollution Control and Resource Reuse, Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, P. R. China
| | - Wenzhe Li
- College of Life Science and Technology, Tongji University, Shanghai 200092, P. R. China
| | - Lei Xue
- College of Life Science and Technology, Tongji University, Shanghai 200092, P. R. China
| | - Laura N Vandenberg
- School of Public Health and Health Sciences, University of Massachusetts - Amherst, Amherst, Massachusetts 01003, United States
| | - Daqiang Yin
- State Key Laboratory of Pollution Control and Resource Reuse, Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, P. R. China
- Shanghai Institute of Pollution Control and Ecological Security, Shanghai 200092, P. R. China
| |
Collapse
|
35
|
Alqaderi H, Tavares M, Al-Mulla F, Al-Ozairi E, Goodson JM. Late bedtime and dental caries incidence in Kuwaiti children: A longitudinal multilevel analysis. Community Dent Oral Epidemiol 2020; 48:181-187. [PMID: 32077126 DOI: 10.1111/cdoe.12523] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Inadequate sleep contributes to several adverse systemic health outcomes due to hormonal and metabolic disorders. The purpose of this study was to determine the effect of bedtime on the development of dental caries and the relationship with salivary ghrelin and leptin in a prospective cohort study of Kuwaiti children. METHODS Data were collected from 5456 10-year-old children in 2012 and repeated in 2014. We selected children from 138 middle schools representing the six governorates of Kuwait. We derived data from oral examinations, self-reported sleep interviews, body and weight measurements, and chemical analysis of whole saliva samples. Leptin and ghrelin were determined by salivary assay in a subset of 744. Two separate analyses were performed. a) Using the entire longitudinal data set (n = 5456), multilevel random intercept analysis was conducted to assess the relationship between reported bedtime and dental caries. b) Using data from a subset of the original sample (n = 744), multiple linear regression analysis was conducted to determine the relationship between dental caries and salivary ghrelin and leptin. The outcome variable was the development of dental caries in children. The independent explanatory variables and confounders were bedtime, sleep duration, salivary ghrelin and leptin; confounders assessed were gingivitis, sex, age and governorate (school location). RESULTS With every additional hour past 8 pm for bedtime, there was a 20% increase in dental caries incidence over two years (B = 0.2, P = .01), adjusting for age, gender, gingivitis and governorate. There was a significant difference in the magnitude of dental caries between the six governorates of Kuwait. Lower levels of salivary leptin and higher levels of salivary ghrelin were associated with increased dental caries, and sleep duration was an effect modifier that negatively affected the relationship between leptin and dental caries (B = -0.09, P < .05) and positively affects the relationship between ghrelin and dental caries (B = 0.07, P < .05). Additionally, there was a significant clustering effect within schools in this cohort. CONCLUSION In a cohort study of Kuwaiti children, late bedtime was associated with increased dental caries incidence. Additionally, dental caries experience increased with higher levels of salivary ghrelin and lower levels of salivary leptin, and sleep duration mediates the relationship between these two biomarkers and dental caries.
Collapse
Affiliation(s)
- Hend Alqaderi
- Department of Oral Health Policy and Epidemiology, Harvard School of Dental Medicine, Boston, MA, USA.,The Forsyth Institute, Cambridge, MA, USA.,Dasman Diabetes Institute, Dasman, Kuwait
| | - Mary Tavares
- Department of Oral Health Policy and Epidemiology, Harvard School of Dental Medicine, Boston, MA, USA.,The Forsyth Institute, Cambridge, MA, USA
| | | | - Ebaa Al-Ozairi
- Dasman Diabetes Institute, Dasman, Kuwait.,Faculty of Medicine, Department of Medicine, Kuwait University, Safat, Kuwait
| | | |
Collapse
|
36
|
Škrlec I, Milić J, Cilenšek I, Petrovič D, Wagner J, Peterlin B. Circadian clock genes and myocardial infarction in patients with type 2 diabetes mellitus. Gene 2019; 701:98-103. [PMID: 30905809 DOI: 10.1016/j.gene.2019.03.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 02/07/2023]
Abstract
Disruption of circadian clock may trigger the onset of diabetes mellitus and myocardial infarction. Type 2 diabetes mellitus (T2DM) is well-known risk factors for cardiovascular diseases and myocardial infarction. We performed a case-control study, where we explored the possible association between single nucleotide polymorphisms in three circadian rhythm genes (ARNTL, CLOCK, and PER2) and myocardial infarction in 657 patients with T2DM. The study group consisted of 231 patients with myocardial infarction and T2DM and a control group of 426 T2DM patients. We hypothesized that variations in the circadian rhythm genes in patients with T2DM could be an additional risk factor for myocardial infarction. The statistically significant difference was found in allelic (p = 1.1 × 10-5) and genotype distribution (p = 1.42 × 10-4) between two groups of the rs12363415 at the ARNTL gene locus. We provide evidence that genetic variability in the ARNTL gene might be associated with myocardial infarction in patients with T2DM.
Collapse
Affiliation(s)
- Ivana Škrlec
- Department of Biology, Faculty of Dental Medicine and Health, J. J. Strossmayer University of Osijek, Croatia; Faculty of Medicine, J. J. Strossmayer University of Osijek, Croatia.
| | - Jakov Milić
- Faculty of Medicine, J. J. Strossmayer University of Osijek, Croatia
| | - Ines Cilenšek
- Institute of Histology and Embryology, Faculty of Medicine, University Ljubljana, Ljubljana, Slovenia
| | - Daniel Petrovič
- Institute of Histology and Embryology, Faculty of Medicine, University Ljubljana, Ljubljana, Slovenia
| | - Jasenka Wagner
- Faculty of Medicine, J. J. Strossmayer University of Osijek, Croatia
| | - Borut Peterlin
- Clinical Institute of Medical Genetics, University Medical Center Ljubljana, Slovenia
| |
Collapse
|
37
|
Hieke ASC, Hubert SM, Athrey G. Circadian disruption and divergent microbiota acquisition under extended photoperiod regimens in chicken. PeerJ 2019; 7:e6592. [PMID: 30886778 PMCID: PMC6421066 DOI: 10.7717/peerj.6592] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 02/09/2019] [Indexed: 12/13/2022] Open
Abstract
The gut microbiota is crucial for metabolic homeostasis, immunity, growth and overall health, and it is recognized that early-life microbiota acquisition is a pivotal event for later-life health. Recent studies show that gut microbiota diversity and functional activity are synchronized with the host circadian rhythms in healthy individuals, and circadian disruption elicits dysbiosis in mammalian models. However, no studies have determined the associations between circadian disruption in early life, microbiota colonization, and the consequences for microbiota structure in birds. Chickens, as a major source of protein around the world, are one of the most important agricultural species, and their gut and metabolic health are significant concerns. The poultry industry routinely employs extended photoperiods (>18 h light) as a management tool, and their impacts on the chicken circadian, its role in gut microbiota acquisition in early life (first 3 weeks of life), and consequences for later life microbiota structure remain unknown. In this study, the objectives were to (a) characterize circadian activity under two different light regimes in layer chicken (12/12 h′ Light/Dark (LD) and 23/1 h LD), (b) characterize gut microbiota acquisition and composition in the first 4 weeks of life, (c) determine if gut microbiota oscillate in synchrony with the host circadian rhythm, and (d) to determine if fecal microbiota is representative of cecal microbiota in early life. Expression of clock genes (clock, bmal1, and per2) was assayed, and fecal and cecal microbiotas were characterized using 16S rRNA gene amplicon analyses from birds raised under two photoperiod treatments. Chickens raised under 12/12 LD photoperiods exhibited rhythmic clock gene activity, which was absent in birds raised under the extended (23/1 LD) photoperiod. There was differential microbiota acquisition under different photoperiod regimes in newly hatched chicks. Gut microbiota members showed a similar oscillating pattern as the host, but this association was not as strong as found in mammals. Finally, the fecal microbiota was found to be not representative of cecal microbiota membership and structure in young birds. This is one of the first studies to demonstrate the use of photoperiods to modulate microbiota acquisition in newly hatched chicks, and show their potential as a tool to promote the colonization of beneficial microorganisms.
Collapse
Affiliation(s)
| | | | - Giridhar Athrey
- Poultry Science Department, Texas A&M University, College Station, TX, USA.,Faculty of Ecology and Evolutionary Biology, Texas A&M University, College Station, TX, USA
| |
Collapse
|
38
|
Virto L, Cano P, Jiménez-Ortega V, Fernández-Mateos P, González J, Haugen HJ, Esquifino AI, Sanz M. Melatonin as adjunctive therapy in the treatment of periodontitis associated with obesity. J Clin Periodontol 2018; 45:1336-1346. [DOI: 10.1111/jcpe.13013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/21/2018] [Accepted: 09/16/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Leire Virto
- Etiology and Therapy of Periodontal Diseases (ETEP) Research Group; University Complutense; Madrid Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Madrid Spain
| | - Pilar Cano
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Madrid Spain
- Department of Biochemical and Molecular Biology (Sección Departamental, Faculty of Medicine); University Complutense; Madrid Spain
| | - Vanesa Jiménez-Ortega
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Madrid Spain
- Department of Biochemical and Molecular Biology (Sección Departamental, Faculty of Medicine); University Complutense; Madrid Spain
| | - Pilar Fernández-Mateos
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Madrid Spain
- Department of Cellular Biology; Faculty of Medicine; University Complutense; Madrid Spain
| | - Jerián González
- Department of Clinical Dental Specialties; Faculty of Odontology; University Complutense; Madrid Spain
| | - Håvard J. Haugen
- Department of Biomaterials; Institute for Clinical Dentistry; University of Oslo; Oslo Norway
| | - Ana Isabel Esquifino
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Madrid Spain
- Department of Biochemical and Molecular Biology (Sección Departamental, Faculty of Medicine); University Complutense; Madrid Spain
| | - Mariano Sanz
- Etiology and Therapy of Periodontal Diseases (ETEP) Research Group; University Complutense; Madrid Spain
- Department of Clinical Dental Specialties; Faculty of Odontology; University Complutense; Madrid Spain
| |
Collapse
|
39
|
Virto L, Haugen HJ, Fernández-Mateos P, Cano P, González J, Jiménez-Ortega V, Esquifino AI, Sanz M. Melatonin expression in periodontitis and obesity: An experimental in-vivo investigation. J Periodontal Res 2018; 53:825-831. [PMID: 29900537 DOI: 10.1111/jre.12571] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND OBJECTIVE Melatonin deficiency has been associated with obesity and systemic inflammation. This study aims to evaluate whether melatonin could interfere with the mechanisms of co-morbidity linking obesity and periodontitis. MATERIAL AND METHODS Twenty-eight male Wistar rats were randomly divided in 4 groups: control group (Con) (fed with standard diet); high-fat diet group (HFD) (fed with a diet containing 35.2% fat); Con group with induced periodontitis (Con-Perio) and HFD group with induced periodontitis (HFD-Perio). To induce periodontitis, the method of oral gavages with Porphyromonas gingivalis ATCC W83K1 and Fusobacterium nucleatum DMSZ 20482 was used. Circulating melatonin levels were analyzed by multiplex immunoassays. Periodontitis was assessed by alveolar bone loss (micro-computed tomography and histology) and by surrogate inflammatory outcomes (periodontal pocket depth, modified gingival index and plaque dental index). RESULTS Plasma melatonin levels were significantly decreased (P < .05) in the obese rats with periodontitis when compared with controls or with either obese or periodontitis rats. Alveolar bone loss increased 27.71% (2.28 µm) in HFD-Perio group compared with the Con group. The histological analysis showed marked periodontal tissue destruction with osteoclast activity, particularly in the HFD-Perio group. A significant negative correlation (P < .05) was found between periodontal pocket depth, modified gingival index and circulating melatonin levels. CONCLUSION Obese and periodontitis demonstrated significantly lower melatonin concentrations when compared with controls, but in obese rats with periodontitis these concentrations were even significantly lower when compared with either periodontitis or obese rats. These results may indicate that melatonin deficiency could be a key mechanism explaining the co-morbidity effect in the association between obesity and periodontitis.
Collapse
Affiliation(s)
- L Virto
- Etiology and Therapy of Periodontal Diseases (ETEP) Research Group, University Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - H J Haugen
- Department of Biomaterials, Institute for Clinical Dentistry, University of Oslo, Oslo, Norway
| | - P Fernández-Mateos
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
- Faculty of Medicine, Department of Cellular Biology, University Complutense, Madrid, Spain
| | - P Cano
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
- Faculty of Medicine, Department of Biochemistry and Molecular Biology III, University Complutense, Madrid, Spain
| | - J González
- Etiology and Therapy of Periodontal Diseases (ETEP) Research Group, University Complutense, Madrid, Spain
| | - V Jiménez-Ortega
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
- Faculty of Medicine, Department of Biochemistry and Molecular Biology III, University Complutense, Madrid, Spain
| | - A I Esquifino
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
- Faculty of Medicine, Department of Biochemistry and Molecular Biology III, University Complutense, Madrid, Spain
| | - M Sanz
- Etiology and Therapy of Periodontal Diseases (ETEP) Research Group, University Complutense, Madrid, Spain
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW In addition to their effects on glycemic control, two specific classes of relatively new anti-diabetic drugs, namely the sodium glucose co-transporter-2 inhibitors (SGLT2i) and glucagon-like peptide-1 receptor agonists (GLP-1RA) have demonstrated reduced rates of major adverse cardiovascular events (MACE) in subjects with type 2 diabetes (T2D) at high risk for cardiovascular disease (CVD). This review summarizes recent experimental results that inform putative molecular mechanisms underlying these benefits. RECENT FINDINGS SGLT2i and GLP-1RA exert cardiovascular effects by targeting in both common and distinctive ways (A) several mediators of macro- and microvascular pathophysiology: namely (A1) inflammation and atherogenesis, (A2) oxidative stress-induced endothelial dysfunction, (A3) vascular smooth muscle cell reactive oxygen species (ROS) production and proliferation, and (A4) thrombosis. These agents also exhibit (B) hemodynamic effects through modulation of (B1) natriuresis/diuresis and (B2) the renin-angiotensin-aldosterone system. This review highlights that while GLP-1RA exert direct effects on vascular (endothelial and smooth muscle) cells, the effects of SGLT2i appear to include the activation of signaling pathways that prevent adverse vascular remodeling. Both SGLT2i and GLP-1RA confer hemodynamic effects that counter adverse cardiac remodeling.
Collapse
Affiliation(s)
- Dorrin Zarrin Khat
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Mansoor Husain
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
- Department of Medicine, University of Toronto, Toronto, Canada.
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.
- Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, Canada.
- Ted Rogers Centre for Heart Research, University Health Network, Toronto, Canada.
- Peter Munk Cardiac Centre, University Health Network, Toronto, Canada.
| |
Collapse
|
41
|
Ren DL, Zhang JL, Yang LQ, Wang XB, Wang ZY, Huang DF, Tian C, Hu B. Circadian genes period1b and period2 differentially regulate inflammatory responses in zebrafish. FISH & SHELLFISH IMMUNOLOGY 2018; 77:139-146. [PMID: 29605504 DOI: 10.1016/j.fsi.2018.03.048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 06/08/2023]
Abstract
The circadian clock has been shown to regulate various immune processes in different animals. Our previous report demonstrated that the innate immune responses in zebrafish show significant rhythmicity that could be regulated by melatonin. Here, we used diurnal zebrafish to determine the role of circadian genes in the inflammatory responses. Our results indicate that circadian genes exhibit rhythmic oscillations in zebrafish leukocytes, and mutations of the clock genes period1b (per1b) and period2 (per2) considerably affect these oscillations. Using a wounded zebrafish inflammation model, we found that under constant dark conditions (DD), the expression of pro-inflammatory cytokines is significantly downregulated in per1b gene mutant zebrafish and significantly upregulated in the per2 gene mutant zebrafish. Furthermore, using real-time imaging technology, we found that the per1b gene markedly disturbs the rhythmic recruitment of neutrophils toward the injury, whereas the per2 gene does not show a significant effect. Taken together, our results reveal differential functions of the circadian genes per1b and per2 in the inflammatory responses, serving as evidence that circadian rhythms play a vital role in immune processes.
Collapse
Affiliation(s)
- Da-Long Ren
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, No.96 Jinzhai Road, Hefei, Anhui Province, 230026, PR China.
| | - Jun-Long Zhang
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, No.96 Jinzhai Road, Hefei, Anhui Province, 230026, PR China
| | - Lei-Qing Yang
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, No.96 Jinzhai Road, Hefei, Anhui Province, 230026, PR China
| | - Xiao-Bo Wang
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, No.96 Jinzhai Road, Hefei, Anhui Province, 230026, PR China
| | - Zong-Yi Wang
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, No.96 Jinzhai Road, Hefei, Anhui Province, 230026, PR China
| | - Deng-Feng Huang
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, No.96 Jinzhai Road, Hefei, Anhui Province, 230026, PR China
| | - Chen Tian
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, No.96 Jinzhai Road, Hefei, Anhui Province, 230026, PR China
| | - Bing Hu
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, No.96 Jinzhai Road, Hefei, Anhui Province, 230026, PR China.
| |
Collapse
|
42
|
Altered Circadian Timing System-Mediated Non-Dipping Pattern of Blood Pressure and Associated Cardiovascular Disorders in Metabolic and Kidney Diseases. Int J Mol Sci 2018; 19:ijms19020400. [PMID: 29385702 PMCID: PMC5855622 DOI: 10.3390/ijms19020400] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/12/2018] [Accepted: 01/20/2018] [Indexed: 12/15/2022] Open
Abstract
The morning surge in blood pressure (BP) coincides with increased cardiovascular (CV) events. This strongly suggests that an altered circadian rhythm of BP plays a crucial role in the development of CV disease (CVD). A disrupted circadian rhythm of BP, such as the non-dipping type of hypertension (i.e., absence of nocturnal BP decline), is frequently observed in metabolic disorders and chronic kidney disease (CKD). The circadian timing system, controlled by the central clock in the suprachiasmatic nucleus of the hypothalamus and/or by peripheral clocks in the heart, vasculature, and kidneys, modulates the 24 h oscillation of BP. However, little information is available regarding the molecular and cellular mechanisms of an altered circadian timing system-mediated disrupted dipping pattern of BP in metabolic disorders and CKD that can lead to the development of CV events. A more thorough understanding of this pathogenesis could provide novel therapeutic strategies for the management of CVD. This short review will address our and others' recent findings on the molecular mechanisms that may affect the dipping pattern of BP in metabolic dysfunction and kidney disease and its association with CV disorders.
Collapse
|
43
|
Hemoglobin A1c and C-reactive protein are independently associated with blunted nocturnal blood pressure dipping in obesity-related prediabetes. Hypertens Res 2017; 41:33-38. [PMID: 29070829 DOI: 10.1038/hr.2017.82] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/11/2017] [Accepted: 06/07/2017] [Indexed: 12/12/2022]
Abstract
Blunted nocturnal dipping in blood pressure (BP) is associated with increased cardiovascular disease (CVD) risk in middle-aged/older adults. The prevalence of blunted nocturnal BP dipping is higher in persons with obesity and diabetes, conditions that are also associated with elevated aortic stiffness and inflammation. Therefore, we hypothesized that elevated glycemia, inflammation and aortic stiffness would be inversely associated with the magnitude of nocturnal systolic BP dipping among middle-aged/older adults with obesity at high CVD risk. Twenty-four hour ambulatory BP monitoring, aortic stiffness (carotid-femoral pulse wave velocity, CF-PWV), hemoglobin A1c (HbA1c) and inflammation (C-reactive protein, CRP) were measured in 86 middle-aged/older adults with obesity and at least one other CVD risk factor (age 40-74 years; 34 male/52 female; body mass index=36.7±0.5 kg m-2; HbA1c=5.7±0.04%). In the entire cohort, CRP (β=0.40±0.20, P=0.04), but not HbA1c or CF-PWV was independently associated with systolic BP dipping percent (Model R2=0.07, P=0.12). In stratified (that is, presence or absence of prediabetes) multiple linear regression analysis, HbA1c (β=6.24±2.6, P=0.02) and CRP (β=0.57±0.2, P=0.01), but not CF-PWV (β=0.14± 2.6, P=0.74), were independently associated with systolic BP dipping percent (Model R2=0.32, P<0.01) in obese adults with prediabetes but were absent in obese adults without prediabetes (Model R2=0.01 P=0.95). However, nocturnal systolic BP dipping percent (P=0.65), CF-PWV (P=0.68) and CRP (P=0.59) were similar between participants with and without prediabetes. These data suggest that impaired long-term glycemic control and higher inflammation may contribute partly to blunted BP dipping in middle-aged/older adults with obesity-related prediabetes.
Collapse
|
44
|
Cold-sensing TRPM8 channel participates in circadian control of the brown adipose tissue. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2415-2427. [PMID: 28943398 DOI: 10.1016/j.bbamcr.2017.09.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/18/2017] [Accepted: 09/19/2017] [Indexed: 01/11/2023]
Abstract
Transient receptor potential (TRP) channels are known to regulate energy metabolism, and TRPM8 has become an interesting player in this context. Here we demonstrate the role of the cold sensor TRPM8 in the regulation of clock gene and clock controlled genes in brown adipose tissue (BAT). We investigated TrpM8 temporal profile in the eyes, suprachiasmatic nucleus and BAT; only BAT showed temporal variation of TrpM8 transcripts. Eyes from mice lacking TRPM8 lost the temporal profile of Per1 in LD cycle. This alteration in the ocular circadian physiology may explain the delay in the onset of locomotor activity in response to light pulse, as compared to wild type animals (WT). Brown adipocytes from TrpM8 KO mice exhibited a larger multilocularity in comparison to WT or TrpV1 KO mice. In addition, Ucp1 and UCP1 expression was significantly reduced in TrpM8 KO mice in comparison to WT mice. Regarding circadian components, the expression of Per1, Per2, Bmal1, Pparα, and Pparβ oscillated in WT mice kept in LD, whereas in the absence of TRPM8 the expression of clock genes was reduced in amplitude and lack temporal oscillation. Thus, our results reveal new roles for TRPM8 channel: it participates in the regulation of clock and clock-controlled genes in the eyes and BAT, and in BAT thermogenesis. Since disruption of the clock machinery has been associated with many metabolic disorders, the pharmacological modulation of TRPM8 channel may become a promising therapeutic target to counterbalance weight gain, through increased thermogenesis, energy expenditure, and clock gene activation.
Collapse
|
45
|
Nio Y, Hotta N, Maruyama M, Hamagami K, Nagi T, Funata M, Sakamoto J, Nakakariya M, Amano N, Okawa T, Arikawa Y, Sasaki S, Okuda S, Kasai S, Habata Y, Nagisa Y. A Selective Bombesin Receptor Subtype 3 Agonist Promotes Weight Loss in Male Diet-Induced-Obese Rats With Circadian Rhythm Change. Endocrinology 2017; 158:1298-1313. [PMID: 28324017 DOI: 10.1210/en.2016-1825] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 03/08/2017] [Indexed: 12/17/2022]
Abstract
Bombesin receptor subtype 3 (BRS-3) is an orphan G protein-coupled receptor. Based on the obese phenotype of male BRS-3-deficient mice, BRS-3 has been considered an attractive target for obesity treatment. Here, we developed a selective BRS-3 agonist (compound-A) and evaluated its antiobesity effects. Compound-A showed anorectic effects and enhanced energy expenditure in diet-induced-obese (DIO)-F344 rats. Moreover, repeated oral administration of compound-A for 7 days resulted in a significant body weight reduction in DIO-F344 rats. We also evaluated compound-A for cardiovascular side effects using telemeterized Sprague-Dawley (SD) rats. Oral administration of compound-A resulted in transient blood pressure increases in SD rats. To investigate the underlying mechanisms of BRS-3 agonist effects, we focused on the suprachiasmatic nucleus (SCN), the main control center of circadian rhythms in the hypothalamus, also regulating sympathetic nervous system. Compound-A significantly increased the messenger RNA expression of Brs-3, c-fos, and circadian rhythm genes in SCN of DIO-F344 rats. Because SCN also controls the hypothalamic-pituitary-adrenal (HPA) axis, we evaluated the relationship between BRS-3 and the HPA axis. Oral administration of compound-A caused a significant increase of plasma corticosterone levels in DIO-F344 rats. On this basis, energy expenditure enhancement by compound-A may be due to a circadian rhythm change in central and peripheral tissues, enhancement of peripheral lipid metabolism, and stimulation of the sympathetic nervous system. Furthermore, the blood pressure increase by compound-A could be associated with sympathetic nervous system stimulation via SCN and elevation of plasma corticosterone levels through activation of the HPA axis.
Collapse
Affiliation(s)
- Yasunori Nio
- Extra Value Generation & General Medicine Drug Discovery Unit, Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Natsu Hotta
- Cardiovascular and Metabolic Drug Discovery Unit, Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Minoru Maruyama
- Cardiovascular and Metabolic Drug Discovery Unit, Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Kenichi Hamagami
- Cardiovascular and Metabolic Drug Discovery Unit, Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Toshimi Nagi
- Central Nervous System Drug Discovery Unit, Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Masaaki Funata
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Junichi Sakamoto
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Masanori Nakakariya
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Nobuyuki Amano
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Tomohiro Okawa
- Central Nervous System Drug Discovery Unit, Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Yasuyoshi Arikawa
- Central Nervous System Drug Discovery Unit, Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Shinobu Sasaki
- Medicinal Chemistry Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Shoki Okuda
- Cardiovascular and Metabolic Drug Discovery Unit, Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Shizuo Kasai
- Cardiovascular and Metabolic Drug Discovery Unit, Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Yugo Habata
- Foods & Nutrients, Yamanashi Gakuin Junior College, Kofu, Yamanashi 400-8575, Japan
| | - Yasutaka Nagisa
- CVM Marketing Japan Pharma Business Unit, Takeda Pharmaceutical Company Ltd, Chuo-ku, Tokyo 103-8686, Japan
| |
Collapse
|
46
|
Moraes MN, Mezzalira N, de Assis LVM, Menaker M, Guler A, Castrucci AML. TRPV1 participates in the activation of clock molecular machinery in the brown adipose tissue in response to light-dark cycle. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:324-335. [DOI: 10.1016/j.bbamcr.2016.11.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 11/07/2016] [Accepted: 11/12/2016] [Indexed: 02/02/2023]
|
47
|
Cardioprotective effects of SGLT2 inhibitors are possibly associated with normalization of the circadian rhythm of blood pressure. Hypertens Res 2017; 40:535-540. [DOI: 10.1038/hr.2016.193] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 11/25/2016] [Accepted: 11/28/2016] [Indexed: 12/11/2022]
|
48
|
Hardeland R. Melatonin and the pathologies of weakened or dysregulated circadian oscillators. J Pineal Res 2017; 62. [PMID: 27763686 DOI: 10.1111/jpi.12377] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/18/2016] [Indexed: 12/20/2022]
Abstract
Dynamic aspects of melatonin's actions merit increasing future attention. This concerns particularly entirely different effects in senescent, weakened oscillators and in dysregulated oscillators of cancer cells that may be epigenetically blocked. This is especially obvious in the case of sirtuin 1, which is upregulated by melatonin in aged tissues, but strongly downregulated in several cancer cells. These findings are not at all controversial, but are explained on the basis of divergent changes in weakened and dysregulated oscillators. Similar findings can be expected to occur in other accessory oscillator components that are modulated by melatonin, among them several transcription factors and metabolic sensors. Another cause of opposite effects concerns differences between nocturnally active laboratory rodents and the diurnally active human. This should be more thoroughly considered in the field of metabolic syndrome and related pathologies, especially with regard to type 2 diabetes and other aspects of insulin resistance. Melatonin was reported to impair glucose tolerance in humans, especially in carriers of the risk allele of the MT2 receptor gene, MTNR1B, that contains the SNP rs10830963. These findings contrast with numerous reports on improvements of glucose tolerance in preclinical studies. However, the relationship between melatonin and insulin may be more complex, as indicated by loss-of-function mutants of the MT2 receptor that are also prodiabetic, by the age-dependent time course of risk allele overexpression, by progressive reduction in circadian amplitudes and melatonin secretion, which are aggravated in diabetes. By supporting high-amplitude rhythms, melatonin may be beneficial in preventing or delaying diabetes.
Collapse
Affiliation(s)
- Rüdiger Hardeland
- Johann Friedrich Blumenbach Institute of Zoology and Anthropology, University of Göttingen, Göttingen, Germany
| |
Collapse
|
49
|
Circadian Rhythm Shapes the Gut Microbiota Affecting Host Radiosensitivity. Int J Mol Sci 2016; 17:ijms17111786. [PMID: 27792172 PMCID: PMC5133787 DOI: 10.3390/ijms17111786] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 10/12/2016] [Accepted: 10/19/2016] [Indexed: 12/11/2022] Open
Abstract
Modern lifestyles, such as shift work, nocturnal social activities, and jet lag, disturb the circadian rhythm. The interaction between mammals and the co-evolved intestinal microbiota modulates host physiopathological processes. Radiotherapy is a cornerstone of modern management of malignancies; however, it was previously unknown whether circadian rhythm disorder impairs prognosis after radiotherapy. To investigate the effect of circadian rhythm on radiotherapy, C57BL/6 mice were housed in different dark/light cycles, and their intestinal bacterial compositions were compared using high throughput sequencing. The survival rate, body weight, and food intake of mice in diverse cohorts were measured following irradiation exposure. Finally, the enteric bacterial composition of irradiated mice that experienced different dark/light cycles was assessed using 16S RNA sequencing. Intriguingly, mice housed in aberrant light cycles harbored a reduction of observed intestinal bacterial species and shifts of gut bacterial composition compared with those of the mice kept under 12 h dark/12 h light cycles, resulting in a decrease of host radioresistance. Moreover, the alteration of enteric bacterial composition of mice in different groups was dissimilar. Our findings provide novel insights into the effects of biological clocks on the gut bacterial composition, and underpin that the circadian rhythm influences the prognosis of patients after radiotherapy in a preclinical setting.
Collapse
|
50
|
Middlemiss JE, McEniery CM. Feeling the pressure: (patho) physiological mechanisms of weight gain and weight loss in humans. Hypertens Res 2016; 40:226-236. [PMID: 27760999 DOI: 10.1038/hr.2016.142] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/30/2016] [Accepted: 09/12/2016] [Indexed: 12/12/2022]
Abstract
Obesity is an ongoing global epidemic and has adverse consequences for cardiovascular health. Obesity is often associated with hypertension, which is, itself, a common condition and an important cause of morbidity and mortality worldwide. Although animal models of obesity have provided extensive data on the links between obesity and hypertension, a greater understanding of the pathways linking obesity and hypertension in humans is likely to assist translation of animal data, and may, itself, identify important treatment strategies. Ultimately, this could have a substantial impact on human health, both at an individual and population level. The current review will focus specifically on studies of experimental weight gain and weight loss in humans and the following key areas, which are strongly related to blood pressure: cardiovascular function, autonomic nervous system function, metabolic function and the impact of cardiorespiratory fitness.
Collapse
Affiliation(s)
- Jessica E Middlemiss
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Carmel M McEniery
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| |
Collapse
|