1
|
Yang S, Ye Z, Chen L, Zhou X, Li W, Cheng F. Circadian Clock Gene Bmal1: A Molecular Bridge from AKI to CKD. Biomolecules 2025; 15:77. [PMID: 39858471 PMCID: PMC11762869 DOI: 10.3390/biom15010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/05/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) represent two frequently observed clinical conditions. AKI is characterized by an abrupt decrease in glomerular filtration rate (GFR), generally associated with elevated serum creatinine (sCr), blood urea nitrogen (BUN), and electrolyte imbalances. This condition usually persists for approximately a week, causing a transient reduction in kidney function. If these abnormalities continue beyond 90 days, the condition is redefined as chronic kidney disease (CKD) or may advance to end-stage renal disease (ESRD). Recent research increasingly indicates that maladaptive repair mechanisms after AKI significantly contribute to the development of CKD. Thus, implementing early interventions to halt the progression from AKI to CKD has the potential to markedly improve patient outcomes. Although considerable research has been conducted, the exact mechanisms linking AKI to CKD are complex, and effective treatments remain limited. Kidney function is influenced by circadian rhythms, with the circadian gene Bmal1 being vital in managing these cycles. Recent research indicates that Bmal1 is significantly involved in the progression of both AKI and CKD. In this study, we conducted a retrospective analysis of Bmal1's role in AKI and CKD, reviewed recent research advancements, and investigated how Bmal1 influences the pathological mechanisms underlying the progression from AKI to CKD. Additionally, we highlighted gaps in the existing research and examined the potential of Bmal1 as a therapeutic target in kidney disease management. This work aims to provide meaningful insights for future studies on the role of the circadian gene Bmal1 in the transition from AKI to CKD, with the goal of identifying therapeutic approaches to mitigate kidney disease progression.
Collapse
Affiliation(s)
- Songyuan Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (S.Y.); (Z.Y.); (L.C.); (X.Z.)
| | - Zehua Ye
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (S.Y.); (Z.Y.); (L.C.); (X.Z.)
| | - Lijia Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (S.Y.); (Z.Y.); (L.C.); (X.Z.)
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (S.Y.); (Z.Y.); (L.C.); (X.Z.)
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (S.Y.); (Z.Y.); (L.C.); (X.Z.)
| |
Collapse
|
2
|
Aten S, Ramirez-Plascencia O, Blake C, Holder G, Fishbein E, Vieth A, Zarghani-Shiraz A, Keister E, Howe S, Appo A, Palmer B, Mahoney CE. A time for sex: circadian regulation of mammalian sexual and reproductive function. Front Neurosci 2025; 18:1516767. [PMID: 39834701 PMCID: PMC11743455 DOI: 10.3389/fnins.2024.1516767] [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: 10/24/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025] Open
Abstract
The circadian clock regulates physiological and biochemical processes in nearly every species. Sexual and reproductive behaviors are two processes controlled by the circadian timing system. Evidence supporting the importance of proper clock function on fertility comes from several lines of work demonstrating that misalignment of biological rhythms or disrupted function of the body's master clock, such as occurs from repeated shift work or chronic jet lag, negatively impacts reproduction by interfering with both male and female fertility. Along these lines, dysregulation of clock genes leads to impairments in fertility within mammals, and disruption of circadian clock timing negatively impacts sex hormone levels and semen quality in males, and it leads to ovulatory deficiencies in females. Here, we review the current understanding of the circadian modulation of both male and female reproductive hormones-from animal models to humans. Further, we discuss neural circuits within the hypothalamus that may regulate circadian changes in mammalian sexual behavior and reproduction, and we explore how knowledge of such circuits in animal models may help to improve human sexual function, fertility, and reproduction.
Collapse
Affiliation(s)
- Sydney Aten
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Oscar Ramirez-Plascencia
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Chiara Blake
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Behavioral Neuroscience, College of Science, Northeastern University, Boston, MA, United States
| | - Gabriel Holder
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Behavioral Neuroscience, College of Science, Northeastern University, Boston, MA, United States
| | - Emma Fishbein
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Department of Biology, School of Arts and Sciences, Tufts University, Medford, MA, United States
| | - Adam Vieth
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Behavioral Neuroscience, College of Science, Northeastern University, Boston, MA, United States
| | - Arman Zarghani-Shiraz
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Department of Psychological and Brain Sciences, College of Arts and Sciences, Boston University, Boston, MA, United States
| | - Evan Keister
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Behavioral Neuroscience, College of Science, Northeastern University, Boston, MA, United States
| | - Shivani Howe
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Behavioral Neuroscience, College of Science, Northeastern University, Boston, MA, United States
| | - Ashley Appo
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Behavioral Neuroscience, College of Science, Northeastern University, Boston, MA, United States
| | - Beatrice Palmer
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Carrie E. Mahoney
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
3
|
Xu W, Li L, Cao Z, Ye J, Gu X. Circadian Rhythms and Lung Cancer in the Context of Aging: A Review of Current Evidence. Aging Dis 2025:AD.2024.1188. [PMID: 39812541 DOI: 10.14336/ad.2024.1188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025] Open
Abstract
Circadian rhythm is the internal homeostatic physiological clock that regulates the 24-hour sleep/wake cycle. This biological clock helps to adapt to environmental changes such as light, dark, temperature, and behaviors. Aging, on the other hand, is a process of physiological changes that results in a progressive decline in cells, tissues, and other vital systems of the body. Both aging and the circadian clock are highly interlinked phenomena with a bidirectional relationship. The process of aging leads to circadian disruptions while dysfunctional circadian rhythms promote age-related complications. Both processes involve diverse physiological, molecular, and cellular changes such as modifications in the DNA repair mechanisms, mechanisms, ROS generation, apoptosis, and cell proliferation. This review aims to examine the role of aging and circadian rhythms in the context of lung cancer. This will also review the existing literature on the role of circadian disruptions in the process of aging and vice versa. Various molecular pathways and genes such as BMAL1, SIRT1, HLF, and PER1 and their implications in aging, circadian rhythms, and lung cancer will also be discussed.
Collapse
Affiliation(s)
- Wenhui Xu
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Lei Li
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Zhendong Cao
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Jinghong Ye
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Xuyu Gu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
4
|
Zhuang Y, Zhang Y, Liu C, Zhong Y. Interplay Between the Circadian Clock and Sirtuins. Int J Mol Sci 2024; 25:11469. [PMID: 39519022 PMCID: PMC11545976 DOI: 10.3390/ijms252111469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
The circadian clock is an autonomous timekeeping system evolved by organisms to adapt to external changes, regulating a variety of important physiological and behavioral processes. Recent studies have shown that the sirtuin family of histone deacetylases is involved in regulating the expression of clock genes and plays an important role in maintaining the normal rhythm of clock gene expression and behavior. Moreover, sirtuins are regulated directly or indirectly by the circadian clock system. The mutual regulation between the circadian clock and sirtuins is likely involved in a variety of signal transduction and metabolism processes. In this review, we discuss the molecular mechanisms and research progress on the intertwined relationship between the circadian clock and sirtuins, mainly in mammals, highlighting sirtuins as molecular links between metabolic control and circadian rhythms and offering our perspectives on future developments in the field.
Collapse
Affiliation(s)
- Yan Zhuang
- School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Yantong Zhang
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Chao Liu
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Yingbin Zhong
- School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| |
Collapse
|
5
|
Duong HA, Baba K, DeBruyne JP, Davidson AJ, Ehlen C, Powell M, Tosini G. Environmental circadian disruption re-writes liver circadian proteomes. Nat Commun 2024; 15:5537. [PMID: 38956413 PMCID: PMC11220080 DOI: 10.1038/s41467-024-49852-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024] Open
Abstract
Circadian gene expression is fundamental to the establishment and functions of the circadian clock, a cell-autonomous and evolutionary-conserved timing system. Yet, how it is affected by environmental-circadian disruption (ECD) such as shiftwork and jetlag are ill-defined. Here, we provided a comprehensive and comparative description of male liver circadian gene expression, encompassing transcriptomes, whole-cell proteomes and nuclear proteomes, under normal and after ECD conditions. Under both conditions, post-translation, rather than transcription, is the dominant contributor to circadian functional outputs. After ECD, post-transcriptional and post-translational processes are the major contributors to whole-cell or nuclear circadian proteome, respectively. Furthermore, ECD re-writes the rhythmicity of 64% transcriptome, 98% whole-cell proteome and 95% nuclear proteome. The re-writing, which is associated with changes of circadian regulatory cis-elements, RNA-processing and protein localization, diminishes circadian regulation of fat and carbohydrate metabolism and persists after one week of ECD-recovery.
Collapse
Affiliation(s)
- Hao A Duong
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, 30310, USA.
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA.
| | - Kenkichi Baba
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Jason P DeBruyne
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Alec J Davidson
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Christopher Ehlen
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Michael Powell
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Gianluca Tosini
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| |
Collapse
|
6
|
Naveed M, Chao OY, Hill JW, Yang YM, Huston JP, Cao R. Circadian neurogenetics and its implications in neurophysiology, behavior, and chronomedicine. Neurosci Biobehav Rev 2024; 157:105523. [PMID: 38142983 PMCID: PMC10872425 DOI: 10.1016/j.neubiorev.2023.105523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 12/26/2023]
Abstract
The circadian rhythm affects multiple physiological processes, and disruption of the circadian system can be involved in a range of disease-related pathways. The genetic underpinnings of the circadian rhythm have been well-studied in model organisms. Significant progress has been made in understanding how clock genes affect the physiological functions of the nervous system. In addition, circadian timing is becoming a key factor in improving drug efficacy and reducing drug toxicity. The circadian biology of the target cell determines how the organ responds to the drug at a specific time of day, thus regulating pharmacodynamics. The current review brings together recent advances that have begun to unravel the molecular mechanisms of how the circadian clock affects neurophysiological and behavioral processes associated with human brain diseases. We start with a brief description of how the ubiquitous circadian rhythms are regulated at the genetic, cellular, and neural circuit levels, based on knowledge derived from extensive research on model organisms. We then summarize the latest findings from genetic studies of human brain disorders, focusing on the role of human clock gene variants in these diseases. Lastly, we discuss the impact of common dietary factors and medications on human circadian rhythms and advocate for a broader application of the concept of chronomedicine.
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Owen Y Chao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Joseph P Huston
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Ruifeng Cao
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA; Department of Neurology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
7
|
Suen TC, DeBruyne JP. Lysine-independent ubiquitination and degradation of REV-ERBα involves a bi-functional degradation control sequence at its N-terminus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.01.538963. [PMID: 37205588 PMCID: PMC10187254 DOI: 10.1101/2023.05.01.538963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
REV-ERBα and REV-ERBβ proteins play crucial roles in linking the circadian system to overt daily rhythms in mammalian physiology and behavior. In most tissues, REV-ERBα protein robustly cycles such that it is detected only within a tight interval of 4-6 hours each day, suggesting both its synthesis and degradation are tightly controlled. Several ubiquitin ligases are known to drive REV-ERBα degradation, but how they interact with REV-ERBα and which lysine residues they ubiquitinate to promote degradation are unknown. In this study, we attempted to identify both ubiquitin-ligase-binding and ubiquitination sites within REV-ERBα required for its degradation. Surprisingly, mutating all lysine residues, the common sites for ubiquitin conjugation, in REV-ERBα to arginines (K20R), did very little to impair its degradation in cells. K20R were degraded much faster by co-expression of two E3 ligases, SIAH2 or SPSB4, suggesting possible N-terminal ubiquitination. To explore this, we examined if small deletions at the N-terminus of REV-ERBα would alter its degradation. Interestingly, deletion of amino acid (AA) residues 2 to 9 (delAA2-9) clearly resulted in a less stable REV-ERBα. We found that it was the length (i.e. 8 AA), and not the specific sequence, that confers stability in this region. Simultaneously, we also mapped the interaction site of the E3 ligase SPSB4 to this same region, specifically requiring AA4-9 of REV-ERBα. Thus, the first 9 AA of REV-ERBα has two opposing roles in regulating REV-ERBα turnover. Further, deleting eight additional AAs (delAA2-17) from the N-terminus strongly prevents REV-ERBα degradation. Combined, these results suggest that complex interactions within the first 25AAs potentially act as an endogenous 'switch' that allows REV-ERBα to exist in a stabilized conformation in order to accumulate at one time of day, but then rapidly shifts to a destabilized form, to enhance its removal at the end of its daily cycle.
Collapse
|
8
|
Srinivasan A, Giri A, Duraisamy SK, Alsup A, Castro M, Sundar IK. Acute HDM exposure shows time-of-day and sex-based differences in the severity of lung inflammation and circadian clock disruption. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:100155. [PMID: 37781650 PMCID: PMC10509939 DOI: 10.1016/j.jacig.2023.100155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 10/03/2023]
Abstract
Background Asthma is a chronic inflammatory disease that shows a time-of-day response to variations in symptoms/severity. However, how the lung circadian clock influences time-of-day response and sex-based differences in house dust mite (HDM)-induced airway inflammation and remodeling has not been thoroughly investigated. Objective We sought to determine whether acute HDM exposure in wild-type mice shows time-of-day response and sex-based differences in allergic airway inflammation and circadian clock disruption in the lungs. Methods Wild-type (C57BL/6J) and Rev-erbα knockout (KO) mice were exposed to either PBS or HDM (for 10 days) intranasally at Zeitgeber time (ZT0: 6 am; ZT12: 6 pm) and euthanized 48 hours after the last exposure. Acute HDM-induced time-of-day response and sex-based differences in lung inflammation, gated cytokines/chemokines, humoral and hormonal responses, and circadian clock gene expression were analyzed. Results Acute HDM-exposed mice showed a time-of-day response and sex-based differences in exaggerated lung inflammation (inflammatory eosinophils and interstitial macrophages) at ZT12 when compared with ZT0. HDM-exposed female mice showed increased inflammatory response at ZT12, but HDM-exposed male mice showed comparatively lower inflammation with no time-of-day response. HDM-exposed female and male mice showed augmented IgE levels at ZT12 when compared with ZT0. Myeloid innate immunity panel, cytokines/chemokines, and mucin genes showed a time-of-day gating response at ZT0 and ZT12 in the HDM group. In addition, HDM exposure altered the expression of circadian clock genes in the lung, which was evident in female mice at ZT12. Overall, female mice showed significant time-of-day responses to all these parameters compared with male mice. Rev-erbα KO mice exposed to acute HDM showed exaggerated lung inflammation associated with increased IgE and proinflammatory cytokines in bronchoalveolar lavage fluid. Interestingly, HDM exposure causes reduced expression of clock genes in flow-sorted resident eosinophils but not alveolar macrophages. Acute HDM exposure reduced the nocturnal locomotor activity in mice 5 days post-HDM exposure until day 10. Conclusions This study shows a time-of-day response to acute HDM exposure and sex-based differences in the severity of lung inflammation and humoral immune response associated with circadian clock disruption. Our findings support the use of separate female and male mice cohorts for preclinical studies to understand the molecular heterogeneity in asthma pathophysiology.
Collapse
Affiliation(s)
| | - Allan Giri
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kan
| | - Santhosh Kumar Duraisamy
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kan
| | - Alexander Alsup
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kan
| | - Mario Castro
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kan
| | - Isaac Kirubakaran Sundar
- Corresponding author: Isaac Kirubakaran Sundar, PhD, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160.
| |
Collapse
|
9
|
Kim DY, Park J, Han IO. Hexosamine biosynthetic pathway and O-GlcNAc cycling of glucose metabolism in brain function and disease. Am J Physiol Cell Physiol 2023; 325:C981-C998. [PMID: 37602414 DOI: 10.1152/ajpcell.00191.2023] [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: 05/23/2023] [Revised: 08/03/2023] [Accepted: 08/03/2023] [Indexed: 08/22/2023]
Abstract
Impaired brain glucose metabolism is considered a hallmark of brain dysfunction and neurodegeneration. Disruption of the hexosamine biosynthetic pathway (HBP) and subsequent O-linked N-acetylglucosamine (O-GlcNAc) cycling has been identified as an emerging link between altered glucose metabolism and defects in the brain. Myriads of cytosolic and nuclear proteins in the nervous system are modified at serine or threonine residues with a single N-acetylglucosamine (O-GlcNAc) molecule by O-GlcNAc transferase (OGT), which can be removed by β-N-acetylglucosaminidase (O-GlcNAcase, OGA). Homeostatic regulation of O-GlcNAc cycling is important for the maintenance of normal brain activity. Although significant evidence linking dysregulated HBP metabolism and aberrant O-GlcNAc cycling to induction or progression of neuronal diseases has been obtained, the issue of whether altered O-GlcNAcylation is causal in brain pathogenesis remains uncertain. Elucidation of the specific functions and regulatory mechanisms of individual O-GlcNAcylated neuronal proteins in both normal and diseased states may facilitate the identification of novel therapeutic targets for various neuronal disorders. The information presented in this review highlights the importance of HBP/O-GlcNAcylation in the neuronal system and summarizes the roles and potential mechanisms of O-GlcNAcylated neuronal proteins in maintaining normal brain function and initiation and progression of neurological diseases.
Collapse
Affiliation(s)
- Dong Yeol Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea
| | - Jiwon Park
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea
| | - Inn-Oc Han
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea
| |
Collapse
|
10
|
BaHammam AS, Pirzada A. Timing Matters: The Interplay between Early Mealtime, Circadian Rhythms, Gene Expression, Circadian Hormones, and Metabolism-A Narrative Review. Clocks Sleep 2023; 5:507-535. [PMID: 37754352 PMCID: PMC10528427 DOI: 10.3390/clockssleep5030034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/24/2023] [Accepted: 09/04/2023] [Indexed: 09/28/2023] Open
Abstract
Achieving synchronization between the central and peripheral body clocks is essential for ensuring optimal metabolic function. Meal timing is an emerging field of research that investigates the influence of eating patterns on our circadian rhythm, metabolism, and overall health. This narrative review examines the relationship between meal timing, circadian rhythm, clock genes, circadian hormones, and metabolic function. It analyzes the existing literature and experimental data to explore the connection between mealtime, circadian rhythms, and metabolic processes. The available evidence highlights the importance of aligning mealtime with the body's natural rhythms to promote metabolic health and prevent metabolic disorders. Specifically, studies show that consuming meals later in the day is associated with an elevated prevalence of metabolic disorders, while early time-restricted eating, such as having an early breakfast and an earlier dinner, improves levels of glucose in the blood and substrate oxidation. Circadian hormones, including cortisol and melatonin, interact with mealtimes and play vital roles in regulating metabolic processes. Cortisol, aligned with dawn in diurnal mammals, activates energy reserves, stimulates appetite, influences clock gene expression, and synchronizes peripheral clocks. Consuming meals during periods of elevated melatonin levels, specifically during the circadian night, has been correlated with potential implications for glucose tolerance. Understanding the mechanisms of central and peripheral clock synchronization, including genetics, interactions with chronotype, sleep duration, and hormonal changes, provides valuable insights for optimizing dietary strategies and timing. This knowledge contributes to improved overall health and well-being by aligning mealtime with the body's natural circadian rhythm.
Collapse
Affiliation(s)
- Ahmed S. BaHammam
- The University Sleep Disorders Center, Department of Medicine, College of Medicine, King Saud University, Riyadh 11324, Saudi Arabia
| | - Abdulrouf Pirzada
- North Cumbria Integrated Care (NCIC), National Health Service (NHS), Carlisle CA2 7HY, UK;
| |
Collapse
|
11
|
Duong HA, Baba K, DeBruyne JP, Davidson AJ, Ehlen C, Powell M, Tosini G. Environmental circadian disruption re-programs liver circadian gene expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555175. [PMID: 37693605 PMCID: PMC10491124 DOI: 10.1101/2023.08.28.555175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Circadian gene expression is fundamental to the establishment and functions of the circadian clock, a cell-autonomous and evolutionary-conserved timing system. Yet, how it is affected by environmental-circadian disruption (ECD) such as shiftwork and jetlag, which impact millions of people worldwide, are ill-defined. Here, we provided the first comprehensive description of liver circadian gene expression under normal and after ECD conditions. We found that post-transcription and post-translation processes are dominant contributors to whole-cell or nuclear circadian proteome, respectively. Furthermore, rhythmicity of 64% transcriptome, 98% whole-cell proteome and 95% nuclear proteome is re-written by ECD. The re-writing, which is associated with changes of circadian cis-regulatory elements, RNA-processing and protein trafficking, diminishes circadian regulation of fat and carbohydrate metabolism and persists after one week of ECD-recovery.
Collapse
Affiliation(s)
- Hao A. Duong
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta GA 30310
- Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310
| | - Kenkichi Baba
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta GA 30310
- Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310
| | - Jason P. DeBruyne
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta GA 30310
- Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310
| | - Alec J. Davidson
- Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310
| | - Christopher Ehlen
- Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310
| | - Michael Powell
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta GA 30310
| | - Gianluca Tosini
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta GA 30310
- Neuroscience Institute, Morehouse School of Medicine, Atlanta GA 30310
| |
Collapse
|
12
|
Davidson K, Pickering AM. The proteasome: A key modulator of nervous system function, brain aging, and neurodegenerative disease. Front Cell Dev Biol 2023; 11:1124907. [PMID: 37123415 PMCID: PMC10133520 DOI: 10.3389/fcell.2023.1124907] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/06/2023] [Indexed: 05/02/2023] Open
Abstract
The proteasome is a large multi-subunit protease responsible for the degradation and removal of oxidized, misfolded, and polyubiquitinated proteins. The proteasome plays critical roles in nervous system processes. This includes maintenance of cellular homeostasis in neurons. It also includes roles in long-term potentiation via modulation of CREB signaling. The proteasome also possesses roles in promoting dendritic spine growth driven by proteasome localization to the dendritic spines in an NMDA/CaMKIIα dependent manner. Proteasome inhibition experiments in varied organisms has been shown to impact memory, consolidation, recollection and extinction. The proteasome has been further shown to impact circadian rhythm through modulation of a range of 'clock' genes, and glial function. Proteasome function is impaired as a consequence both of aging and neurodegenerative diseases. Many studies have demonstrated an impairment in 26S proteasome function in the brain and other tissues as a consequence of age, driven by a disassembly of 26S proteasome in favor of 20S proteasome. Some studies also show proteasome augmentation to correct age-related deficits. In amyotrophic lateral sclerosis Alzheimer's, Parkinson's and Huntington's disease proteasome function is impaired through distinct mechanisms with impacts on disease susceptibility and progression. Age and neurodegenerative-related deficits in the function of the constitutive proteasome are often also accompanied by an increase in an alternative form of proteasome called the immunoproteasome. This article discusses the critical role of the proteasome in the nervous system. We then describe how proteasome dysfunction contributes to brain aging and neurodegenerative disease.
Collapse
Affiliation(s)
- Kanisa Davidson
- Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andrew M. Pickering
- Center for Neurodegeneration and Experimental Therapeutics (CNET), Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
13
|
Yu Y, Li W, Xu L, Wang Y. Circadian rhythm of plasminogen activator inhibitor-1 and cardiovascular complications in type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1124353. [PMID: 37020596 PMCID: PMC10067678 DOI: 10.3389/fendo.2023.1124353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/27/2023] [Indexed: 03/22/2023] Open
Abstract
Cardiovascular complications are a common death cause in type 2 diabetes patients, as they are often combined. Plasminogen-activator Inhibitor 1 (PAI-1) participates in the development and progression of cardiovascular complications in diabetes. Insulin resistance increases PAI-1 production, and high PAI-1 levels lead to an environment conducive to thrombosis and earlier and more severe vascular disease. Current evidence also suggests that PAI-1 has a rhythmic profile of circadian fluctuations and acrophase in the morning within a single day, which might explain the high morning incidence of cardiovascular events. Thus, PAI-1 is a possible drug target. Although several PAI-1 inhibitors have been developed, none have yet been allowed for clinical use. Research on rhythm has also led to the concept of "chronotherapy", a rhythm-based drug regimen expected to improve the treatment of cardiovascular complications in diabetic patients. Herein, we searched several databases and reviewed relevant articles to describe the circadian rhythm characteristics and endogenous molecular mechanisms of PAI-1, its relationship with insulin resistance, the causes of cardiovascular complications caused by PAI-1, and the current development of PAI-1 inhibitors. We also summarized the possibility of using the circadian rhythm of PAI-1 to treat cardiovascular complications in diabetic patients.
Collapse
|
14
|
Singh A, Li C, Diernfellner ACR, Höfer T, Brunner M. Data-driven modelling captures dynamics of the circadian clock of Neurospora crassa. PLoS Comput Biol 2022; 18:e1010331. [PMID: 35951637 PMCID: PMC9397904 DOI: 10.1371/journal.pcbi.1010331] [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: 03/07/2022] [Revised: 08/23/2022] [Accepted: 06/23/2022] [Indexed: 11/18/2022] Open
Abstract
Eukaryotic circadian clocks are based on self-sustaining, cell-autonomous oscillatory feedback loops that can synchronize with the environment via recurrent stimuli (zeitgebers) such as light. The components of biological clocks and their network interactions are becoming increasingly known, calling for a quantitative understanding of their role for clock function. However, the development of data-driven mathematical clock models has remained limited by the lack of sufficiently accurate data. Here we present a comprehensive model of the circadian clock of Neurospora crassa that describe free-running oscillations in constant darkness and entrainment in light-dark cycles. To parameterize the model, we measured high-resolution time courses of luciferase reporters of morning and evening specific clock genes in WT and a mutant strain. Fitting the model to such comprehensive data allowed estimating parameters governing circadian phase, period length and amplitude, and the response of genes to light cues. Our model suggests that functional maturation of the core clock protein Frequency causes a delay in negative feedback that is critical for generating circadian rhythms. Circadian rhythms are endogenous autonomous clocks that emancipate daily rhythms in physiology and behavior. Lately, a large body of research has contributed to our understanding of clocks’ genetic and mechanistic basis across kingdoms of life, i.e., mammals, fungi, plants, and bacteria. Several mathematical models have made key contributions to our current understanding of the design principles of the Neurospora crassa circadian clock and conditions for self-sustained oscillations. However, previous models uncovered and described the principle properties of the clock in generic manner due to a lack of experimental data. In this study, we developed a mathematical model based on systems of differential equations to describe the core clock components and estimated model parameters from luciferase data that capture experimental observations. We demonstrate the model predictive control simulation emphasizing the importance of functional maturation of the core clock protein Frequency in generating circadian rhythms.
Collapse
Affiliation(s)
- Amit Singh
- Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Congxin Li
- Theoretical Systems Biology [B086] Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | | | - Thomas Höfer
- Theoretical Systems Biology [B086] Deutsches Krebsforschungszentrum, Heidelberg, Germany
- * E-mail: (TH); (MB)
| | - Michael Brunner
- Heidelberg University Biochemistry Center, Heidelberg, Germany
- * E-mail: (TH); (MB)
| |
Collapse
|
15
|
Comprehensive analysis of the circadian nuclear and cytoplasmic transcriptome in mouse liver. PLoS Genet 2022; 18:e1009903. [PMID: 35921362 PMCID: PMC9377612 DOI: 10.1371/journal.pgen.1009903] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 08/15/2022] [Accepted: 07/06/2022] [Indexed: 11/19/2022] Open
Abstract
In eukaryotes, RNA is synthesised in the nucleus, spliced, and exported to the cytoplasm where it is translated and finally degraded. Any of these steps could be subject to temporal regulation during the circadian cycle, resulting in daily fluctuations of RNA accumulation and affecting the distribution of transcripts in different subcellular compartments. Our study analysed the nuclear and cytoplasmic, poly(A) and total transcriptomes of mouse livers collected over the course of a day. These data provide a genome-wide temporal inventory of enrichment in subcellular RNA, and revealed specific signatures of splicing, nuclear export and cytoplasmic mRNA stability related to transcript and gene lengths. Combined with a mathematical model describing rhythmic RNA profiles, we could test the rhythmicity of export rates and cytoplasmic degradation rates of approximately 1400 genes. With nuclear export times usually much shorter than cytoplasmic half-lives, we found that nuclear export contributes to the modulation and generation of rhythmic profiles of 10% of the cycling nuclear mRNAs. This study contributes to a better understanding of the dynamic regulation of the transcriptome during the day-night cycle.
Collapse
|
16
|
Dekens MPS, Fontinha BM, Gallach M, Pflügler S, Tessmar‐Raible K. Melanopsin elevates locomotor activity during the wake state of the diurnal zebrafish. EMBO Rep 2022; 23:e51528. [PMID: 35233929 PMCID: PMC9066073 DOI: 10.15252/embr.202051528] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 01/24/2022] [Accepted: 02/04/2022] [Indexed: 11/28/2022] Open
Abstract
Mammalian and fish pineals play a key role in adapting behaviour to the ambient light conditions through the release of melatonin. In mice, light inhibits nocturnal locomotor activity via the non‐visual photoreceptor Melanopsin. In contrast to the extensively studied function of Melanopsin in the indirect regulation of the rodent pineal, its role in the intrinsically photosensitive zebrafish pineal has not been elucidated. Therefore, it is not evident if the light signalling mechanism is conserved between distant vertebrates, and how Melanopsin could affect diurnal behaviour. A double knockout of melanopsins (opn4.1‐opn4xb) was generated in the diurnal zebrafish, which manifests attenuated locomotor activity during the wake state. Transcriptome sequencing gave insight into pathways downstream of Melanopsin, implying that sustained repression of the melatonin pathway is required to elevate locomotor activity during the diurnal wake state. Moreover, we show that light induces locomotor activity during the diurnal wake state in an intensity‐dependent manner. These observations suggest a common Melanopsin‐driven mechanism between zebrafish and mammals, while the diurnal and nocturnal chronotypes are inversely regulated downstream of melatonin.
Collapse
Affiliation(s)
- Marcus P S Dekens
- Max Perutz Laboratory Centre for Molecular Biology University of Vienna and Medical University of Vienna Vienna Austria
| | - Bruno M Fontinha
- Max Perutz Laboratory Centre for Molecular Biology University of Vienna and Medical University of Vienna Vienna Austria
| | - Miguel Gallach
- Max Perutz Laboratory Centre for Molecular Biology University of Vienna and Medical University of Vienna Vienna Austria
- Max Perutz Laboratory Centre for Integrative Bioinformatics University of Vienna and Medical University of Vienna Vienna Austria
| | - Sandra Pflügler
- Max Perutz Laboratory Centre for Molecular Biology University of Vienna and Medical University of Vienna Vienna Austria
| | - Kristin Tessmar‐Raible
- Max Perutz Laboratory Centre for Molecular Biology University of Vienna and Medical University of Vienna Vienna Austria
- Research Platform “Marine Rhythms of Life” University of Vienna Vienna Austria
| |
Collapse
|
17
|
Zadeh-Haghighi H, Simon C. Radical pairs can explain magnetic field and lithium effects on the circadian clock. Sci Rep 2022; 12:269. [PMID: 34997158 PMCID: PMC8742017 DOI: 10.1038/s41598-021-04334-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/14/2021] [Indexed: 12/21/2022] Open
Abstract
Drosophila's circadian clock can be perturbed by magnetic fields, as well as by lithium administration. Cryptochromes are critical for the circadian clock. Further, the radical pairs in cryptochrome also can explain magnetoreception in animals. Based on a simple radical pair mechanism model of the animal magnetic compass, we show that both magnetic fields and lithium can influence the spin dynamics of the naturally occurring radical pairs and hence modulate the circadian clock's rhythms. Using a simple chemical oscillator model for the circadian clock, we show that the spin dynamics influence a rate in the chemical oscillator model, which translates into a change in the circadian period. Our model can reproduce the results of two independent experiments, magnetic field and lithium effects on the circadian clock. Our model predicts that stronger magnetic fields would shorten the clock's period. We also predict that lithium influences the clock in an isotope-dependent manner. Furthermore, our model also predicts that magnetic fields and hyperfine interactions modulate oxidative stress. The findings of this work suggest that the quantum nature of radical pairs might play roles in the brain, as another piece of evidence in addition to recent results on xenon anesthesia and lithium effects on hyperactivity.
Collapse
Affiliation(s)
- Hadi Zadeh-Haghighi
- Department of Physics and Astronomy, University of Calgary, Calgary, AB, T2N 1N4, Canada.
- Institute for Quantum Science and Technology, University of Calgary, Calgary, AB, T2N 1N4, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| | - Christoph Simon
- Department of Physics and Astronomy, University of Calgary, Calgary, AB, T2N 1N4, Canada.
- Institute for Quantum Science and Technology, University of Calgary, Calgary, AB, T2N 1N4, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
18
|
Chen LC, Hsieh YL, Tan GYT, Kuo TY, Chou YC, Hsu PH, Hwang-Verslues WW. Differential effects of SUMO1 and SUMO2 on circadian protein PER2 stability and function. Sci Rep 2021; 11:14431. [PMID: 34257372 PMCID: PMC8277905 DOI: 10.1038/s41598-021-93933-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 07/05/2021] [Indexed: 11/09/2022] Open
Abstract
Posttranslational modification (PTM) of core circadian clock proteins, including Period2 (PER2), is required for proper circadian regulation. PER2 function is regulated by casein kinase 1 (CK1)-mediated phosphorylation and ubiquitination but little is known about other PER2 PTMs or their interaction with PER2 phosphorylation. We found that PER2 can be SUMOylated by both SUMO1 and SUMO2; however, SUMO1 versus SUMO2 conjugation had different effects on PER2 turnover and transcriptional suppressor function. SUMO2 conjugation facilitated PER2 interaction with β-TrCP leading to PER2 proteasomal degradation. In contrast, SUMO1 conjugation, mediated by E3 SUMO-protein ligase RanBP2, enhanced CK1-mediated PER2S662 phosphorylation, inhibited PER2 degradation and increased PER2 transcriptional suppressor function. PER2 K736 was critical for both SUMO1- and SUMO2-conjugation. A PER2K736R mutation was sufficient to alter PER2 protein oscillation and reduce PER2-mediated transcriptional suppression. Together, our data revealed that SUMO1 versus SUMO2 conjugation acts as a determinant of PER2 stability and function and thereby affects the circadian regulatory system and the expression of clock-controlled genes.
Collapse
Affiliation(s)
- Ling-Chih Chen
- Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Road, Taipei, 115, Taiwan, ROC
| | - Yung-Lin Hsieh
- Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Road, Taipei, 115, Taiwan, ROC
| | - Grace Y T Tan
- Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Road, Taipei, 115, Taiwan, ROC
| | - Tai-Yun Kuo
- Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Road, Taipei, 115, Taiwan, ROC
| | - Yu-Chi Chou
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 115, Taiwan, ROC
| | - Pang-Hung Hsu
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung City, 202, Taiwan, ROC
| | - Wendy W Hwang-Verslues
- Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Road, Taipei, 115, Taiwan, ROC.
| |
Collapse
|
19
|
Gao Y, Wu Y, Zhang N, Yuan H, Wang F, Xu H, Yu J, Ma J, Hou S, Cao X. IDH1 gene mutation activates Smad signaling molecules to regulate the expression levels of cell cycle and biological rhythm genes in human glioma U87‑MG cells. Mol Med Rep 2021; 23:354. [PMID: 33760141 PMCID: PMC7974315 DOI: 10.3892/mmr.2021.11993] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Isocitrate dehydrogenase1 (IDH1) mutation is the most important genetic change in glioma. The most common IDH1 mutation results in the amino acid substitution of arginine 132 (Arg/R132), which is located at the active site of the enzyme. IDH1 Arg132His (R132H) mutation can reduce the proliferative rate of glioma cells. Numerous diseases follow circadian rhythms, and there is growing evidence that circadian disruption may be a risk factor for cancer in humans. Dysregulation of the circadian clock serves an important role in the development of malignant tumors, including glioma. Brain-Muscle Arnt-Like protein 1 (BMAL1) and Circadian Locomotor Output Cycles Kaput (CLOCK) are the main biological rhythm genes. The present study aimed to further study whether there is an association between IDH1 R132H mutation and biological rhythm in glioma, and whether this affects the occurrence of glioma. The Cancer Genome Atlas (TCGA) database was used to detect the expression levels of the biological rhythm genes BMAL1 and CLOCK in various types of tumor. Additionally, U87-MG cells were infected with wild-type and mutant IDH1 lentiviruses. Colony formation experiments were used to detect cell proliferation in each group, cell cycle distribution was detected by flow cytometry and western blotting was used to detect the expression levels of wild-type and mutant IDH1, cyclins, biological rhythm genes and Smad signaling pathway-associated genes in U87-MG cells. TCGA database results suggested that BMAL1 and CLOCK were abnormally expressed in glioma. Cells were successfully infected with wild-type and mutant IDH1 lentiviruses. Colony formation assay revealed decreased cell proliferation in the IDH1 R132H mutant group. The cell cycle distribution detected by flow cytometry indicated that IDH1 gene mutation increased the G1 phase ratio and decreased the S phase ratio in U87-MG cells. The western blotting results demonstrated that IDH1 R132H mutation decreased the expression levels of the S phase-associated proteins Cyclin A and CDK2, and increased the expression levels of the G1 phase-associated proteins Cyclin D3 and CDK4, but did not significantly change the expression levels of the G2/M phase-associated protein Cyclin B1. The expression levels of the positive and negative rhythm regulation genes BMAL1, CLOCK, period (PER s (PER1, 2 and 3) and cryptochrom (CRY)s (CRY1 and 2) were significantly decreased, those of the Smad signaling pathway-associated genes Smad2, Smad3 and Smad2-3 were decreased, and those of phosphorylated (p)-Smad2, p-Smad3 and Smad4 were increased. Therefore, the present results suggested that the IDH1 R132H mutation may alter the cell cycle and biological rhythm genes in U87-MG cells through the TGF-β/Smad signaling pathway.
Collapse
Affiliation(s)
- Yongying Gao
- Department of Pathology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yanwei Wu
- Department of Pathology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Ningmei Zhang
- Department of Pathology, Tumor Hospital, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Hongmei Yuan
- Functional Department, Ningxia Hui Autonomous Region People's Hospital, Yinchuan, Ningxia 750021, P.R. China
| | - Fei Wang
- Department of Pathology, The First People's Hospital of Yinchuan, Yinchuan, Ningxia 750001, P.R. China
| | - Hui Xu
- Department of Pathology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jiaxiang Yu
- Department of Pathology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jie Ma
- Department of Pathology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Shaozhang Hou
- Department of Pathology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xiangmei Cao
- Department of Pathology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
20
|
Circadian Misalignment Induced by Chronic Night Shift Work Promotes Endoplasmic Reticulum Stress Activation Impacting Directly on Human Metabolism. BIOLOGY 2021; 10:biology10030197. [PMID: 33807589 PMCID: PMC7998626 DOI: 10.3390/biology10030197] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 01/04/2023]
Abstract
Simple Summary The demands of modern society have made shift work a necessity. Night work is associated with an increased risk of metabolic problems such as obesity and diabetes, which is mainly due to the misalignment of circadian rhythms that play a crucial role in many biological processes. This study performed clinical, anthropometric, and molecular analyses on 40 hospital workers who work day or night. We demonstrated that night workers had increased glucose levels, triglycerides, waist circumference, and blood pressure compared to day workers. Surprisingly, we report that night workers have significant changes in the expression of circadian clock genes and an up-regulation of genes related to endoplasmic reticulum stress (ERS). These findings provide new insights into the effects of night shift work on the expression of circadian cycle genes and ERS activation, leading to metabolic stress and the development of metabolic diseases associated with night work. Abstract Night work has become necessary in our modern society. However, sleep deprivation induces a circadian misalignment that effectively contributes to the development of diseases associated with metabolic syndrome, such as obesity and diabetes. Here, we evaluated the pattern of circadian clock genes and endoplasmic reticulum stress (ERS) genes in addition to metabolic and anthropometric measures in subjects that work during a nocturnal period compared with day workers. We study 20 night workers (NW) and 20 day workers (DW) submitted to a work schedule of 12 h of work for 36 h of rest for at least 5 years in a hospital. The present report shows that NW have increased fasting blood glucose, glycated hemoglobin (HbA1c), triglycerides, and low-density lipoprotein (LDL)-cholesterol levels, and lower high-density lipoprotein (HDL)-cholesterol levels compared to DW. In addition, we observed that waist circumference (WC), waist–hip ratio (WHR), and systemic blood pressure are also increased in NW. Interestingly, gene expression analysis showed changes in CLOCK gene expression in peripheral blood mononuclear cells (PBMC) samples of NW compared to the DW, evidencing a peripheral circadian misalignment. This metabolic adaptation was accompanied by the up-regulation of many genes of ERS in NW. These findings support the hypothesis that night shift work results in disturbed glycemic and lipid control and affects the circadian cycle through the deregulation of peripheral CLOCK genes, which is possibly due to the activation of ERS. Thus, night work induces important metabolic changes that increase the risk of developing metabolic syndrome.
Collapse
|
21
|
Heyde I, Begemann K, Oster H. Contributions of white and brown adipose tissues to the circadian regulation of energy metabolism. Endocrinology 2021; 162:6102571. [PMID: 33453099 PMCID: PMC7864004 DOI: 10.1210/endocr/bqab009] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 12/17/2022]
Abstract
The term energy metabolism comprises the entirety of chemical processes associated with uptake, conversion, storage, and breakdown of nutrients. All these must be tightly regulated in time and space to ensure metabolic homeostasis in an environment characterized by cycles such as the succession of day and night. Most organisms evolved endogenous circadian clocks to achieve this goal. In mammals, a ubiquitous network of cellular clocks is coordinated by a pacemaker residing in the hypothalamic suprachiasmatic nucleus. Adipocytes harbor their own circadian clocks, and large aspects of adipose physiology are regulated in a circadian manner through transcriptional regulation of clock-controlled genes. White adipose tissue (WAT) stores energy in the form of triglycerides at times of high energy levels that then serve as fuel in times of need. It also functions as an endocrine organ, releasing factors in a circadian manner to regulate food intake and energy turnover in other tissues. Brown adipose tissue (BAT) produces heat through nonshivering thermogenesis, a process also controlled by the circadian clock. We here review how WAT and BAT contribute to the circadian regulation of energy metabolism. We describe how adipose rhythms are regulated by the interplay of systemic signals and local clocks and summarize how adipose-originating circadian factors feed-back on metabolic homeostasis. The role of adipose tissue in the circadian control of metabolism becomes increasingly clear as circadian disruption leads to alterations in adipose tissue regulation, promoting obesity and its sequelae. Stabilizing adipose tissue rhythms, in turn, may help to combat disrupted energy homeostasis and obesity.
Collapse
Affiliation(s)
- Isabel Heyde
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | | | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
- Correspondence: Henrik Oster, PhD, Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany.
| |
Collapse
|
22
|
Liu JA, Walton JC, DeVries AC, Nelson RJ. Disruptions of Circadian Rhythms and Thrombolytic Therapy During Ischemic Stroke Intervention. Front Neurosci 2021; 15:675732. [PMID: 34177452 PMCID: PMC8222607 DOI: 10.3389/fnins.2021.675732] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/11/2021] [Indexed: 11/24/2022] Open
Abstract
Several endogenous and exogenous factors interact to influence stroke occurrence, in turn contributing to discernable daily distribution patterns in the frequency and severity of cerebrovascular events. Specifically, strokes that occur during the morning tend to be more severe and are associated with elevated diastolic blood pressure, increased hospital stay, and worse outcomes, including mortality, compared to strokes that occur later in the day. Furthermore, disrupted circadian rhythms are linked to higher risk for stroke and play a role in stroke outcome. In this review, we discuss the interrelation among core clock genes and several factors contributing to ischemic outcomes, sources of disrupted circadian rhythms, the implications of disrupted circadian rhythms in foundational stroke scientific literature, followed by a review of clinical implications. In addition to highlighting the distinct daily pattern of onset, several aspects of physiology including immune response, endothelial/vascular and blood brain barrier function, and fibrinolysis are under circadian clock regulation; disrupted core clock gene expression patterns can adversely affect these physiological processes, leading to a prothrombotic state. Lastly, we discuss how the timing of ischemic onset increases morning resistance to thrombolytic therapy and the risk of hemorrhagic transformation.
Collapse
Affiliation(s)
- Jennifer A Liu
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - James C Walton
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - A Courtney DeVries
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States.,Department of Medicine, Division of Oncology/Hematology, West Virginia University, Morgantown, WV, United States.,West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, United States
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
23
|
Yaw AM, McLane-Svoboda AK, Hoffmann HM. Shiftwork and Light at Night Negatively Impact Molecular and Endocrine Timekeeping in the Female Reproductive Axis in Humans and Rodents. Int J Mol Sci 2020; 22:E324. [PMID: 33396885 PMCID: PMC7795361 DOI: 10.3390/ijms22010324] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/24/2020] [Accepted: 12/25/2020] [Indexed: 01/17/2023] Open
Abstract
Shiftwork, including work that takes place at night (nightshift) and/or rotates between day and nightshifts, plays an important role in our society, but is associated with decreased health, including reproductive dysfunction. One key factor in shiftwork, exposure to light at night, has been identified as a likely contributor to the underlying health risks associated with shiftwork. Light at night disrupts the behavioral and molecular circadian timekeeping system, which is important for coordinated timing of physiological processes, causing mistimed hormone release and impaired physiological functions. This review focuses on the impact of shiftwork on reproductive function and pregnancy in women and laboratory rodents and potential underlying molecular mechanisms. We summarize the negative impact of shiftwork on female fertility and compare these findings to studies in rodent models of light shifts. Light-shift rodent models recapitulate several aspects of reproductive dysfunction found in shift workers, and their comparison with human studies can enable a deeper understanding of physiological and hormonal responses to light shifts and the underlying molecular mechanisms that may lead to reproductive disruption in human shift workers. The contributions of human and rodent studies are essential to identify the origins of impaired fertility in women employed in shiftwork.
Collapse
Affiliation(s)
| | | | - Hanne M. Hoffmann
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
24
|
Abstract
Circadian rhythms are biological systems that synchronize cellular circadian oscillators with the organism's daily feeding-fasting or rest-activity cycles in mammals. Circadian rhythms regulate nutrient absorption and utilization at the cellular level and are closely related to obesity and metabolic disorders. Bile acids are important modulators that facilitate nutrient absorption and regulate energy metabolism. Here, we provide an overview of the current connections and future perspectives between the circadian clock and bile acid metabolism as well as related metabolic diseases. Feeding and fasting cycles influence bile acid pool size and composition, and bile acid signaling can respond to acute lipid and glucose utilization and mediate energy balance. Disruption of circadian rhythms such as shift work, irregular diet, and gene mutations can contribute to altered bile acid metabolism and heighten obesity risk. High-fat diets, alcohol, and gene mutations related to bile acid signaling result in desynchronized circadian rhythms. Gut microbiome also plays a role in connecting circadian rhythms with bile acid metabolism. The underlying mechanism of how circadian rhythms interact with bile acid metabolism has not been fully explored. Sustaining bile acid homeostasis based on circadian rhythms may be a potential therapy to alleviate metabolic disturbance.
Collapse
Affiliation(s)
- Yunxia Yang
- Center for Molecular Metabolism, Nanjing University of Science and Technology, Nanjing, China
| | - Jianfa Zhang
- Center for Molecular Metabolism, Nanjing University of Science and Technology, Nanjing, China
| |
Collapse
|
25
|
Malaguarnera R, Ledda C, Filippello A, Frasca F, Francavilla VC, Ramaci T, Parisi MC, Rapisarda V, Piro S. Thyroid Cancer and Circadian Clock Disruption. Cancers (Basel) 2020; 12:E3109. [PMID: 33114365 PMCID: PMC7690860 DOI: 10.3390/cancers12113109] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/18/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
Thyroid cancer (TC) represents the most common malignancy of the endocrine system, with an increased incidence across continents attributable to both improvement of diagnostic procedures and environmental factors. Among the modifiable risk factors, insulin resistance might influence the development of TC. A relationship between circadian clock machinery disfunction and TC has recently been proposed. The circadian clock machinery comprises a set of rhythmically expressed genes responsible for circadian rhythms. Perturbation of this system contributes to the development of pathological states such as cancer. Several clock genes have been found deregulated upon thyroid nodule malignant transformation. The molecular mechanisms linking circadian clock disruption and TC are still unknown but could include insulin resistance. Circadian misalignment occurring during shift work, jet lag, high fat food intake, is associated with increased insulin resistance. This metabolic alteration, in turn, is associated with a well-known risk factor for TC i.e., hyperthyrotropinemia, which could also be induced by sleep disturbances. In this review, we describe the mechanisms controlling the circadian clock function and its involvement in the cell cycle, stemness and cancer. Moreover, we discuss the evidence supporting the link between circadian clockwork disruption and TC development/progression, highlighting its potential implications for TC prevention, diagnosis and therapy.
Collapse
Affiliation(s)
- Roberta Malaguarnera
- School of Human and Social Sciences, “Kore” University of Enna, 94100 Enna, Italy; (R.M.); (V.C.F.); (T.R.); (M.C.P.)
| | - Caterina Ledda
- Department of Clinical and Experimental Medicine, Occupational Medicine, University of Catania, 95100 Catania, Italy;
| | - Agnese Filippello
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, Italy; (A.F.); (S.P.)
| | - Francesco Frasca
- Endocrinology Unit, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, Italy;
| | - Vincenzo Cristian Francavilla
- School of Human and Social Sciences, “Kore” University of Enna, 94100 Enna, Italy; (R.M.); (V.C.F.); (T.R.); (M.C.P.)
| | - Tiziana Ramaci
- School of Human and Social Sciences, “Kore” University of Enna, 94100 Enna, Italy; (R.M.); (V.C.F.); (T.R.); (M.C.P.)
| | - Maria Chiara Parisi
- School of Human and Social Sciences, “Kore” University of Enna, 94100 Enna, Italy; (R.M.); (V.C.F.); (T.R.); (M.C.P.)
| | - Venerando Rapisarda
- Department of Clinical and Experimental Medicine, Occupational Medicine, University of Catania, 95100 Catania, Italy;
| | - Salvatore Piro
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, Italy; (A.F.); (S.P.)
| |
Collapse
|
26
|
Li Y, Li G, Li J, Cai X, Sun Y, Zhang B, Zhao H. Depression-like behavior is associated with lower Per2 mRNA expression in the lateral habenula of rats. GENES BRAIN AND BEHAVIOR 2020; 20:e12702. [PMID: 32964673 DOI: 10.1111/gbb.12702] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 09/10/2020] [Accepted: 09/12/2020] [Indexed: 12/22/2022]
Abstract
Circadian rhythm dysfunction is primary symptom of depression and is closely related to depression onset. The role of the lateral habenula (LHb) of the thalamus in the pathogenesis of depression has been a research topic of great interest. The neuronal activity of this structure has circadian characteristics, which are related to the regulation of circadian rhythms. However, in depression model of rats, the role of clock genes in the LHb has not been assessed. To address this gap, we used a clomipramine (CLI) injection-induced depression model in rats to assess the daily expression of rhythmic genes in the LHb and depression-like behavior in rats at multiple time points. In determining the role of the Per2 gene in the development of depression-like behavior in the LHb, we found that the expression of this clock gene differed in a circadian manner. Per2 expression was also significantly decreased in CLI-treated rats in late afternoon (17:00) and in the middle of the night (1:00). Furthermore, silencing Per2 in the LHb of normal rats induced depression-like behavior at night, suggesting that Per2 may play an important role in the pathogenesis of depression. Collectively, these results indicate that decreased Per2 expression in the LHb may be related to increased depression-like behavior at night in depression model of rats.
Collapse
Affiliation(s)
- Yang Li
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Guangjian Li
- Department of Neurology and Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| | - Jicheng Li
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xuewei Cai
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yanfei Sun
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Beilin Zhang
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Hua Zhao
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China.,Department of Neurology and Neuroscience Research Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
27
|
An K, Zhao H, Miao Y, Xu Q, Li YF, Ma YQ, Shi YM, Shen JW, Meng JJ, Yao YG, Zhang Z, Chen JT, Bao J, Zhang M, Xue T. A circadian rhythm-gated subcortical pathway for nighttime-light-induced depressive-like behaviors in mice. Nat Neurosci 2020; 23:869-880. [PMID: 32483349 DOI: 10.1038/s41593-020-0640-8] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/10/2020] [Indexed: 12/11/2022]
Abstract
Besides generating vision, light modulates various physiological functions, including mood. While light therapy applied in the daytime is known to have anti-depressive properties, excessive light exposure at night has been reportedly associated with depressive symptoms. The neural mechanisms underlying this day-night difference in the effects of light are unknown. Using a light-at-night (LAN) paradigm in mice, we showed that LAN induced depressive-like behaviors without disturbing the circadian rhythm. This effect was mediated by a neural pathway from retinal melanopsin-expressing ganglion cells to the dorsal perihabenular nucleus (dpHb) to the nucleus accumbens (NAc). Importantly, the dpHb was gated by the circadian rhythm, being more excitable at night than during the day. This indicates that the ipRGC→dpHb→NAc pathway preferentially conducts light signals at night, thereby mediating LAN-induced depressive-like behaviors. These findings may be relevant when considering the mental health effects of the prevalent nighttime illumination in the industrial world.
Collapse
Affiliation(s)
- Kai An
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huan Zhao
- College of Biology, Food and Environment, Hefei University, Hefei, China.
| | - Ying Miao
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Qi Xu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yu-Fei Li
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yu-Qian Ma
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yi-Ming Shi
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jia-Wei Shen
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jian-Jun Meng
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zhi Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ju-Tao Chen
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jin Bao
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Mei Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tian Xue
- Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. .,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China. .,Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
28
|
Jin Y, Olsen RE, Harvey TN, Østensen MA, Li K, Santi N, Vadstein O, Bones AM, Vik JO, Sandve SR, Olsen Y. Comparative transcriptomics reveals domestication-associated features of Atlantic salmon lipid metabolism. Mol Ecol 2020; 29:1860-1872. [PMID: 32293070 DOI: 10.1111/mec.15446] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 04/01/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022]
Abstract
Domestication of animals imposes strong targeted selection for desired traits but can also result in unintended selection due to new domestic environments. Atlantic salmon (Salmo salmar) was domesticated in the 1970s and has subsequently been selected for faster growth in systematic breeding programmes. More recently, salmon aquaculture has replaced fish oils (FOs) with vegetable oils (VOs) in feed, radically changing the levels of essential long-chain polyunsaturated fatty acids (LC-PUFAs). Our aim here was to study the impact of domestication on metabolism and explore the hypothesis that the shift to VO diets has unintentionally selected for a domestication-specific lipid metabolism. We conducted a 96-day feeding trial of domesticated and wild salmon fed diets based on FOs, VOs or phospholipids, and compared transcriptomes and fatty acids in tissues involved in lipid absorption (pyloric caeca) and lipid turnover and synthesis (liver). Domesticated salmon had faster growth and higher gene expression in glucose and lipid metabolism compared to wild fish, possibly linked to differences in regulation of circadian rhythm pathways. Only the domesticated salmon increased expression of LC-PUFA synthesis genes when given VOs. This transcriptome response difference was mirrored at the physiological level, with domesticated salmon having higher LC-PUFA levels but lower 18:3n-3 and 18:2n-6 levels. In line with this, the VO diet decreased growth rate in wild but not domesticated salmon. Our study revealed a clear impact of domestication on transcriptomic regulation linked to metabolism and suggests that unintentional selection in the domestic environment has resulted in evolution of stronger compensatory mechanisms to a diet low in LC-PUFAs.
Collapse
Affiliation(s)
- Yang Jin
- Department of Biology, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Rolf Erik Olsen
- Department of Biology, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Thomas Nelson Harvey
- Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Mari-Ann Østensen
- Department of Biology, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | | | | | - Olav Vadstein
- Department of Biotechnology and Food Science, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Atle Magnar Bones
- Department of Biology, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Jon Olav Vik
- Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Simen Rød Sandve
- Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Yngvar Olsen
- Department of Biology, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
29
|
Sinturel F, Petrenko V, Dibner C. Circadian Clocks Make Metabolism Run. J Mol Biol 2020; 432:3680-3699. [PMID: 31996313 DOI: 10.1016/j.jmb.2020.01.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/16/2020] [Accepted: 01/16/2020] [Indexed: 12/12/2022]
Abstract
Most organisms adapt to the 24-h cycle of the Earth's rotation by anticipating the time of the day through light-dark cycles. The internal time-keeping system of the circadian clocks has been developed to ensure this anticipation. The circadian system governs the rhythmicity of nearly all physiological and behavioral processes in mammals. In this review, we summarize current knowledge stemming from rodent and human studies on the tight interconnection between the circadian system and metabolism in the body. In particular, we highlight recent advances emphasizing the roles of the peripheral clocks located in the metabolic organs in regulating glucose, lipid, and protein homeostasis at the organismal and cellular levels. Experimental disruption of circadian system in rodents is associated with various metabolic disturbance phenotypes. Similarly, perturbation of the clockwork in humans is linked to the development of metabolic diseases. We discuss recent studies that reveal roles of the circadian system in the temporal coordination of metabolism under physiological conditions and in the development of human pathologies.
Collapse
Affiliation(s)
- Flore Sinturel
- Department of Medicine, Division of Endocrinology, Diabetes, Hypertension and Nutrition, Faculty of Medicine, University of Geneva, Rue Michel-Servet, 1, CH-1211, Geneva, 14, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, Geneva, Switzerland.
| | - Volodymyr Petrenko
- Department of Medicine, Division of Endocrinology, Diabetes, Hypertension and Nutrition, Faculty of Medicine, University of Geneva, Rue Michel-Servet, 1, CH-1211, Geneva, 14, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, Geneva, Switzerland
| | - Charna Dibner
- Department of Medicine, Division of Endocrinology, Diabetes, Hypertension and Nutrition, Faculty of Medicine, University of Geneva, Rue Michel-Servet, 1, CH-1211, Geneva, 14, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, Geneva, Switzerland.
| |
Collapse
|
30
|
Bano‐Otalora B, Madrid JA, Rol MA. Melatonin alleviates circadian system disruption induced by chronic shifts of the light-dark cycle in Octodon degus. J Pineal Res 2020; 68:e12619. [PMID: 31677295 PMCID: PMC6916290 DOI: 10.1111/jpi.12619] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 10/14/2019] [Accepted: 10/20/2019] [Indexed: 12/19/2022]
Abstract
Modern 24-h society lifestyle is associated with experiencing frequent shifts in the lighting conditions which can negatively impact human health. Here, we use the degus, a species exhibiting diurnal and nocturnal chronotypes, to: (a) assess the impact of chronic shifts of the light:dark (LD) cycle in the animal's physiology and behaviour and (b) test the therapeutic potential of melatonin in enhancing rhythmicity under these conditions. Degus were subjected to a "5d + 2d" LD-shifting schedule for 19 weeks. This protocol aims to mimic lighting conditions experienced by humans during shift work: LD cycle was weekly delayed by 8h during 5 "working" days (Morning, Afternoon and Night schedule); during weekends (2 days), animals were kept under Morning schedule. After 9 weeks, melatonin was provided daily for 6h in the drinking water. The "5d + 2d" shifting LD schedule led to a disruption in wheel-running activity (WRA) and body temperature (Tb) rhythms which manifested up to three separate periods in the circadian range. This chronodisruption was more evident in nocturnal than in diurnal degus, particularly during the Afternoon schedule when a phase misalignment between WRA and Tb rhythms appeared. Melatonin treatment and, to a lesser extent, water restriction enhanced the 24-h component, suggesting a potential role in ameliorating the disruptive effects of shift work.
Collapse
Affiliation(s)
- Beatriz Bano‐Otalora
- Chronobiology LabDepartment of PhysiologyFaculty of BiologyUniversity of MurciaIUIEIMIB‐ArrixacaMurciaSpain
- Present address:
Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Juan Antonio Madrid
- Chronobiology LabDepartment of PhysiologyFaculty of BiologyUniversity of MurciaIUIEIMIB‐ArrixacaMurciaSpain
- Ciber Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Maria Angeles Rol
- Chronobiology LabDepartment of PhysiologyFaculty of BiologyUniversity of MurciaIUIEIMIB‐ArrixacaMurciaSpain
- Ciber Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| |
Collapse
|
31
|
Circadian rhythms in Per1, PER2 and Ca 2+ of a solitary SCN neuron cultured on a microisland. Sci Rep 2019; 9:18271. [PMID: 31797953 PMCID: PMC6892917 DOI: 10.1038/s41598-019-54654-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/01/2019] [Indexed: 01/08/2023] Open
Abstract
Circadian rhythms in Per1, PER2 expression and intracellular Ca2+ were measured from a solitary SCN neuron or glial cell which was physically isolated from other cells. Dispersed cells were cultured on a platform of microisland (100–200 μm in diameter) in a culture dish. Significant circadian rhythms were detected in 57.1% for Per1 and 70.0% for PER2 expression. When two neurons were located on the same island, the circadian rhythms showed desynchronization, indicating a lack of oscillatory coupling. Circadian rhythms were also detected in intracellular Ca2+ of solitary SCN neurons. The ratio of circadian positive neurons was significantly larger without co-habitant of glial cells (84.4%) than with it (25.0%). A relatively large fraction of SCN neurons generates the intrinsic circadian oscillation without neural or humoral networks. In addition, glial cells seem to interrupt the expression of the circadian rhythmicity of intracellular Ca2+ under these conditions.
Collapse
|
32
|
Abstract
Circadian clocks drive daily rhythms of physiology and behavior in multiple organisms and synchronize these rhythms to environmental cycles of light and temperature. The basic mechanism of the clock consists of a transcription-translation feedback loop, in which key clock proteins negatively regulate their own transcription. Although much of the focus with respect to clock mechanisms has been on the regulation of transcription and on the stability and activity of clock proteins, it is clear that other regulatory processes also have to be involved to explain aspects of clock function. Here, we review the role of alternative splicing in circadian clocks. Starting with a discussion of the Drosophila clock and then extending to other major circadian model systems, we describe how the control of alternative splicing enables organisms to maintain their circadian clocks as well as to respond to environmental inputs, in particular to temperature changes.
Collapse
Affiliation(s)
- Iryna Shakhmantsir
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amita Sehgal
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
33
|
Bagherzadeh-Azbari S, Khazaie H, Zarei M, Spiegelhalder K, Walter M, Leerssen J, Van Someren EJW, Sepehry AA, Tahmasian M. Neuroimaging insights into the link between depression and Insomnia: A systematic review. J Affect Disord 2019; 258:133-143. [PMID: 31401541 DOI: 10.1016/j.jad.2019.07.089] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 07/06/2019] [Accepted: 07/30/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Insomnia is a common symptom of Major Depressive Disorder (MDD) and genome-wide association studies pointed to their strong genetic association. Although the prevalence of insomnia symptoms in MDD is noticeable and evidence supports their strong bidirectional association, the number of available neuroimaging findings on patients of MDD with insomnia symptoms is limited. However, such neuroimaging studies could verily improve our understanding of their shared pathophysiology and advance corresponding theories. METHODS Based on the preferred reporting items for systematic reviews and meta-analysis (PRISMA) guideline, we have conducted a literature search using PubMed, EMBASE, and Scopus databases and systematically explored 640 studies using various neuroimaging modalities in MDD patients with different degrees of insomnia symptoms. RESULTS Despite inconsistencies, current findings from eight studies suggested structural and functional disturbances in several brain regions including the amygdala, prefrontal cortex and anterior cingulate cortex and insula. The aberrant functional connectivity within and between the main hubs of the salience and default mode networks could potentially yield new insights into the link between MDD and insomnia, which needs further assessment. LIMITATIONS The number of studies reviewed herein is limited. The applied methods for assessing structural and functional neural mechanisms of insomnia and depression were variable. CONCLUSION Neuroimaging methods demonstrated the overlapping underlying neural mechanisms between MDD and insomnia. Future studies may facilitate better understanding of their pathophysiology to allow development of specific treatment.
Collapse
Affiliation(s)
- Shadi Bagherzadeh-Azbari
- Institute of Medical Sciences and Technology, Shahid Beheshti University, Tehran, Iran; Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Habibolah Khazaie
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mojtaba Zarei
- Institute of Medical Sciences and Technology, Shahid Beheshti University, Tehran, Iran
| | - Kai Spiegelhalder
- Department of Psychiatry and Psychotherapy, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Martin Walter
- Department of Psychiatry, University of Tübingen, Tübingen, Germany; Clinical Affective Neuroimaging Laboratory, Leibniz Institute for Neurobiology, Otto-von-Guericke University, Magdeburg, Germany
| | - Jeanne Leerssen
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, Netherlands; Departments of Psychiatry and Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, Vrije Universtiteit Amsterdam, Amsterdam UMC, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - Eus J W Van Someren
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, Netherlands; Departments of Psychiatry and Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, Vrije Universtiteit Amsterdam, Amsterdam UMC, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - Amir A Sepehry
- Clinical and Counselling Psychology Program, Adler University, Vancouver, BC, Canada
| | - Masoud Tahmasian
- Institute of Medical Sciences and Technology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
34
|
Pibiri M. Liver regeneration in aged mice: new insights. Aging (Albany NY) 2019; 10:1801-1824. [PMID: 30157472 PMCID: PMC6128415 DOI: 10.18632/aging.101524] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023]
Abstract
The regenerative capacity of the liver after resection is reduced with aging. Recent studies on rodents revealed that both intracellular and extracellular factors are involved in the impairment of liver mass recovery during aging. Among the intracellular factors, age-dependent decrease of BubR1 (budding uninhibited by benzimidazole-related 1), YAP (Yes-associated protein) and SIRT1 (Sirtuin-1) have been associated to dampening of tissue reconstitution and inhibition of cell cycle genes following partial hepatectomy. Extra-cellular factors, such as age-dependent changes in hepatic stellate cells affect liver regeneration through inhibition of progenitor cells and reduction of liver perfusion. Furthermore, chronic release of pro-inflammatory proteins by senescent cells (SASP) affects cell proliferation suggesting that senescent cell clearance might improve tissue regeneration. Accordingly, young plasma restores liver regeneration in aged animals through autophagy re-establishment. This review will discuss how intracellular and extracellular factors cooperate to guarantee a proper liver regeneration and the possible causes of its impairment during aging. The possibility that an improvement of the liver regenerative capacity in elderly might be achieved through elimination of senescent cells via autophagy or by administration of direct mitogenic agents devoid of cytotoxicity will also be entertained.
Collapse
Affiliation(s)
- Monica Pibiri
- Department of Biomedical Sciences, Oncology and Molecular Pathology Unit, University of Cagliari, Cagliari 09124, Italy
| |
Collapse
|
35
|
Melatonin in Alzheimer’s Disease: A Latent Endogenous Regulator of Neurogenesis to Mitigate Alzheimer’s Neuropathology. Mol Neurobiol 2019; 56:8255-8276. [DOI: 10.1007/s12035-019-01660-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/20/2019] [Accepted: 05/22/2019] [Indexed: 12/15/2022]
|
36
|
Chang WH, Lai AG. Timing gone awry: distinct tumour suppressive and oncogenic roles of the circadian clock and crosstalk with hypoxia signalling in diverse malignancies. J Transl Med 2019; 17:132. [PMID: 31014368 PMCID: PMC6480786 DOI: 10.1186/s12967-019-1880-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/09/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The circadian clock governs a large variety of fundamentally important physiological processes in all three domains of life. Consequently, asynchrony in timekeeping mechanisms could give rise to cellular dysfunction underpinning many disease pathologies including human neoplasms. Yet, detailed pan-cancer evidence supporting this notion has been limited. METHODS In an integrated approach uniting genomic, transcriptomic and clinical data of 21 cancer types (n = 18,484), we interrogated copy number and transcript profiles of 32 circadian clock genes to identify putative loss-of-function (ClockLoss) and gain-of-function (ClockGain) players. Kaplan-Meier, Cox regression and receiver operating characteristic analyses were employed to evaluate the prognostic significance of both gene sets. RESULTS ClockLoss and ClockGain were associated with tumour-suppressing and tumour-promoting roles respectively. Downregulation of ClockLoss genes resulted in significantly higher mortality rates in five cancer cohorts (n = 2914): bladder (P = 0.027), glioma (P < 0.0001), pan-kidney (P = 0.011), clear cell renal cell (P < 0.0001) and stomach (P = 0.0007). In contrast, patients with high expression of oncogenic ClockGain genes had poorer survival outcomes (n = 2784): glioma (P < 0.0001), pan-kidney (P = 0.0034), clear cell renal cell (P = 0.014), lung (P = 0.046) and pancreas (P = 0.0059). Both gene sets were independent of other clinicopathological features to permit further delineation of tumours within the same stage. Circadian reprogramming of tumour genomes resulted in activation of numerous oncogenic pathways including those associated with cancer stem cells, suggesting that the circadian clock may influence self-renewal mechanisms. Within the hypoxic tumour microenvironment, circadian dysregulation is exacerbated by tumour hypoxia in glioma, renal, lung and pancreatic cancers, resulting in additional death risks. Tumour suppressive ClockLoss genes were negatively correlated with hypoxia inducible factor-1A targets in glioma patients, providing a novel framework for investigating the hypoxia-clock signalling axis. CONCLUSIONS Loss of timekeeping fidelity promotes tumour progression and influences clinical outcomes. ClockLoss and ClockGain may offer novel druggable targets for improving patient prognosis. Both gene sets can be used for patient stratification in adjuvant chronotherapy treatment. Emerging interactions between the circadian clock and hypoxia may be harnessed to achieve therapeutic advantage using hypoxia-modifying compounds in combination with first-line treatments.
Collapse
Affiliation(s)
- Wai Hoong Chang
- Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, OX3 7FZ UK
| | - Alvina G. Lai
- Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, OX3 7FZ UK
| |
Collapse
|
37
|
Cai T, Hua B, Luo D, Xu L, Cheng Q, Yuan G, Yan Z, Sun N, Hua L, Lu C. The circadian protein CLOCK regulates cell metabolism via the mitochondrial carrier SLC25A10. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1310-1321. [PMID: 30943427 DOI: 10.1016/j.bbamcr.2019.03.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/11/2019] [Accepted: 03/29/2019] [Indexed: 12/17/2022]
Abstract
Physiological function and metabolic regulation are the most important outputs of circadian clock controls in mammals. Mitochondrial respiration and ROS production show rhythmic activity. Mitochondrial carriers, which are responsible for mitochondrial substance transfer, are vital for mitochondrial metabolism. Clock (Circadian Locomotor Output Cycles Kaput) is the first core circadian gene identified in mammalian animals. However, whether CLOCK protein can regulate mitochondrial functions via mitochondrial carriers is unclear. Here, we showed that CLOCK can bind to the mitochondrial carrier SLC25A10. For further analysis, we established a Slc25a10-/--Hepa1-6 cell line using CRISPR/Cas9 gene-editing technology. Slc25a10-/--Hepa1-6 cells showed disordered glucose homeostasis, increased oxidative stress levels, and damaged electron transport chains. Next, using an immunoprecipitation assay, we found that amino acids 43-84 and 169-210 in SLC25A10 are key sites that respond to CLOCK binding. Finally, forced expression of wild-type SLC25A10 in Slc25a10-/--Hepa1-6 cells could compensate for the loss of SLC25A10; the decreased glucose metabolism, severe oxidative stress and damaged electron transport chain were recovered. In addition, a mutant Slc25a10 with changes in two key sites did not show a rescue effect. In conclusion, we identified a new protein-protein interaction mechanism in which CLOCK can directly regulate cell metabolism via the mitochondrial membrane transporter SLC25A10. Our study might provide some new insights into the relationship between circadian clock and mitochondrial metabolism.
Collapse
Affiliation(s)
- Tingting Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Bingxuan Hua
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dawei Luo
- Department of Ophthalmology, Shanghai First People's Hospital affiliated with Shanghai Jiaotong University, Shanghai, China
| | - Lirong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Qianyun Cheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Gongsheng Yuan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Zuoqin Yan
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ning Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Luchun Hua
- Department of Surgery, Huashan Hospital, Fudan University, Shanghai, China.
| | - Chao Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China; Research Center on Aging and Medicine, Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China.
| |
Collapse
|
38
|
Smith DF, Amin RS. OSA and Cardiovascular Risk in Pediatrics. Chest 2019; 156:402-413. [PMID: 30790552 DOI: 10.1016/j.chest.2019.02.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 01/21/2019] [Accepted: 02/08/2019] [Indexed: 02/07/2023] Open
Abstract
OSA occurs in approximately 1% to 5% of children in the United States. Long-term cardiovascular risks associated with OSA in the adult population are well documented. Although changes in BP regulation occur in children with OSA, the pathways leading to chronic cardiovascular risks of OSA in children are less clear. Risk factors associated with cardiovascular disease in adult populations could carry the same future risk for children. It is imperative to determine whether known mechanisms of cardiovascular diseases in adults are like those that lead to pediatric disease. Early pathophysiologic changes may lead to a lifetime burden of cardiovascular disease and early mortality. With this perspective in mind, our review discusses pathways leading to cardiovascular pathology in children with OSA and provides a comprehensive overview of recent research findings related to cardiovascular sequelae in the pediatric population.
Collapse
Affiliation(s)
- David F Smith
- Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Pulmonary and Sleep Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Otolaryngology-Head and Neck Surgery, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Raouf S Amin
- Division of Pulmonary and Sleep Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.
| |
Collapse
|
39
|
A splice variant of human Bmal1 acts as a negative regulator of the molecular circadian clock. Exp Mol Med 2018; 50:1-10. [PMID: 30523262 PMCID: PMC6283877 DOI: 10.1038/s12276-018-0187-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 12/04/2022] Open
Abstract
Bmal1 is one of the key molecules that controls the mammalian molecular clock. In humans, two isoforms of Bmal1 are generated by alternative RNA splicing. Unlike the extensively studied hBmal1b, the canonical form of Bmal1 in most species, the expression and/or function of another human-specific isoform, hBmal1a, are poorly understood. Due to the lack of the N-terminal nuclear localization signal (NLS), hBMAL1a does not enter the nucleus as hBMAL1b does. However, despite the lack of the NLS, hBMAL1a still dimerizes with either hCLOCK or hBMAL1b and thereby promotes cytoplasmic retention or protein degradation, respectively. Consequently, hBMAL1a interferes with hCLOCK:hBMAL1b-induced transcriptional activation and the circadian oscillation of Period2. Moreover, when the expression of endogenous hBmal1a is aborted by CRISPR/Cas9-mediated knockout, the rhythmic expression of hPer2 and hBmal1b is restored in cultured HeLa cells. Together, these results suggest a role for hBMAL1a as a negative regulator of the mammalian molecular clock. An alternative form of a key ‘clock’ protein involved in the maintenance of daily cellular rhythms serves as a negative regulator of the cell’s 24-hour cycle. A team led by Ilmin Kwon from Sungkyunkwan University School of Medicine, Suwon, and Kyungjin Kim from Daegu Gyeongbuk Institute of Science and Technology, both in South Korea, detailed the function of BMAL1a, a lesser-studied variant of the clock protein BMAL1b, in human cells. Whereas BMAL1b enters the nucleus, where it works in concert with another protein called CLOCK to control circadian dynamics, BMAL1a stays in the cytoplasm, where it binds BMAL1b and CLOCK, interfering with their function. Genetically inhibiting BMAL1a helped restore normal rhythmic cycles. Drugs targeting BMAL1a may thus aid in sleep disorders and other circadian-linked health problems.
Collapse
|
40
|
von Allmen DC, Francey LJ, Rogers GM, Ruben MD, Cohen AP, Wu G, Schmidt RE, Ishman SL, Amin RS, Hogenesch JB, Smith DF. Circadian Dysregulation: The Next Frontier in Obstructive Sleep Apnea Research. Otolaryngol Head Neck Surg 2018; 159:948-955. [PMID: 30200807 DOI: 10.1177/0194599818797311] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To review the effects of the circadian clock on homeostasis, the functional interaction between the circadian clock and hypoxia-inducible factors, and the role of circadian dysregulation in the progression of cardiopulmonary disease in obstructive sleep apnea (OSA). DATA SOURCES The MEDLINE database was accessed through PubMed. REVIEW METHODS A general review is presented on molecular pathways disrupted in OSA, circadian rhythms and the role of the circadian clock, hypoxia signaling, crosstalk between the circadian and hypoxia systems, the role of the circadian clock in cardiovascular disease, and implications for practice. Studies included in this State of the Art Review demonstrate the potential contribution of the circadian clock and hypoxia in animal models or human disease. CONCLUSIONS Molecular crosstalk between the circadian clock and hypoxia-inducible factors has not been evaluated in disease models of OSA. IMPLICATIONS FOR PRACTICE Pediatric OSA is highly prevalent and, if left untreated, may lead to cardiopulmonary sequelae. Changes in inflammatory markers that normally demonstrate circadian rhythmicity are also seen among patients with OSA. Hypoxia-inducible transcription factors interact with core circadian clock transcription factors; however, the interplay between these pathways has not been elucidated in the cardiopulmonary system. This gap in knowledge hinders our ability to identify potential biomarkers of OSA and develop alternative therapeutic strategies. A deeper understanding of the mechanisms by which OSA impinges on clock function and the impact of clock dysregulation on the cardiopulmonary system may lead to future advancements for the care of patients with OSA. The aim of this review is to shed light on this important clinical topic.
Collapse
Affiliation(s)
- Douglas C von Allmen
- 1 Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lauren J Francey
- 2 Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Garrett M Rogers
- 3 College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Marc D Ruben
- 2 Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Aliza P Cohen
- 4 Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Gang Wu
- 2 Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Robert E Schmidt
- 2 Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Stacey L Ishman
- 1 Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
- 4 Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- 5 Division of Pulmonary and Sleep Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Raouf S Amin
- 5 Division of Pulmonary and Sleep Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- 6 Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - John B Hogenesch
- 2 Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- 6 Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - David F Smith
- 1 Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
- 4 Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- 5 Division of Pulmonary and Sleep Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
41
|
Modeling Reveals a Key Mechanism for Light-Dependent Phase Shifts of Neurospora Circadian Rhythms. Biophys J 2018; 115:1093-1102. [PMID: 30139524 DOI: 10.1016/j.bpj.2018.07.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 07/11/2018] [Accepted: 07/24/2018] [Indexed: 12/14/2022] Open
Abstract
Light shifts and synchronizes the phase of the circadian clock to daily environments, which is critical for maintaining the daily activities of an organism. It has been proposed that such light-dependent phase shifts are triggered by light-induced upregulation of a negative element of the core circadian clock (i.e., frq, Per1/2) in many organisms, including fungi. However, we find, using systematic mathematical modeling of the Neurospora crassa circadian clock, that the upregulation of the frq gene expression alone is unable to reproduce the observed light-dependent phase responses. Indeed, we find that the depression of the transcriptional activator white-collar-1, previously shown to be promoted by FRQ and VVD, is a key molecular mechanism for accurately simulating light-induced phase response curves for wild-type and mutant strains of Neurospora. Our findings elucidate specific molecular pathways that can be utilized to control phase resetting of circadian rhythms.
Collapse
|
42
|
Hida A, Kitamura S, Kadotani H, Uchiyama M, Ebisawa T, Inoue Y, Kamei Y, Mishima K. Lack of association between PER3 variable number tandem repeat and circadian rhythm sleep-wake disorders. Hum Genome Var 2018; 5:17. [PMID: 30083361 PMCID: PMC6043536 DOI: 10.1038/s41439-018-0017-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 05/27/2018] [Indexed: 12/02/2022] Open
Abstract
Circadian rhythm sleep–wake disorders (CRSWDs) are characterized by disturbed sleep–wake patterns. We genotyped a PER3 variable number tandem repeat (VNTR) in 248 CRSWD individuals and 925 controls and found no significant association between the VNTR and CRSWDs or morningness–eveningness (diurnal) preferences in the Japanese population. Although the VNTR has been associated with circadian and sleep phenotypes in some other populations, the polymorphism may not be a universal genetic marker.
Collapse
Affiliation(s)
- Akiko Hida
- 1Department of Sleep-Wake Disorders, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, 187-8553 Japan
| | - Shingo Kitamura
- 1Department of Sleep-Wake Disorders, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, 187-8553 Japan
| | - Hiroshi Kadotani
- 2Department of Sleep and Behavioral Sciences, Shiga University of Medical Science, Shiga, 520-2192 Japan
| | - Makoto Uchiyama
- 3Department of Psychiatry, Nihon University School of Medicine, Tokyo, 173-8610 Japan
| | - Takashi Ebisawa
- Yokohama Clinic for Psychosomatic Medicine and Psychiatry, Medical Corporation Warakukai, Kanagawa, 220-0004 Japan
| | - Yuichi Inoue
- 5Department of Somnology, Tokyo Medical University, Tokyo, 160-8402 Japan.,Yoyogi Sleep Disorder Center, Tokyo, 151-0053 Japan
| | - Yuichi Kamei
- 7Department of Laboratory Medicine, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, 187-8551 Japan
| | - Kazuo Mishima
- 1Department of Sleep-Wake Disorders, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, 187-8553 Japan
| |
Collapse
|
43
|
Ao Y, Zhao Q, Yang K, Zheng G, Lv X, Su X. A role for the clock period circadian regulator 2 gene in regulating the clock gene network in human oral squamous cell carcinoma cells. Oncol Lett 2018. [PMID: 29541184 PMCID: PMC5835870 DOI: 10.3892/ol.2018.7825] [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] [Indexed: 12/05/2022] Open
Abstract
Clock genes are the core of the circadian rhythms in the human body and are important in regulating normal physiological functions. To date, research has indicated that the clock gene, period circadian clock 2 (PER2), is downregulated in numerous types of cancer, and that it is associated with cancer occurrence and progression via the regulation of various downstream cell cycle genes. However, it remains unclear whether the decreased expression of PER2 influences the expression of other clock genes in cancer cells. In the present study, short hairpin RNA interference was used to knockdown PER2 effectively in human oral squamous cell carcinoma SCC15 cells. Quantitative polymerase chain reaction was used to assess the mRNA expression levels of various clock genes and revealed that, following the knockdown of PER2 in SCC15 cells, the mRNA expression levels of PER3, brain and muscle ARNT-like 1, deleted in esophageal cancer (DEC)1, DEC2, cryptochrome circadian clock (CRY)2, timeless circadian clock, retinoic acid receptor-related orphan receptor-alpha and neuronal PAS domain protein 2 were significantly downregulated, while the mRNA expression levels of PER1 and nuclear receptor subfamily 1 group D member 1 were significantly upregulated. In addition, flow cytometric analysis demonstrated that proliferation was enhanced and apoptosis was reduced following PER2 knockdown in SCC15 cells (P<0.05). To the best of our knowledge, the present study is the first to report that PER2 is important for the regulation of other clock genes of the clock gene network in cancer cells. This is of great significance in elucidating the molecular function and tumor suppression mechanism of PER2.
Collapse
Affiliation(s)
- Yiran Ao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Qin Zhao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Kai Yang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Gang Zheng
- Anorectal Department, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, P.R. China
| | - Xiaoqing Lv
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiaoli Su
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
44
|
Mayeuf-Louchart A, Zecchin M, Staels B, Duez H. Circadian control of metabolism and pathological consequences of clock perturbations. Biochimie 2017; 143:42-50. [DOI: 10.1016/j.biochi.2017.07.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/31/2017] [Indexed: 01/08/2023]
|
45
|
Ozburn AR, Kern J, Parekh PK, Logan RW, Liu Z, Falcon E, Becker-Krail D, Purohit K, Edgar NM, Huang Y, McClung CA. NPAS2 Regulation of Anxiety-Like Behavior and GABAA Receptors. Front Mol Neurosci 2017; 10:360. [PMID: 29163035 PMCID: PMC5675889 DOI: 10.3389/fnmol.2017.00360] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/19/2017] [Indexed: 12/26/2022] Open
Abstract
Abnormal circadian rhythms and circadian genes are strongly associated with several psychiatric disorders. Neuronal PAS Domain Protein 2 (NPAS2) is a core component of the molecular clock that acts as a transcription factor and is highly expressed in reward- and stress-related brain regions such as the striatum. However, the mechanism by which NPAS2 is involved in mood-related behaviors is still unclear. We measured anxiety-like behaviors in mice with a global null mutation in Npas2 (Npas2 null mutant mice) and found that Npas2 null mutant mice exhibit less anxiety-like behavior than their wild-type (WT) littermates (in elevated plus maze, light/dark box and open field assay). We assessed the effects of acute or chronic stress on striatal Npas2 expression, and found that both stressors increased levels of Npas2. Moreover, knockdown of Npas2 in the ventral striatum resulted in a similar reduction of anxiety-like behaviors as seen in the Npas2 null mutant mouse. Additionally, we identified Gabra genes as transcriptional targets of NPAS2, found that Npas2 null mutant mice exhibit reduced sensitivity to the GABAa positive allosteric modulator, diazepam and that knockdown of Npas2 reduced Gabra1 expression and response to diazepam in the ventral striatum. These results: (1) implicate Npas2 in the response to stress and the development of anxiety; and (2) provide functional evidence for the regulation of GABAergic neurotransmission by NPAS2 in the ventral striatum.
Collapse
Affiliation(s)
- Angela R Ozburn
- Portland Veterans Affairs Medical Center, Research and Development Service, Portland, OR, United States.,Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States.,Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Joseph Kern
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Puja K Parekh
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Ryan W Logan
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Zheng Liu
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Edgardo Falcon
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Darius Becker-Krail
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Kush Purohit
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Nicole M Edgar
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Yanhua Huang
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Colleen A McClung
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| |
Collapse
|
46
|
Distribution of tyrosine hydroxylase-immunoreactive neurons in the brain of the viviparous fish Gambusia affinis. J Chem Neuroanat 2017; 85:1-12. [DOI: 10.1016/j.jchemneu.2017.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/14/2017] [Accepted: 05/19/2017] [Indexed: 11/18/2022]
|
47
|
Mark PJ, Crew RC, Wharfe MD, Waddell BJ. Rhythmic Three-Part Harmony: The Complex Interaction of Maternal, Placental and Fetal Circadian Systems. J Biol Rhythms 2017; 32:534-549. [PMID: 28920512 DOI: 10.1177/0748730417728671] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
From the perspective of circadian biology, mammalian pregnancy presents an unusual biological scenario in which an entire circadian system (i.e., that of the fetus) is embodied within another (i.e., that of the mother). Moreover, both systems are likely to be influenced at their interface by a third player, the placenta. Successful pregnancy requires major adaptations in maternal physiology, many of which involve circadian changes that support the high metabolic demands of the growing fetus. A functional role for maternal circadian adaptations is implied by the effects of circadian disruption, which result in pregnancy complications including higher risks for miscarriage, preterm labor, and low birth weight. Various aspects of fetal physiology lead to circadian variation, at least in late gestation, but it remains unclear what drives this rhythmicity. It likely involves contributions from the maternal environment and possibly from the placenta and the developing intrinsic molecular clocks within fetal tissues. The role of the placenta is of particular significance because it serves not only to relay signals about the external environment (via the mother) but may also exhibit its own circadian rhythmicity. This review considers how the fetus may be influenced by dynamic circadian signals from the mother and the placenta during gestation, and how, in the face of these changing influences, a new fetal circadian system emerges. Particular emphasis is placed on the role of endocrine signals, most notably melatonin and glucocorticoids, as mediators of maternal-fetal circadian interactions, and on the expression of the clock gene in the 3 compartments. Further study is required to understand how the mother, placenta, and fetus interact across pregnancy to optimize circadian adaptations that support adequate growth and development of the fetus and its transition to postnatal life in a circadian environment.
Collapse
Affiliation(s)
- Peter J Mark
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Rachael C Crew
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Michaela D Wharfe
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Brendan J Waddell
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
48
|
Williams CT, Barnes BM, Yan L, Buck CL. Entraining to the polar day: circadian rhythms in arctic ground squirrels. ACTA ACUST UNITED AC 2017. [PMID: 28623226 DOI: 10.1242/jeb.159889] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Circadian systems are principally entrained to 24 h light-dark cycles, but this cue is seasonally absent in polar environments. Although some resident polar vertebrates have weak circadian clocks and are seasonally arrhythmic, the arctic ground squirrel (AGS) maintains daily rhythms of physiology and behavior throughout the summer, which includes 6 weeks of constant daylight. Here, we show that persistent daily rhythms in AGS are maintained through a circadian system that readily entrains to the polar day yet remains insensitive to entrainment by rapid light-dark transitions, which AGS generate naturally as a consequence of their semi-fossorial behavior. Additionally, AGS do not show 'jet lag', the slow realignment of circadian rhythms induced by the inertia of an intrinsically stable master circadian clock in the suprachiasmatic nucleus (SCN). We suggest this is due to the low expression of arginine vasopressin in the SCN of AGS, as vasopressin is associated with inter-neuronal coupling and robust rhythmicity.
Collapse
Affiliation(s)
- Cory T Williams
- Department of Biological Sciences and Center for Bioengineering Innovation, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Brian M Barnes
- Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK 99775, USA
| | - Lily Yan
- Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
| | - C Loren Buck
- Department of Biological Sciences and Center for Bioengineering Innovation, Northern Arizona University, Flagstaff, AZ 86011, USA
| |
Collapse
|
49
|
Kurupati RK, Kossenkoff A, Kannan S, Haut LH, Doyle S, Yin X, Schmader KE, Liu Q, Showe L, Ertl HCJ. The effect of timing of influenza vaccination and sample collection on antibody titers and responses in the aged. Vaccine 2017; 35:3700-3708. [PMID: 28583307 DOI: 10.1016/j.vaccine.2017.05.074] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 04/25/2017] [Accepted: 05/23/2017] [Indexed: 02/07/2023]
Abstract
Antibody responses, B cell subset distribution in blood and the blood transcriptome were analyzed in younger and aged human subjects before and after vaccination with the inactivated influenza vaccine. In the aged, but not the younger, individuals we saw a clear difference in antibody titers including those at baseline depending on the time of vaccination and sample collection. Differences in baseline titers in aged individuals treated in the morning or afternoon in turn affected responsiveness to the vaccine. In both younger and aged individuals, the time of sample collection also affected relative numbers of some of the B cell subsets in blood. A global gene expression analysis with whole blood samples from the aged showed small but statistically significant differences depending on the time of sample collection. Our data do not indicate that timing of vaccination affects immune responsiveness of the aged, but rather shows that in clinical influenza vaccine trials timing of collection of samples can have a major and potentially misleading influence on study outcome. In future vaccine trials, timing of vaccination and sample collection should be recorded carefully to allow for its use as a study covariant.
Collapse
Affiliation(s)
| | | | - Senthil Kannan
- The Wistar Institute, Philadelphia, PA, USA; Biomedical Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Susan Doyle
- GRECC, Durham VA Medical Center and Center for the Study of Aging and Human, Development and Division of Geriatrics, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | | | - Kenneth E Schmader
- GRECC, Durham VA Medical Center and Center for the Study of Aging and Human, Development and Division of Geriatrics, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Qin Liu
- The Wistar Institute, Philadelphia, PA, USA
| | | | | |
Collapse
|
50
|
Mange F, Praz V, Migliavacca E, Willis IM, Schütz F, Hernandez N. Diurnal regulation of RNA polymerase III transcription is under the control of both the feeding-fasting response and the circadian clock. Genome Res 2017; 27:973-984. [PMID: 28341772 PMCID: PMC5453330 DOI: 10.1101/gr.217521.116] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/17/2017] [Indexed: 11/25/2022]
Abstract
RNA polymerase III (Pol III) synthesizes short noncoding RNAs, many of which are essential for translation. Accordingly, Pol III activity is tightly regulated with cell growth and proliferation by factors such as MYC, RB1, TRP53, and MAF1. MAF1 is a repressor of Pol III transcription whose activity is controlled by phosphorylation; in particular, it is inactivated through phosphorylation by the TORC1 kinase complex, a sensor of nutrient availability. Pol III regulation is thus sensitive to environmental cues, yet a diurnal profile of Pol III transcription activity is so far lacking. Here, we first use gene expression arrays to measure mRNA accumulation during the diurnal cycle in the livers of (1) wild-type mice, (2) arrhythmic Arntl knockout mice, (3) mice fed at regular intervals during both night and day, and (4) mice lacking the Maf1 gene, and so provide a comprehensive view of the changes in cyclic mRNA accumulation occurring in these different systems. We then show that Pol III occupancy of its target genes rises before the onset of the night, stays high during the night, when mice normally ingest food and when translation is known to be increased, and decreases in daytime. Whereas higher Pol III occupancy during the night reflects a MAF1-dependent response to feeding, the rise of Pol III occupancy before the onset of the night reflects a circadian clock-dependent response. Thus, Pol III transcription during the diurnal cycle is regulated both in response to nutrients and by the circadian clock, which allows anticipatory Pol III transcription.
Collapse
Affiliation(s)
- François Mange
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Viviane Praz
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Eugenia Migliavacca
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Ian M Willis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Frédéric Schütz
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
- Bioinformatics Core Facility, Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Nouria Hernandez
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|