1
|
Horiguchi M, Yoshihara K, Watanabe K, Tsurudome Y, Mizukami Y, Ushijima K. Circadian Rhythms of Clock Genes After Transplantation of Mesenchymal Stem Cells with Type 2 Diabetes Mellitus. Int J Mol Sci 2024; 25:13145. [PMID: 39684854 DOI: 10.3390/ijms252313145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Regenerative therapy involving stem cell transplantation has become an option for the radical treatment of diabetes mellitus. Disruption in the clock genes of stem cells affects the homeostasis of transplanted tissues. We examined the circadian rhythm of genes in transplanted adipose-derived mesenchymal stem cells derived from a patient with type 2 diabetes mellitus (T2DM-ADSC). The clock genes (PER2, CLOCK1, CRY1, and ARNTL[BMAL1]) exhibited similar daily fluctuations in phase and amplitude between a group transplanted with adipose-derived mesenchymal stem cells derived from a healthy individual (N-ADSC) and a group transplanted with T2DM-ADSC. The findings demonstrated that clock genes in stem cells are synchronized with those in living organisms. Next-generation sequencing was then employed to categorize genes that exhibited variation in expression between N-ADSC and T2DM-ADSC. MTATP8P1 and NDUFA7_2 gene expression was significantly reduced at two time points (ZT6 and ZT18), and daily fluctuations were lost. The present study reports, for the first time, that the circadian rhythms of MTATP8P1 and NDUFA7_2, genes involved in mitochondrial processes, are altered in T2DM-ADSC.
Collapse
Affiliation(s)
- Michiko Horiguchi
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan
| | - Kenichi Yoshihara
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan
| | - Kenji Watanabe
- Institute of Gene Research, Yamaguchi University Science Research Center, Yamaguchi 753-0841, Japan
| | - Yuya Tsurudome
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan
| | - Yoichi Mizukami
- Institute of Gene Research, Yamaguchi University Science Research Center, Yamaguchi 753-0841, Japan
| | - Kentaro Ushijima
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan
| |
Collapse
|
2
|
Ding H, Meng L, Zhang Y, Bryant AJ, Xing C, Esser KA, Chen L, Huo Z. A Bayesian Framework for Genome-wide Circadian Rhythmicity Biomarker Detection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620703. [PMID: 39554018 PMCID: PMC11565714 DOI: 10.1101/2024.10.28.620703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Circadian rhythms are endogenous ∼24-hour cycles that significantly influence physiological and behavioral processes. These rhythms are governed by a transcriptional-translational feedback loop of core circadian genes and are essential for maintaining overall health. The study of circadian rhythms has expanded into various omics datasets, necessitating accurate analytical methodology for circadian biomarker detection. Here, we introduce a novel Bayesian framework for the genome-wide detection of circadian rhythms that is capable of incorporating prior biological knowledge and adjusting for multiple testing issue via a false discovery rate approach. Our framework leverages a Bayesian hierarchical model and employs a reverse jump Markov chain Monte Carlo (rjMCMC) technique for model selection. Through extensive simulations, our method, BayesCircRhy, demonstrated superior false discovery rate control over competing methods, robustness against heavier-tailed error distributions, and better performance compared to existing approaches. The method's efficacy was further validated in two RNA-Sequencing data, including a human resitrcted feeding data and a mouse aging data, where it successfully identified known and novel circadian genes. R package "BayesianCircadian" for the method is publicly available on GitHub https://github.com/jxncdhc/BayesianCircadian .
Collapse
|
3
|
Zhu X, Han X, Li Z, Zhou X, Yoo SH, Chen Z, Ji Z. CircaKB: a comprehensive knowledgebase of circadian genes across multiple species. Nucleic Acids Res 2024:gkae817. [PMID: 39329269 DOI: 10.1093/nar/gkae817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/30/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024] Open
Abstract
Circadian rhythms, which are the natural cycles that dictate various physiological processes over a 24-h period, have been increasingly recognized as important in the management and treatment of various human diseases. However, the lack of sufficient data and reliable analysis methods have been a major obstacle to understanding the bidirectional interaction between circadian variation and human health. We have developed CircaKB, a comprehensive knowledgebase of circadian genes across multiple species. CircaKB is the first knowledgebase that provides systematic annotations of the oscillatory patterns of gene expression at a genome-wide level for 15 representative species. Currently, CircaKB contains 226 time-course transcriptome datasets, covering a wide variety of tissues, organs, and cell lines. In addition, CircaKB integrates 12 computational models to facilitate reliable data analysis and identify oscillatory patterns and their variations in gene expression. CircaKB also offers powerful functionalities to its users, including easy search, fast browsing, strong visualization, and custom upload. We believe that CircaKB will be a valuable tool and resource for the circadian research community, contributing to the identification of new targets for disease prevention and treatment. We have made CircaKB freely accessible at https://cdsic.njau.edu.cn/CircaKB.
Collapse
Affiliation(s)
- Xingchen Zhu
- College of Artificial Intelligence, Nanjing Agricultural University, No. 1 Weigang Rd., Nanjing, Jiangsu 210095, China
- Center for Data Science and Intelligent Computing, Nanjing Agricultural University, No. 1 Weigang Rd., Nanjing, Jiangsu 210095, China
| | - Xiao Han
- College of Artificial Intelligence, Nanjing Agricultural University, No. 1 Weigang Rd., Nanjing, Jiangsu 210095, China
- Center for Data Science and Intelligent Computing, Nanjing Agricultural University, No. 1 Weigang Rd., Nanjing, Jiangsu 210095, China
| | - Zhijin Li
- Department of Neurosurgery, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Xiaobo Zhou
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Houston, TX 77030, USA
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhiwei Ji
- College of Artificial Intelligence, Nanjing Agricultural University, No. 1 Weigang Rd., Nanjing, Jiangsu 210095, China
- Center for Data Science and Intelligent Computing, Nanjing Agricultural University, No. 1 Weigang Rd., Nanjing, Jiangsu 210095, China
| |
Collapse
|
4
|
Ferrell JM. Chronobiology of Cancers in the Liver and Gut. Cancers (Basel) 2024; 16:2925. [PMID: 39272783 PMCID: PMC11394324 DOI: 10.3390/cancers16172925] [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: 07/29/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Circadian rhythms dictate the timing of cellular and organismal physiology to maintain homeostasis. Within the liver and gut, circadian rhythms influence lipid and glucose homeostasis, xenobiotic metabolism, and nutrient absorption. Disruption of this orchestrated timing is known to negatively impact human health and contribute to disease progression, including carcinogenesis. Dysfunctional core clock timing has been identified in malignant growths and may be used as a molecular signature of disease progression. Likewise, the circadian clock and its downstream effectors also represent potential for novel therapeutic targets. Here, the role of circadian rhythms in the pathogenesis of cancers of the liver and gut will be reviewed, and chronotherapy and chronopharmacology will be explored as potential treatment options.
Collapse
Affiliation(s)
- Jessica M Ferrell
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| |
Collapse
|
5
|
Kuo LT, Lu HY, Chen YH. Traumatic brain injury-induced disruption of the circadian clock. J Mol Med (Berl) 2024; 102:403-414. [PMID: 38285094 PMCID: PMC10879350 DOI: 10.1007/s00109-024-02416-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 12/16/2023] [Accepted: 01/03/2024] [Indexed: 01/30/2024]
Abstract
Disturbances in the circadian rhythm have been reported in patients following traumatic brain injury (TBI). However, the rhythmic expression of circadian genes in peripheral blood leukocytes (PBL) following TBI has not yet been studied. The messenger ribonucleic acid (mRNA) expression of period 1 (Per1), Per2, Per3, cryptochrome 1 (Cry1), Cry2, brain and muscle aryl hydrocarbon receptor nuclear translocator-like 1 (Bmal1), and circadian locomotor output cycles kaput (Clock) was quantified in PBLs from sham-operated rats and rats with acute subdural hematoma (ASDH) over a 48-h period. The rectal temperature of the animals was measured every 4 h over 2 days. The mesor, rhythm, amplitude, and acrophase were estimated using cosinor analysis. Cosinor analysis revealed that Per2, Cry1, and Bmal1 mRNAs were rhythmically expressed in the PBLs of sham-operated rats. In contrast, fluctuations in rhythmic expression were not observed following ASDH. The rectal temperature of sham-operated rats also exhibited rhythmicity. ASDH rats had a disrupted rectal temperature rhythm, a diminished amplitude, and an acrophase shift. TBI with ASDH results in dysregulated expression of some circadian genes and changes in body temperature rhythm. Further research is required to understand the pathophysiology of altered circadian networks following TBI. KEY MESSAGES: First to investigate the mRNA expression of circadian genes in PBLs of ASDH rats. ASDH rats had disrupted rhythmicity of Per2, Cry1, and Bmal1 mRNA expression. Cosinor analysis showed that ASDH rats had a disrupted rectal temperature rhythm.
Collapse
Affiliation(s)
- Lu-Ting Kuo
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, 7 Chun-Shan South Road, Taipei, 100, Taiwan.
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Yun-Lin Branch, Douliu, Yunlin, 640, Taiwan.
| | - Hsueh-Yi Lu
- Department of Industrial Engineering and Management, National Yunlin University of Science and Technology, Douliu, Yunlin, 640, Taiwan
| | - Yi-Hsing Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, 7 Chun-Shan South Road, Taipei, 100, Taiwan
| |
Collapse
|
6
|
Engin A. Misalignment of Circadian Rhythms in Diet-Induced Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:27-71. [PMID: 39287848 DOI: 10.1007/978-3-031-63657-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The biological clocks of the circadian timing system coordinate cellular and physiological processes and synchronize them with daily cycles. While the central clock in the suprachiasmatic nucleus (SCN) is mainly synchronized by the light/dark cycles, the peripheral clocks react to other stimuli, including the feeding/fasting state, nutrients, sleep-wake cycles, and physical activity. During the disruption of circadian rhythms due to genetic mutations or social and occupational obligations, incorrect arrangement between the internal clock system and environmental rhythms leads to the development of obesity. Desynchronization between the central and peripheral clocks by altered timing of food intake and diet composition leads to uncoupling of the peripheral clocks from the central pacemaker and to the development of metabolic disorders. The strong coupling of the SCN to the light-dark cycle creates a situation of misalignment when food is ingested during the "wrong" time of day. Food-anticipatory activity is mediated by a self-sustained circadian timing, and its principal component is a food-entrainable oscillator. Modifying the time of feeding alone greatly affects body weight, whereas ketogenic diet (KD) influences circadian biology, through the modulation of clock gene expression. Night-eating behavior is one of the causes of circadian disruption, and night eaters have compulsive and uncontrolled eating with severe obesity. By contrast, time-restricted eating (TRE) restores circadian rhythms through maintaining an appropriate daily rhythm of the eating-fasting cycle. The hypothalamus has a crucial role in the regulation of energy balance rather than food intake. While circadian locomotor output cycles kaput (CLOCK) expression levels increase with high-fat diet-induced obesity, peroxisome proliferator-activated receptor-alpha (PPARα) increases the transcriptional level of brain and muscle aryl hydrocarbon receptor nuclear translocator (ARNT)-like 1 (BMAL1) in obese subjects. In this context, effective timing of chronotherapies aiming to correct SCN-driven rhythms depends on an accurate assessment of the SCN phase. In fact, in a multi-oscillator system, local rhythmicity and its disruption reflects the disruption of either local clocks or central clocks, thus imposing rhythmicity on those local tissues, whereas misalignment of peripheral oscillators is due to exosome-based intercellular communication.Consequently, disruption of clock genes results in dyslipidemia, insulin resistance, and obesity, while light exposure during the daytime, food intake during the daytime, and sleeping during the biological night promote circadian alignment between the central and peripheral clocks. Thus, shift work is associated with an increased risk of obesity, diabetes, and cardiovascular diseases because of unusual eating times as well as unusual light exposure and disruption of the circadian rhythm.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
7
|
Zong W, Seney ML, Ketchesin KD, Gorczyca MT, Liu AC, Esser KA, Tseng GC, McClung CA, Huo Z. Experimental design and power calculation in omics circadian rhythmicity detection using the cosinor model. Stat Med 2023; 42:3236-3258. [PMID: 37265194 PMCID: PMC10425922 DOI: 10.1002/sim.9803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/27/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023]
Abstract
Circadian clocks are 24-h endogenous oscillators in physiological and behavioral processes. Though recent transcriptomic studies have been successful in revealing the circadian rhythmicity in gene expression, the power calculation for omics circadian analysis have not been fully explored. In this paper, we develop a statistical method, namely CircaPower, to perform power calculation for circadian pattern detection. Our theoretical framework is determined by three key factors in circadian gene detection: sample size, intrinsic effect size and sampling design. Via simulations, we systematically investigate the impact of these key factors on circadian power calculation. We not only demonstrate that CircaPower is fast and accurate, but also show its underlying cosinor model is robust against variety of violations of model assumptions. In real applications, we demonstrate the performance of CircaPower using mouse pan-tissue data and human post-mortem brain data, and illustrate how to perform circadian power calculation using mouse skeleton muscle RNA-Seq pilot as case study. Our method CircaPower has been implemented in an R package, which is made publicly available on GitHub ( https://github.com/circaPower/circaPower).
Collapse
Affiliation(s)
- Wei Zong
- Department of Biostatistics, University of Pittsburgh, PA, USA
| | - Marianne L. Seney
- Translational Neuroscience Program, Department of Psychiatry, Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Kyle D. Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Michael T. Gorczyca
- Department of Computational and Systems Biology, University of Pittsburgh, PA, USA
| | - Andrew C. Liu
- Department of Physiology and Aging, University of Florida, FL, USA
| | - Karyn A. Esser
- Department of Physiology and Aging, University of Florida, FL, USA
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, PA, USA
| | - Colleen A. McClung
- Translational Neuroscience Program, Department of Psychiatry, Center for Neuroscience, University of Pittsburgh, PA, USA
| | - Zhiguang Huo
- Department of Biostatistics, University of Florida, FL, USA
| |
Collapse
|
8
|
Silva MSB, Navarro VM, Chachlaki K. Editorial: The neuroendocrine female brain: from normal reproductive function to disease. Front Neurosci 2023; 17:1243349. [PMID: 37521681 PMCID: PMC10374340 DOI: 10.3389/fnins.2023.1243349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 08/01/2023] Open
Affiliation(s)
- Mauro S. B. Silva
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Victor M. Navarro
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Konstantina Chachlaki
- Université de Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille, France
- FHU 1000 Days for Health, School of Medicine, Lille, France
- University Research Institute of Child Health and Precision Medicine, National and Kapodistrian University of Athens, “Aghia Sophia” Children's Hospital, Athens, Greece
| |
Collapse
|
9
|
Makrygianni EA, Chrousos GP. Neural Progenitor Cells and the Hypothalamus. Cells 2023; 12:1822. [PMID: 37508487 PMCID: PMC10378393 DOI: 10.3390/cells12141822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 07/30/2023] Open
Abstract
Neural progenitor cells (NPCs) are multipotent neural stem cells (NSCs) capable of self-renewing and differentiating into neurons, astrocytes and oligodendrocytes. In the postnatal/adult brain, NPCs are primarily located in the subventricular zone (SVZ) of the lateral ventricles (LVs) and subgranular zone (SGZ) of the hippocampal dentate gyrus (DG). There is evidence that NPCs are also present in the postnatal/adult hypothalamus, a highly conserved brain region involved in the regulation of core homeostatic processes, such as feeding, metabolism, reproduction, neuroendocrine integration and autonomic output. In the rodent postnatal/adult hypothalamus, NPCs mainly comprise different subtypes of tanycytes lining the wall of the 3rd ventricle. In the postnatal/adult human hypothalamus, the neurogenic niche is constituted by tanycytes at the floor of the 3rd ventricle, ependymal cells and ribbon cells (showing a gap-and-ribbon organization similar to that in the SVZ), as well as suprachiasmatic cells. We speculate that in the postnatal/adult human hypothalamus, neurogenesis occurs in a highly complex, exquisitely sophisticated neurogenic niche consisting of at least four subniches; this structure has a key role in the regulation of extrahypothalamic neurogenesis, and hypothalamic and extrahypothalamic neural circuits, partly through the release of neurotransmitters, neuropeptides, extracellular vesicles (EVs) and non-coding RNAs (ncRNAs).
Collapse
Affiliation(s)
- Evanthia A Makrygianni
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
10
|
Allgood JE, Roe A, Sparks BB, Castillo M, Cruz A, Brooks AE, Brooks BD. The Correlation of Sleep Disturbance and Location of Glioma Tumors: A Narrative Review. J Clin Med 2023; 12:4058. [PMID: 37373751 DOI: 10.3390/jcm12124058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Sleep disturbance can occur when sleep centers of the brain, regions that are responsible for coordinating and generating healthy amounts of sleep, are disrupted by glioma growth or surgical resection. Several disorders cause disruptions to the average duration, quality, or patterns of sleep, resulting in sleep disturbance. It is unknown whether specific sleep disorders can be reliably correlated with glioma growth, but there are sufficient numbers of case reports to suggest that a connection is possible. In this manuscript, these case reports and retrospective chart reviews are considered in the context of the current primary literature on sleep disturbance and glioma diagnosis to identify a new and useful connection which warrants further systematic and scientific examination in preclinical animal models. Confirmation of the relationship between disruption of the sleep centers in the brain and glioma location could have significant implications for diagnostics, treatment, monitoring of metastasis/recurrence, and end-of-life considerations.
Collapse
Affiliation(s)
- JuliAnne E Allgood
- Department of Neuroscience, University of Wyoming, Laramie, WY 82071, USA
| | - Avery Roe
- College of Osteopathic Medicine, Rocky Vista University, Greenwood Village, CO 80112, USA
| | - Bridger B Sparks
- Department of Neuroscience, University of Wyoming, Laramie, WY 82071, USA
| | - Mercedes Castillo
- College of Osteopathic Medicine, Rocky Vista University, Greenwood Village, CO 80112, USA
| | - Angel Cruz
- College of Osteopathic Medicine, Rocky Vista University, Greenwood Village, CO 80112, USA
| | - Amanda E Brooks
- College of Osteopathic Medicine, Rocky Vista University, Greenwood Village, CO 80112, USA
| | - Benjamin D Brooks
- College of Osteopathic Medicine, Rocky Vista University, Greenwood Village, CO 80112, USA
| |
Collapse
|
11
|
Li X, Sun Z. Circadian clock and temporal meal pattern. MEDICAL REVIEW (2021) 2023; 3:85-101. [PMID: 37724110 PMCID: PMC10471112 DOI: 10.1515/mr-2022-0021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/02/2022] [Indexed: 09/20/2023]
Abstract
The central circadian clock in the brain controls the time-of-the-day variations in acute meal responses, with a low glycemic response but a high satiety/thermogenic response to meals consumed at waking compared to other time points. Consistently, studies show that consuming a significant proportion of calories, particularly carbohydrates, in breakfast is beneficial for the chronic management of obesity and its associated metabolic syndrome, compared to consuming identical meals at dinner. Conversely, breakfast skipping or/and late dinner can have unfavorable metabolic outcomes. It remains controversial how meal frequency affects metabolic health. In contrast, irregular meals, especially irregular breakfasts, show consistent adverse metabolic consequences. Time-restricted feeding (TRF), with all calories consumed within less than 12-h per day, can improve metabolism and extend lifespan. A major component of TRF in humans is caloric restriction, which contributes significantly to the beneficial effects of TRF in humans. By comparison, TRF effects in rodents can be independent of caloric restriction and show day/night phase specificity. TRF could alleviate metabolic abnormalities due to circadian disruption, but its effects appear independent of the circadian clock in rodents. Understanding neuroendocrine mechanisms underlying clock-mediated metabolic regulation will shed light on the metabolic effects of temporal meal patterns.
Collapse
Affiliation(s)
- Xin Li
- Department of Medicine – Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Zheng Sun
- Department of Medicine – Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
12
|
Zheng Y, Pan L, Wang F, Yan J, Wang T, Xia Y, Yao L, Deng K, Zheng Y, Xia X, Su Z, Chen H, Lin J, Ding Z, Zhang K, Zhang M, Chen Y. Neural function of Bmal1: an overview. Cell Biosci 2023; 13:1. [PMID: 36593479 PMCID: PMC9806909 DOI: 10.1186/s13578-022-00947-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023] Open
Abstract
Bmal1 (Brain and muscle arnt-like, or Arntl) is a bHLH/PAS domain transcription factor central to the transcription/translation feedback loop of the biologic clock. Although Bmal1 is well-established as a major regulator of circadian rhythm, a growing number of studies in recent years have shown that dysfunction of Bmal1 underlies a variety of psychiatric, neurodegenerative-like, and endocrine metabolism-related disorders, as well as potential oncogenic roles. In this review, we systematically summarized Bmal1 expression in different brain regions, its neurological functions related or not to circadian rhythm and biological clock, and pathological phenotypes arising from Bmal1 knockout. This review also discusses oscillation and rhythmicity, especially in the suprachiasmatic nucleus, and provides perspective on future progress in Bmal1 research.
Collapse
Affiliation(s)
- Yuanjia Zheng
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China ,grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingyun Pan
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Feixue Wang
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinglan Yan
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Taiyi Wang
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yucen Xia
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Yao
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kelin Deng
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuqi Zheng
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoye Xia
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhikai Su
- grid.411866.c0000 0000 8848 7685The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong China
| | - Hongjie Chen
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jie Lin
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhenwei Ding
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kaitong Zhang
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meng Zhang
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yongjun Chen
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China ,grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China ,Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, China
| |
Collapse
|
13
|
Inyushkin AN, Mistryugov KA, Ledyaeva OV, Romanova ID, Isakova TS, Inyushkin AA. The Effects of Insulin on Spike Activity of the Suprachiasmatic Nucleus Neurones and Functional State of Afferent Inputs from the Arcuate Nucleus in Rats. J EVOL BIOCHEM PHYS+ 2023. [DOI: 10.1134/s0022093023010210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
14
|
Iyer AR, Sheeba V. A new player in circadian networks: Role of electrical synapses in regulating functions of the circadian clock. Front Physiol 2022; 13:968574. [PMID: 36406999 PMCID: PMC9669436 DOI: 10.3389/fphys.2022.968574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Several studies have indicated that coherent circadian rhythms in behaviour can be manifested only when the underlying circadian oscillators function as a well-coupled network. The current literature suggests that circadian pacemaker neuronal networks rely heavily on communication mediated by chemical synapses comprising neuropeptides and neurotransmitters to regulate several behaviours and physiological processes. It has become increasingly clear that chemical synapses closely interact with electrical synapses and function together in the neuronal networks of most organisms. However, there are only a few studies which have examined the role of electrical synapses in circadian networks and here, we review our current understanding of gap junction proteins in circadian networks of various model systems. We describe the general mechanisms by which electrical synapses function in neural networks, their interactions with chemical neuromodulators and their contributions to the regulation of circadian rhythms. We also discuss the various methods available to characterize functional electrical synapses in these networks and the potential directions that remain to be explored to understand the roles of this relatively understudied mechanism of communication in modulating circadian behaviour.
Collapse
Affiliation(s)
- Aishwarya Ramakrishnan Iyer
- Chronobiology and Behavioural Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, Karnataka, India
- Department of Neuroscience and Behavior, Barnard College of Columbia University, New York, NY, United States
| | - Vasu Sheeba
- Chronobiology and Behavioural Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, Karnataka, India
- *Correspondence: Vasu Sheeba,
| |
Collapse
|
15
|
Pfeffer M, von Gall C, Wicht H, Korf HW. The Role of the Melatoninergic System in Circadian and Seasonal Rhythms—Insights From Different Mouse Strains. Front Physiol 2022; 13:883637. [PMID: 35492605 PMCID: PMC9039042 DOI: 10.3389/fphys.2022.883637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/22/2022] [Indexed: 01/01/2023] Open
Abstract
The melatoninergic system comprises the neurohormone melatonin and its molecular targets. The major source of melatonin is the pineal organ where melatonin is rhythmically produced during darkness. In mammals, melatonin biosynthesis is controlled by the central circadian rhythm generator in the suprachiasmatic nucleus (SCN) and photoreceptors in the retina. Melatonin elicits its function principally through two specific receptors called MT1 and MT2. MT1 is highly expressed in the SCN and the hypophysial pars tuberalis (PT), an important interface for control of seasonal functions. The expression of the MT2 is more widespread. The role of the melatoninergic system in the control of seasonal functions, such as reproduction, has been known for more than 4 decades, but investigations on its impact on the circadian system under normal (entrained) conditions started 2 decades later by comparing mouse strains with a fully functional melatoninergic system with mouse strains which either produce insufficient amounts of melatonin or lack the melatonin receptors MT1 and MT2. These studies revealed that an intact melatoninergic system is not required for the generation or maintenance of rhythmic behavior under physiological entrained conditions. As shown by jet lag experiments, the melatoninergic system facilitated faster re-entrainment of locomotor activity accompanied by a more rapid adaptation of the molecular clock work in the SCN. This action depended on MT2. Further studies indicated that the endogenous melatoninergic system stabilizes the locomotor activity under entrained conditions. Notably, these effects of the endogenous melatoninergic system are subtle, suggesting that other signals such as corticosterone or temperature contribute to the synchronization of locomotor activity. Outdoor experiments lasting for a whole year indicate a seasonal plasticity of the chronotype which depends on the melatoninergic system. The comparison between mice with an intact or a compromised melatoninergic system also points toward an impact of this system on sleep, memory and metabolism.
Collapse
Affiliation(s)
- Martina Pfeffer
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- *Correspondence: Martina Pfeffer,
| | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Helmut Wicht
- Dr. Senckenbergische Anatomie II, Fachbereich Medizin der Goethe-Universität, Frankfurt am Main, Germany
| | - Horst-Werner Korf
- Institute of Anatomy I, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
16
|
Early development of sleep and brain functional connectivity in term-born and preterm infants. Pediatr Res 2022; 91:771-786. [PMID: 33859364 DOI: 10.1038/s41390-021-01497-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/11/2021] [Accepted: 03/11/2021] [Indexed: 12/22/2022]
Abstract
The proper development of sleep and sleep-wake rhythms during early neonatal life is crucial to lifelong neurological well-being. Recent data suggests that infants who have poor quality sleep demonstrate a risk for impaired neurocognitive outcomes. Sleep ontogenesis is a complex process, whereby alternations between rudimentary brain states-active vs. wake and active sleep vs. quiet sleep-mature during the last trimester of pregnancy. If the infant is born preterm, much of this process occurs in the neonatal intensive care unit, where environmental conditions might interfere with sleep. Functional brain connectivity (FC), which reflects the brain's ability to process and integrate information, may become impaired, with ensuing risks of compromised neurodevelopment. However, the specific mechanisms linking sleep ontogenesis to the emergence of FC are poorly understood and have received little investigation, mainly due to the challenges of studying causal links between developmental phenomena and assessing FC in newborn infants. Recent advancements in infant neuromonitoring and neuroimaging strategies will allow for the design of interventions to improve infant sleep quality and quantity. This review discusses how sleep and FC develop in early life, the dynamic relationship between sleep, preterm birth, and FC, and the challenges associated with understanding these processes. IMPACT: Sleep in early life is essential for proper functional brain development, which is essential for the brain to integrate and process information. This process may be impaired in infants born preterm. The connection between preterm birth, early development of brain functional connectivity, and sleep is poorly understood. This review discusses how sleep and brain functional connectivity develop in early life, how these processes might become impaired, and the challenges associated with understanding these processes. Potential solutions to these challenges are presented to provide direction for future research.
Collapse
|
17
|
Ding H, Meng L, Liu AC, Gumz ML, Bryant AJ, Mcclung CA, Tseng GC, Esser KA, Huo Z. Likelihood-based tests for detecting circadian rhythmicity and differential circadian patterns in transcriptomic applications. Brief Bioinform 2021; 22:bbab224. [PMID: 34117739 PMCID: PMC8575021 DOI: 10.1093/bib/bbab224] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/06/2021] [Accepted: 05/22/2021] [Indexed: 01/08/2023] Open
Abstract
Circadian rhythmicity in transcriptomic profiles has been shown in many physiological processes, and the disruption of circadian patterns has been found to associate with several diseases. In this paper, we developed a series of likelihood-based methods to detect (i) circadian rhythmicity (denoted as LR_rhythmicity) and (ii) differential circadian patterns comparing two experimental conditions (denoted as LR_diff). In terms of circadian rhythmicity detection, we demonstrated that our proposed LR_rhythmicity could better control the type I error rate compared to existing methods under a wide variety of simulation settings. In terms of differential circadian patterns, we developed methods in detecting differential amplitude, differential phase, differential basal level and differential fit, which also successfully controlled the type I error rate. In addition, we demonstrated that the proposed LR_diff could achieve higher statistical power in detecting differential fit, compared to existing methods. The superior performance of LR_rhythmicity and LR_diff was demonstrated in four real data applications, including a brain aging data (gene expression microarray data of human postmortem brain), a time-restricted feeding data (RNA sequencing data of human skeletal muscles) and a scRNAseq data (single cell RNA sequencing data of mouse suprachiasmatic nucleus). An R package for our methods is publicly available on GitHub https://github.com/diffCircadian/diffCircadian.
Collapse
Affiliation(s)
- Haocheng Ding
- Department of Biostatistics at the University of Florida, Gainesville, FL, 32608, USA
| | - Lingsong Meng
- Department of Biostatistics at the University of Florida, Gainesville, FL, 32608, USA
| | - Andrew C Liu
- Department of Physiology and Functional Genomics at the University of Florida College of Medicine, Gainesville, FL, 32608, USA
| | - Michelle L Gumz
- Department of Medicine at the University of Florida, Gainesville, FL, 32608, USA
| | - Andrew J Bryant
- Department of Medicine at the University of Florida, Gainesville, FL, 32608, USA
| | - Colleen A Mcclung
- Psychiatry and Clinical and Translational Science at the University of Pittsburgh, Gainesville, FL, 32608, USA
| | - George C Tseng
- Department of Biostatistics at the University of Pittsburgh, Gainesville, FL, 32608, USA
| | - Karyn A Esser
- Department of Physiology and Functional Genomics at the University of Florida College of Medicine, Gainesville, FL, 32608, USA
| | - Zhiguang Huo
- Department of Biostatistics at the University of Florida, Gainesville, FL, 32608, USA
| |
Collapse
|
18
|
Sueviriyapan N, Granados-Fuentes D, Simon T, Herzog ED, Henson MA. Modelling the functional roles of synaptic and extra-synaptic γ-aminobutyric acid receptor dynamics in circadian timekeeping. J R Soc Interface 2021; 18:20210454. [PMID: 34520693 PMCID: PMC8440032 DOI: 10.1098/rsif.2021.0454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/23/2021] [Indexed: 11/12/2022] Open
Abstract
In the suprachiasmatic nucleus (SCN), γ-aminobutyric acid (GABA) is a primary neurotransmitter. GABA can signal through two types of GABAA receptor subunits, often referred to as synaptic GABAA (gamma subunit) and extra-synaptic GABAA (delta subunit). To test the functional roles of these distinct GABAA in regulating circadian rhythms, we developed a multicellular SCN model where we could separately compare the effects of manipulating GABA neurotransmitter or receptor dynamics. Our model predicted that blocking GABA signalling modestly increased synchrony among circadian cells, consistent with published SCN pharmacology. Conversely, the model predicted that lowering GABAA receptor density reduced firing rate, circadian cell fraction, amplitude and synchrony among individual neurons. When we tested these predictions, we found that the knockdown of delta GABAA reduced the amplitude and synchrony of clock gene expression among cells in SCN explants. The model further predicted that increasing gamma GABAA densities could enhance synchrony, as opposed to increasing delta GABAA densities. Overall, our model reveals how blocking GABAA receptors can modestly increase synchrony, while increasing the relative density of gamma over delta subunits can dramatically increase synchrony. We hypothesize that increased gamma GABAA density in the winter could underlie the tighter phase relationships among SCN cells.
Collapse
Affiliation(s)
- Natthapong Sueviriyapan
- Department of Chemical Engineering and the Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| | | | - Tatiana Simon
- Department of Biology, Washington University in St Louis, Saint Louis, MO, USA
| | - Erik D. Herzog
- Department of Biology, Washington University in St Louis, Saint Louis, MO, USA
| | - Michael A. Henson
- Department of Chemical Engineering and the Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
19
|
Cellular Calcium Levels Influenced by NCA-2 Impact Circadian Period Determination in Neurospora. mBio 2021; 12:e0149321. [PMID: 34182778 PMCID: PMC8262947 DOI: 10.1128/mbio.01493-21] [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: 11/20/2022] Open
Abstract
Intracellular calcium signaling has been implicated in the control of a variety of circadian processes in animals and plants, but its role in microbial clocks has remained largely cryptic. To examine the role of intracellular Ca2+ in the Neurospora clock, we screened mutants with knockouts of calcium transporter genes and identified a gene encoding a calcium exporter, nca-2, uniquely as having significant period effects. The loss of NCA-2 results in an increase in the cytosolic calcium level, and this leads to hyper-phosphorylation of core clock components, FRQ and WC-1, and a short period, as measured by both the core oscillator and the overt clock. Genetic analyses showed that mutations in certain frq phospho-sites and in Ca2+-calmodulin-dependent kinase 2 (camk-2) are epistatic to nca-2 in controlling the pace of the oscillator. These data are consistent with a model in which elevated intracellular Ca2+ leads to the increased activity of CAMK-2, leading to enhanced FRQ phosphorylation, accelerated closure of the circadian feedback loop, and a shortened circadian period length. At a mechanistic level, some CAMKs undergo more auto-phosphorylations in the Δnca-2 mutant, consistent with high calcium levels in the Δnca-2 mutant influencing the enzymatic activities of CAMKs. NCA-2 interacts with multiple proteins, including CSP-6, a protein known to be required for circadian output. Most importantly, the expression of nca-2 is circadian clock-controlled at both the transcriptional and translational levels, and this in combination with the period effects seen in strains lacking NCA-2 firmly places calcium signaling within the larger circadian system, where it acts as both an input to and an output from the core clock.
Collapse
|
20
|
Brooks C, Shaafi Kabiri N, Bhangu J, Cai X, Pickering E, Erb MK, Auerbach S, Bonato P, Moore TL, Mortazavi F, Thomas K. The impact of chronotype on circadian rest-activity rhythm and sleep characteristics across the week. Chronobiol Int 2021; 38:1575-1590. [PMID: 34134581 DOI: 10.1080/07420528.2021.1937197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Circadian rhythms are maintained by a complex "system of systems" that continuously coordinates biological processes with each other and the environment. Although humans predominantly entrain to solar time, individual persons vary in their precise behavioral timing due to endogenous and exogenous factors. Endogenous differences in the timing of individual circadian rhythms relative to a common environmental cue are known as chronotypes, ranging from earlier than average (Morningness) to later than average (Eveningness). Furthermore, individual behavior is often constrained by social constructs such as the 7-day week, and the "sociogenic" impact our social calendar has on our behavioral rhythms is likely modified by chronotype. Our aim in this study was to identify and characterize differences in sleep and rest-activity rhythms (RAR) between weekends and weekdays and between-chronotypes. Male volunteers (n = 24, mean age = 23.46 y) were actigraphically monitored for 4 weeks to derive objective behavioral measures of sleep and RARs. Chronotype was assessed through self-report on the Morningness-Eveningness Questionnaire. Sleep characteristics were derived using Actiware; daily rest-activity rhythms were modeled using a basic 3-parameter cosinor function. We observed that both Eveningness and Morningness Chronotypes were more active and slept later on the weekends than on weekdays. Significant between-chronotype differences in sleep timing and duration were observed within individual days of the week, especially during transitions between weekends and the workweek. Moreover, chronotypes significantly varied in their weekly rhythms: e.g. Morningness Chronotypes generally shifted their sleep duration, timing and quality across work/rest transitions quicker than Eveningness Chronotypes. Although our results should be interpreted with caution due to the limitations of our cosinor model and a homogenous cohort, they reinforce a growing body of evidence that day of the week, chronotype and their interactions must be accounted for in observational studies of human behavior, especially when circadian rhythms are of interest.
Collapse
Affiliation(s)
- Chris Brooks
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Nina Shaafi Kabiri
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jaspreet Bhangu
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Xuemei Cai
- Early Clinical Development, Pfizer Inc., Cambridge, Massachusetts, USA
| | - Eve Pickering
- Early Clinical Development, Pfizer Inc., Cambridge, Massachusetts, USA
| | | | - Sanford Auerbach
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Paolo Bonato
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA
| | - Tara L Moore
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts, USA.,Center for Systems Neuroscience, Boston University, Boston, Massachusetts, USA
| | - Farzad Mortazavi
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kevin Thomas
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Maywood ES, Chesham JE, Winsky-Sommerer R, Smyllie NJ, Hastings MH. Circadian Chimeric Mice Reveal an Interplay Between the Suprachiasmatic Nucleus and Local Brain Clocks in the Control of Sleep and Memory. Front Neurosci 2021; 15:639281. [PMID: 33679317 PMCID: PMC7935531 DOI: 10.3389/fnins.2021.639281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/29/2021] [Indexed: 12/11/2022] Open
Abstract
Sleep is regulated by circadian and homeostatic processes. Whereas the suprachiasmatic nucleus (SCN) is viewed as the principal mediator of circadian control, the contributions of sub-ordinate local circadian clocks distributed across the brain are unknown. To test whether the SCN and local brain clocks interact to regulate sleep, we used intersectional genetics to create temporally chimeric CK1ε Tau mice, in which dopamine 1a receptor (Drd1a)-expressing cells, a powerful pacemaking sub-population of the SCN, had a cell-autonomous circadian period of 24 h whereas the rest of the SCN and the brain had intrinsic periods of 20 h. We compared these mice with non-chimeric 24 h wild-types (WT) and 20 h CK1ε Tau mutants. The periods of the SCN ex vivo and the in vivo circadian behavior of chimeric mice were 24 h, as with WT, whereas other tissues in the chimeras had ex vivo periods of 20 h, as did all tissues from Tau mice. Nevertheless, the chimeric SCN imposed its 24 h period on the circadian patterning of sleep. When compared to 24 h WT and 20 h Tau mice, however, the sleep/wake cycle of chimeric mice under free-running conditions was disrupted, with more fragmented sleep and an increased number of short NREMS and REMS episodes. Even though the chimeras could entrain to 20 h light:dark cycles, the onset of activity and wakefulness was delayed, suggesting that SCN Drd1a-Cre cells regulate the sleep/wake transition. Chimeric mice also displayed a blunted homeostatic response to 6 h sleep deprivation (SD) with an impaired ability to recover lost sleep. Furthermore, sleep-dependent memory was compromised in chimeras, which performed significantly worse than 24 h WT and 20 h Tau mice. These results demonstrate a central role for the circadian clocks of SCN Drd1a cells in circadian sleep regulation, but they also indicate a role for extra-SCN clocks. In circumstances where the SCN and sub-ordinate local clocks are temporally mis-aligned, the SCN can maintain overall circadian control, but sleep consolidation and recovery from SD are compromised. The importance of temporal alignment between SCN and extra-SCN clocks for maintaining vigilance state, restorative sleep and memory may have relevance to circadian misalignment in humans, with environmental (e.g., shift work) causes.
Collapse
Affiliation(s)
| | | | - Raphaelle Winsky-Sommerer
- Surrey Sleep Research Centre, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Nicola Jane Smyllie
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | |
Collapse
|
22
|
Sabado V, Nagoshi E. Fluorescence Live Imaging of Drosophila Circadian Pacemaker Neurons. Methods Mol Biol 2021; 2130:207-219. [PMID: 33284447 DOI: 10.1007/978-1-0716-0381-9_16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Live imaging of the molecular clockwork within the circadian pacemaker neurons offers the unique possibility to study complex interactions between the molecular clock and neuronal communication within individual neurons and throughout the entire circadian circuitry. Here we describe how to establish brain explants and dissociated neuron culture from Drosophila larvae, guidelines for time-lapse fluorescence microscopy, and the method of image analysis. This approach enables the long-term monitoring of fluorescence signals of circadian reporters at single-cell resolution and can be also applicable to analyze real-time expression of other fluorescent probes in Drosophila neurons.
Collapse
Affiliation(s)
- Virginie Sabado
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
| | - Emi Nagoshi
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
23
|
Szechtman H, Harvey BH, Woody EZ, Hoffman KL. The Psychopharmacology of Obsessive-Compulsive Disorder: A Preclinical Roadmap. Pharmacol Rev 2020; 72:80-151. [PMID: 31826934 DOI: 10.1124/pr.119.017772] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This review evaluates current knowledge about obsessive-compulsive disorder (OCD), with the goal of providing a roadmap for future directions in research on the psychopharmacology of the disorder. It first addresses issues in the description and diagnosis of OCD, including the structure, measurement, and appropriate description of the disorder and issues of differential diagnosis. Current pharmacotherapies for OCD are then reviewed, including monotherapy with serotonin reuptake inhibitors and augmentation with antipsychotic medication and with psychologic treatment. Neuromodulatory therapies for OCD are also described, including psychosurgery, deep brain stimulation, and noninvasive brain stimulation. Psychotherapies for OCD are then reviewed, focusing on behavior therapy, including exposure and response prevention and cognitive therapy, and the efficacy of these interventions is discussed, touching on issues such as the timing of sessions, the adjunctive role of pharmacotherapy, and the underlying mechanisms. Next, current research on the neurobiology of OCD is examined, including work probing the role of various neurotransmitters and other endogenous processes and etiology as clues to the neurobiological fault that may underlie OCD. A new perspective on preclinical research is advanced, using the Research Domain Criteria to propose an adaptationist viewpoint that regards OCD as the dysfunction of a normal motivational system. A systems-design approach introduces the security motivation system (SMS) theory of OCD as a framework for research. Finally, a new perspective on psychopharmacological research for OCD is advanced, exploring three approaches: boosting infrastructure facilities of the brain, facilitating psychotherapeutic relearning, and targeting specific pathways of the SMS network to fix deficient SMS shut-down processes. SIGNIFICANCE STATEMENT: A significant proportion of patients with obsessive-compulsive disorder (OCD) do not achieve remission with current treatments, indicating the need for innovations in psychopharmacology for the disorder. OCD may be conceptualized as the dysfunction of a normal, special motivation system that evolved to manage the prospect of potential danger. This perspective, together with a wide-ranging review of the literature, suggests novel directions for psychopharmacological research, including boosting support systems of the brain, facilitating relearning that occurs in psychotherapy, and targeting specific pathways in the brain that provide deficient stopping processes in OCD.
Collapse
Affiliation(s)
- Henry Szechtman
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| | - Brian H Harvey
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| | - Erik Z Woody
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| | - Kurt Leroy Hoffman
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| |
Collapse
|
24
|
Álvarez-Rendón JP, Riesgo-Escovar JR. Circadian and rhythmic-related behavioral co-morbidities of the diabetic state in Drosophila melanogaster. Gen Comp Endocrinol 2020; 295:113477. [PMID: 32240709 DOI: 10.1016/j.ygcen.2020.113477] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 03/25/2020] [Accepted: 03/28/2020] [Indexed: 12/12/2022]
Abstract
Circadian phenomena rule many activities of life on earth. Disruptions in circadian rhythmicity and rhythms have been recognized as a contributing factor for diseased states, for instance metabolic disruptions like diabetes. Diabetes develops as a consequence of faulty insulin pathway signaling, either by lack of insulin production (diabetes type I), or by loss of responsiveness in target tissues (diabetes type 2). In this work we use the model organism Drosophila melanogaster with three different mutant hypomorphic conditions at different levels of the insulin pathway. The insulin pathway is a very evolutionarily conserved pathway. We study these different diabetic conditions as a source of circadian rhythm abnormalities and circadian-related co-morbidities. We do so by studying circadian rhythmicity, activity, sleep and sleep structure, and feeding behavior. Results show that flies with impaired insulin signaling show circadian rhythm and rhythmic-related co-morbidities, especially female flies, as a consequence of the diabetic state. The most extreme disruptions occur in flies with impaired insulin receptor signaling, which stands at the beginning of the insulin pathway, in principle affecting most if not all aspects of this pathway. Our work shows that defective insulin signaling is a source of circadian rhythm and rhythmic related co-morbidities.
Collapse
Affiliation(s)
- Jessica Paloma Álvarez-Rendón
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM Juriquilla, 3001 Boulevard Juriquilla, Juriquilla, Querétaro, cp 76230, Mexico
| | - Juan Rafael Riesgo-Escovar
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM Juriquilla, 3001 Boulevard Juriquilla, Juriquilla, Querétaro, cp 76230, Mexico.
| |
Collapse
|
25
|
Reiter RJ, Rosales-Corral S, Sharma R. Circadian disruption, melatonin rhythm perturbations and their contributions to chaotic physiology. Adv Med Sci 2020; 65:394-402. [PMID: 32763813 DOI: 10.1016/j.advms.2020.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 06/15/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023]
Abstract
The aim of this report is to summarize the data documenting the vital nature of well-regulated cellular and organismal circadian rhythms, which are also reflected in a stable melatonin cycle, in supporting optimal health. Cellular fluctuations in physiology exist in most cells of multicellular organisms with their stability relying on the prevailing light:dark cycle, since it regulates, via specialized intrinsically-photoreceptive retinal ganglion cells (ipRGC) and the retinohypothalamic tract, the master circadian oscillator, i.e., the suprachiasmatic nuclei (SCN). The output message of the SCN, as determined by the light:dark cycle, is transferred to peripheral oscillators, so-called slave cellular oscillators, directly via the autonomic nervous system with its limited distribution. and indirectly via the pineal-derived circulating melatonin rhythm, which contacts every cell. Via its regulatory effects on the neuroendocrine system, particularly the hypothalamo-pituitary-adrenal axis, the SCN also has a major influence on the adrenal glucocorticoid rhythm which impacts neurological diseases and psychological behaviors. Moreover, the SCN regulates the circadian production and secretion of melatonin. When the central circadian oscillator is disturbed, such as by light at night, it passes misinformation to all organs in the body. When this occurs the physiology of cells becomes altered and normal cellular functions are compromised. This physiological upheaval is a precursor to pathologies. The deterioration of the SCN/pineal network is often a normal consequence of aging and its related diseases, but in today's societies where manufactured light is becoming progressively more common worldwide, the associated pathologies may also be occurring at an earlier age.
Collapse
Affiliation(s)
- Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX, USA.
| | - Sergio Rosales-Corral
- Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Mexico
| | - Ramaswamy Sharma
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX, USA
| |
Collapse
|
26
|
Zhang Z, Zhai Q, Gu Y, Zhang T, Huang Z, Liu Z, Liu Y, Xu Y. Impaired function of the suprachiasmatic nucleus rescues the loss of body temperature homeostasis caused by time-restricted feeding. Sci Bull (Beijing) 2020; 65:1268-1280. [PMID: 32864176 PMCID: PMC7455017 DOI: 10.1016/j.scib.2020.03.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The suprachiasmatic nucleus (SCN) is the master circadian pacemaker that drives body temperature rhythm. Time-restricted feeding (TRF) has potential as a preventative or therapeutic approach against many diseases. The potential side effects of TRF remain unknown. Here we show that a 4-hour TRF stimulus in mice can severely impair body temperature homeostasis and can result in lethality. Nearly half of the mice died at 21 °C, and all mice died at 18 °C during 4-hour TRF. Moreover, this effect was modulated by the circadian clock and was associated with severe hypothermia due to loss of body temperature homeostasis, which is different from "torpor", an adaptive response under food deprivation. Disrupting the circadian clock by the SCN lesions or a non-invasive method (constant light) which disrupts circadian clock rescued lethality during TRF. Analysis of circadian gene expression in the dorsomedial hypothalamus (DMH) demonstrated that TRF reprograms rhythmic transcriptome in DMH and suppresses expression of genes, such as Ccr5 and Calcrl, which are involved in thermoregulation. We demonstrate a side effect of 4-hour TRF on the homeostasis of body temperature and a rescue function by impairing the SCN function. Altogether, our results suggested that constructing a circadian arrhythmicity may have a beneficial effect on the host response to an acute stress.
Collapse
Affiliation(s)
- Zhihui Zhang
- Model Animal Research Center, Nanjing University, 12 Xuefu Road, Pukou District, Nanjing 210061, China
| | - Qiaocheng Zhai
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yue Gu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou, Jiangsu 215123, China
| | - Tao Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhengyun Huang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhiwei Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yi Liu
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390,Correspondence to: (Y.X.), (Y.L.)
| | - Ying Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou, Jiangsu 215123, China,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China,Correspondence to: (Y.X.), (Y.L.)
| |
Collapse
|
27
|
Astrocytes Function as an Intermediate for Retrograde Endocannabinoid Signaling in the Suprachiasmatic Nucleus to Influence Circadian Clock Timing. eNeuro 2020; 7:7/4/ENEURO.0323-20.2020. [PMID: 32792412 PMCID: PMC7433895 DOI: 10.1523/eneuro.0323-20.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Highlighted Research Paper:Cannabinoid Signaling Recruits Astrocytes to Modulate Presynaptic Function in the Suprachiasmatic Nucleus. Lauren M. Hablitz, Ali N. Gunesch, Olga Cravetchi, Michael Moldavan and Charles N. Allen.
Collapse
|
28
|
Patton AP, Edwards MD, Smyllie NJ, Hamnett R, Chesham JE, Brancaccio M, Maywood ES, Hastings MH. The VIP-VPAC2 neuropeptidergic axis is a cellular pacemaking hub of the suprachiasmatic nucleus circadian circuit. Nat Commun 2020; 11:3394. [PMID: 32636383 PMCID: PMC7341843 DOI: 10.1038/s41467-020-17110-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 06/05/2020] [Indexed: 12/01/2022] Open
Abstract
The hypothalamic suprachiasmatic nuclei (SCN) are the principal mammalian circadian timekeeper, co-ordinating organism-wide daily and seasonal rhythms. To achieve this, cell-autonomous circadian timing by the ~20,000 SCN cells is welded into a tight circuit-wide ensemble oscillation. This creates essential, network-level emergent properties of precise, high-amplitude oscillation with tightly defined ensemble period and phase. Although synchronised, regional cell groups exhibit differentially phased activity, creating stereotypical spatiotemporal circadian waves of cellular activation across the circuit. The cellular circuit pacemaking components that generate these critical emergent properties are unknown. Using intersectional genetics and real-time imaging, we show that SCN cells expressing vasoactive intestinal polypeptide (VIP) or its cognate receptor, VPAC2, are neurochemically and electrophysiologically distinct, but together they control de novo rhythmicity, setting ensemble period and phase with circuit-level spatiotemporal complexity. The VIP/VPAC2 cellular axis is therefore a neurochemically and topologically specific pacemaker hub that determines the emergent properties of the SCN timekeeper. Circadian activity modulation in the suprachiasmatic nucleus (SCN) is a network-level emergent property that requires neuropeptide VIP signaling, yet the precise cellular mechanisms are unknown. Patton et al. show that cells expressing VIP or its receptor VPAC2 together determine these emergent properties of the SCN.
Collapse
Affiliation(s)
- Andrew P Patton
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Mathew D Edwards
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK.,UCL Sainsbury Wellcome Centre for Neural Circuits and Behaviour, London, UK
| | - Nicola J Smyllie
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Ryan Hamnett
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK.,Department of Neurosurgery, Stanford University, Stanford, USA
| | - Johanna E Chesham
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Marco Brancaccio
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK.,Department of Brain Sciences, UK Dementia Research Institute, Imperial College London, London, UK
| | - Elizabeth S Maywood
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Michael H Hastings
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK.
| |
Collapse
|
29
|
Méndez-Hernández R, Escobar C, Buijs RM. Suprachiasmatic Nucleus-Arcuate Nucleus Axis: Interaction Between Time and Metabolism Essential for Health. Obesity (Silver Spring) 2020; 28 Suppl 1:S10-S17. [PMID: 32538539 DOI: 10.1002/oby.22774] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023]
Abstract
In mammals, time and metabolism are tightly coupled variables; this relationship can be illustrated by numerous examples, such as the circadian variation in food intake or the circadian response to a glucose bolus. We review evidence that the interaction between the suprachiasmatic nucleus and the arcuate nucleus plays a key role in the execution of these functions. The nuclei are reciprocally connected via different projections, and this interaction provides an ideal anatomical framework to modify the temporal output of the hypothalamus to metabolic organs as a consequence of the feedback from the periphery. The suprachiasmatic nucleus-arcuate nucleus relationship is essential to integrate metabolic information into the circadian system and thus adapt circadian rhythms in core body temperature, locomotor activity, food intake, and circulating molecules such as glucose and corticosterone. With the rise in obesity-associated diseases in the world population, gaining knowledge about this relationship, and the consequences of disturbing this liaison, is essential to understand the pathogenesis of obesity.
Collapse
Affiliation(s)
- Rebeca Méndez-Hernández
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Carolina Escobar
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Ruud M Buijs
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| |
Collapse
|
30
|
Sueviriyapan N, Tso CF, Herzog ED, Henson MA. Astrocytic Modulation of Neuronal Activity in the Suprachiasmatic Nucleus: Insights from Mathematical Modeling. J Biol Rhythms 2020; 35:287-301. [PMID: 32285754 PMCID: PMC7401727 DOI: 10.1177/0748730420913672] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus consists of a highly heterogeneous neuronal population networked together to allow precise and robust circadian timekeeping in mammals. While the critical importance of SCN neurons in regulating circadian rhythms has been extensively studied, the roles of SCN astrocytes in circadian system function are not well understood. Recent experiments have demonstrated that SCN astrocytes are circadian oscillators with the same functional clock genes as SCN neurons. Astrocytes generate rhythmic outputs that are thought to modulate neuronal activity through pre- and postsynaptic interactions. In this study, we developed an in silico multicellular model of the SCN clock to investigate the impact of astrocytes in modulating neuronal activity and affecting key clock properties such as circadian rhythmicity, period, and synchronization. The model predicted that astrocytes could alter the rhythmic activity of neurons via bidirectional interactions at tripartite synapses. Specifically, astrocyte-regulated extracellular glutamate was predicted to increase neuropeptide signaling from neurons. Consistent with experimental results, we found that astrocytes could increase the circadian period and enhance neural synchronization according to their endogenous circadian period. The impact of astrocytic modulation of circadian rhythm amplitude, period, and synchronization was predicted to be strongest when astrocytes had periods between 0 and 2 h longer than neurons. Increasing the number of neurons coupled to the astrocyte also increased its impact on period modulation and synchrony. These computational results suggest that signals that modulate astrocytic rhythms or signaling (e.g., as a function of season, age, or treatment) could cause disruptions in circadian rhythm or serve as putative therapeutic targets.
Collapse
Affiliation(s)
- Natthapong Sueviriyapan
- Department of Chemical Engineering and the Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Chak Foon Tso
- Department of Biology, Washington University in St. Louis, Saint Louis, MO 63130, USA
- Current Affiliation: Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Erik D. Herzog
- Department of Biology, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Michael A. Henson
- Department of Chemical Engineering and the Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
31
|
Ding L, Xiao XH. Gut microbiota: closely tied to the regulation of circadian clock in the development of type 2 diabetes mellitus. Chin Med J (Engl) 2020; 133:817-825. [PMID: 32106122 PMCID: PMC7147650 DOI: 10.1097/cm9.0000000000000702] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Indexed: 12/20/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM), a worldwide epidemic disease, has caused tremendous economic and social burden, but the pathogenesis remains uncertain. Nowadays, the impact of unrhythmic circadian clock caused by irregular sleep and unhealthy diet on T2DM has be increasingly studied. However, the contribution of the endogenous circadian clock system to the development of T2DM has not yet been satisfactorily explored. It is now becoming clear that the gut microbiota and the circadian clock interact with each other to regulate the host metabolism. Considering all these above, we reviewed the literature related to the gut microbiota, circadian clock, and T2DM to elucidate the idea that the gut microbiota is closely tied to the regulation of the circadian clock in the development of T2DM, which provides potential for gut microbiota-directed therapies to ameliorate the effects of circadian disruptions linked to the occurrence and development of T2DM.
Collapse
Affiliation(s)
- Lu Ding
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Diabetes Research Center of Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | | |
Collapse
|
32
|
Seo DY, Yoon CS, Dizon LA, Lee SR, Youm JB, Yang WS, Kwak HB, Ko TH, Kim HK, Han J, McGregor RA. Circadian modulation of the cardiac proteome underpins differential adaptation to morning and evening exercise training: an LC-MS/MS analysis. Pflugers Arch 2020; 472:259-269. [PMID: 32025886 DOI: 10.1007/s00424-020-02350-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/09/2020] [Accepted: 01/16/2020] [Indexed: 12/11/2022]
Abstract
All living beings on earth are influenced by the circadian rhythm, the rising and the setting of the sun. The ubiquitous effect of exercise is widely believed to maximize health benefits but has not been formally investigated for cardiac responses in the exercise-induced circadian rhythms. We hypothesized that the exercise-related proteome is differentially influenced by circadian rhythm and analyzed the differences between the effects of morning and evening exercise. Twenty-four Sprague-Dawley rats were randomly divided into four groups (n = 6 per group): morning control, morning exercise, evening control, and evening exercise groups. The exercise groups were subjected to 12-week treadmill exercise (5 days/week) performed either during daytime or nighttime. After 12 weeks, the physiological characteristics (e.g., body weight, heart weight, visceral fat, and blood metabolites), cardiovascular capacity (ejection fraction (%) and fractional shortening (%)), circadian gene expression levels (clock, ball1, per1, per2, cry1, and cry2), and the proteomic data were obtained and subjected to univariate and multivariate analysis. The mRNA levels of per1 and cry2 increased in the evening group compared with those in the morning group. We also found that per2 decreased and cry2 increased in the evening exercise groups. The evening exercise groups showed more decreased triacylglycerides and increased blood insulin levels than the morning exercise group. The principal component analysis, partial least squares discriminant analysis, and orthogonal partial least squares discriminant analysis indicated that the circadian rhythm differently influenced the protein networks of the exercise groups. In the morning exercise group, the transcription-translation feedback loop (TTFL) (clock, per1, per2, cry1, and cry2) formed a protein-protein interaction network with Nme2, Hint1, Ddt, Ndufb8, Ldha, and Eef1a2. In contrast, the TTFL group appeared close to Maoa, Hist2h4, and Macrod1 in the evening exercise group. Interestingly, the evening exercise group decreased the mRNA level of per2 but not per1. Per1 and Per2 are known to transport Cry1 and Cry2 into the nucleus. Taken together, we summarized the characteristics of enriched proteins in the aspect of their molecular function, cellular component, and biological process. Our results might provide a better understanding of the circadian effect on exercise-related proteins.
Collapse
Affiliation(s)
- Dae Yun Seo
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Bokji-ro 75, Busanjin-gu, Busan, 47392, South Korea
| | - Chang Shin Yoon
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Bokji-ro 75, Busanjin-gu, Busan, 47392, South Korea
| | - Louise Anne Dizon
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Bokji-ro 75, Busanjin-gu, Busan, 47392, South Korea
| | - Sung Ryul Lee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Bokji-ro 75, Busanjin-gu, Busan, 47392, South Korea
| | - Jae Boum Youm
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Bokji-ro 75, Busanjin-gu, Busan, 47392, South Korea
| | - Won Suk Yang
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University, Seoul, 151-742, South Korea
| | - Hyo-Bum Kwak
- Department of Kinesiology, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, South Korea
| | - Tae Hee Ko
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Bokji-ro 75, Busanjin-gu, Busan, 47392, South Korea
| | - Hyoung Kyu Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Bokji-ro 75, Busanjin-gu, Busan, 47392, South Korea
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Bokji-ro 75, Busanjin-gu, Busan, 47392, South Korea.
| | - Robin A McGregor
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Project Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Bokji-ro 75, Busanjin-gu, Busan, 47392, South Korea
| |
Collapse
|
33
|
Pilorz V, Astiz M, Heinen KO, Rawashdeh O, Oster H. The Concept of Coupling in the Mammalian Circadian Clock Network. J Mol Biol 2020; 432:3618-3638. [PMID: 31926953 DOI: 10.1016/j.jmb.2019.12.037] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/22/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022]
Abstract
The circadian clock network regulates daily rhythms in mammalian physiology and behavior to optimally adapt the organism to the 24-h day/night cycle. A central pacemaker, the hypothalamic suprachiasmatic nucleus (SCN), coordinates subordinate cellular oscillators in the brain, as well as in peripheral organs to align with each other and external time. Stability and coordination of this vast network of cellular oscillators is achieved through different levels of coupling. Although coupling at the molecular level and across the SCN is well established and believed to define its function as pacemaker structure, the notion of coupling in other tissues and across the whole system is less well understood. In this review, we describe the different levels of coupling in the mammalian circadian clock system - from molecules to the whole organism. We highlight recent advances in gaining knowledge of the complex organization and function of circadian network regulation and its significance for the generation of stable but plastic intrinsic 24-h rhythms.
Collapse
Affiliation(s)
- Violetta Pilorz
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behavior and Metabolism, Marie-Curie-Strasse, 23562, Luebeck, Germany
| | - Mariana Astiz
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behavior and Metabolism, Marie-Curie-Strasse, 23562, Luebeck, Germany
| | - Keno Ole Heinen
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behavior and Metabolism, Marie-Curie-Strasse, 23562, Luebeck, Germany
| | - Oliver Rawashdeh
- The University of Queensland, School of Biomedical Sciences, Faculty of Medicine, St Lucia Qld, 4071, Australia
| | - Henrik Oster
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behavior and Metabolism, Marie-Curie-Strasse, 23562, Luebeck, Germany.
| |
Collapse
|
34
|
Furtado A, Astaburuaga R, Costa A, Duarte AC, Gonçalves I, Cipolla-Neto J, Lemos MC, Carro E, Relógio A, Santos CRA, Quintela T. The Rhythmicity of Clock Genes is Disrupted in the Choroid Plexus of the APP/PS1 Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2020; 77:795-806. [PMID: 32741824 DOI: 10.3233/jad-200331] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The choroid plexus (CP), which constitutes the blood-cerebrospinal fluid barrier, was recently identified as an important component of the circadian clock system. OBJECTIVE The fact that circadian rhythm disruption is closely associated to Alzheimer's disease (AD) led us to investigate whether AD pathology can contribute to disturbances of the circadian clock in the CP. METHODS For this purpose, we evaluated the expression of core-clock genes at different time points, in 6- and 12-month-old female and male APP/PS1 mouse models of AD. In addition, we also assessed the effect of melatonin pre-treatment in vitro before amyloid-β stimulus in the daily pattern of brain and muscle Arnt-like protein 1 (Bmal1) expression. RESULTS Our results showed a dysregulation of circadian rhythmicity of Bmal1 expression in female and male APP/PS1 transgenic 12-month-old mice and of Period 2 (Per2) expression in male mice. In addition, a significant circadian pattern of Bmal1 was measured the intermittent melatonin pre-treatment group, showing that melatonin can reset the CP circadian clock. CONCLUSION These results demonstrated a connection between AD and the disruption of circadian rhythm in the CP, representing an attractive target for disease prevention and/or treatment.
Collapse
Affiliation(s)
- André Furtado
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - Rosario Astaburuaga
- Institute for Theoretical Biology (ITB), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Germany
- Medical Department of Hematology, Oncology, and Tumor Immunology and Molekulares Krebsforschungszentrum (MKFZ), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Germany
| | - Ana Costa
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - Ana C Duarte
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - Isabel Gonçalves
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - José Cipolla-Neto
- Laboratory of Neurobiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Manuel C Lemos
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - Eva Carro
- Networked Biomedical Research Center in Neurodegenerative Diseases (CIBERNED), Spain
- Group of Neurodegenerative Diseases, Hospital 12 de Octubre Research Institute (imas12), Madrid, Spain
| | - Angela Relógio
- Institute for Theoretical Biology (ITB), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Germany
- Medical Department of Hematology, Oncology, and Tumor Immunology and Molekulares Krebsforschungszentrum (MKFZ), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin, Berlin Institute of Health, Germany
- Department of Human Medicine, Institute for Systems Medicine and Bioinformatics, MSH Medical School Hamburg - University of Applied Sciences and Medical University, Hamburg, Germany
| | - Cecília R A Santos
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - Telma Quintela
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
35
|
Helfrich-Förster C. Light input pathways to the circadian clock of insects with an emphasis on the fruit fly Drosophila melanogaster. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2019; 206:259-272. [PMID: 31691095 PMCID: PMC7069913 DOI: 10.1007/s00359-019-01379-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/19/2019] [Accepted: 10/26/2019] [Indexed: 12/26/2022]
Abstract
Light is the most important Zeitgeber for entraining animal activity rhythms to the 24-h day. In all animals, the eyes are the main visual organs that are not only responsible for motion and colour (image) vision, but also transfer light information to the circadian clock in the brain. The way in which light entrains the circadian clock appears, however, variable in different species. As do vertebrates, insects possess extraretinal photoreceptors in addition to their eyes (and ocelli) that are sometimes located close to (underneath) the eyes, but sometimes even in the central brain. These extraretinal photoreceptors contribute to entrainment of their circadian clocks to different degrees. The fruit fly Drosophila melanogaster is special, because it expresses the blue light-sensitive cryptochrome (CRY) directly in its circadian clock neurons, and CRY is usually regarded as the fly’s main circadian photoreceptor. Nevertheless, recent studies show that the retinal and extraretinal eyes transfer light information to almost every clock neuron and that the eyes are similarly important for entraining the fly’s activity rhythm as in other insects, or more generally spoken in other animals. Here, I compare the light input pathways between selected insect species with a focus on Drosophila’s special case.
Collapse
|
36
|
Gall AJ, Goodwin AM, Khacherian OS, Teal LB. Superior Colliculus Lesions Lead to Disrupted Responses to Light in Diurnal Grass Rats ( Arvicanthis niloticus). J Biol Rhythms 2019; 35:45-57. [PMID: 31619104 DOI: 10.1177/0748730419881920] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The circadian system regulates daily rhythms of physiology and behavior. Although extraordinary advances have been made to elucidate the brain mechanisms underlying the circadian system in nocturnal species, less is known in diurnal species. Recent studies have shown that retinorecipient brain areas such as the intergeniculate leaflet (IGL) and olivary pretectal nucleus (OPT) are critical for the display of normal patterns of daily activity in diurnal grass rats (Arvicanthis niloticus). Specifically, grass rats with IGL and OPT lesions respond to light in similar ways to intact nocturnal animals. Importantly, both the IGL and OPT project to one another in nocturnal species, and there is evidence that these 2 brain regions also project to the superior colliculus (SC). The SC receives direct retinal input, is involved in the triggering of rapid eye movement sleep in nocturnal rats, and is disproportionately large in the diurnal grass rat. The objective of the current study was to use diurnal grass rats to test the hypothesis that the SC is critical for the expression of diurnal behavior and physiology. We performed bilateral electrolytic lesions of the SC in female grass rats to examine behavioral patterns and acute responses to light. Most grass rats with SC lesions expressed significantly reduced activity in the presence of light. Exposing these grass rats to constant darkness reinstated activity levels during the subjective day, suggesting that light masks their ability to display a diurnal activity profile in 12:12 LD. Altogether, our data suggest that the SC is critical for maintaining normal responses to light in female grass rats.
Collapse
Affiliation(s)
- Andrew J Gall
- Department of Psychology and Neuroscience Program, Hope College, Holland, Michigan
| | - Alyssa M Goodwin
- Department of Psychology and Neuroscience Program, Hope College, Holland, Michigan
| | - Ohanes S Khacherian
- Department of Psychology and Neuroscience Program, Hope College, Holland, Michigan
| | - Laura B Teal
- Department of Psychology and Neuroscience Program, Hope College, Holland, Michigan
| |
Collapse
|
37
|
Liu J, Gao D, Dan J, Liu D, Peng L, Zhou R, Luo Y. The protective effect of cycloastragenol on aging mouse circadian rhythmic disorder induced by d-galactose. J Cell Biochem 2019; 120:16408-16415. [PMID: 31310357 DOI: 10.1002/jcb.28587] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/19/2019] [Accepted: 01/24/2019] [Indexed: 01/04/2023]
Abstract
Aging process in mammals is associated with a decline in amplitude and a long period of circadian behaviors which are regulated by a central circadian regulator in the suprachiasmatic nucleus (SCN) and local oscillators in peripheral tissues. It is unclear whether enhancing clock function can retard aging. Using fibroblasts expressing per2::lucSV and senescent cells, we revealed cycloastragenol (CAG), a natural aglycone derivative from astragaloside IV, as a clock amplitude enhancing small molecule. CAG could activate telomerase to antiaging, but no reports focused on its effects on circadian rhythm disorders in aging mice. Here we analyze the potential effects of CAG on d-galactose-induced aging mice on the circadian behavior and expression of clock genes. For this purpose, CAG (20 mg/kg orally), was administered daily to d-galactose (150 mg/kg, subcutaneous) mice model of aging for 6 weeks. An actogram analysis of free-running activity of these mice showed that CAG significantly enhances the locomotor activity. We further found that CAG increase expressions of per2 and bmal1 genes in liver and kidney of aging mouse. Furthermore, CAG enhanced clock protein BMAL1 and PER2 levels in aging mouse liver and SCN. Our results indicated that the CAG could restore the behavior of circadian rhythm in aging mice induced by d-galactose. These data of present study suggested that CAG could be used as a novel therapeutic strategy for the treatment of age-related circadian rhythm disruption.
Collapse
Affiliation(s)
- Jing Liu
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Dongxiao Gao
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Juhua Dan
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Dan Liu
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Lei Peng
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Ruoyu Zhou
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Ying Luo
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| |
Collapse
|
38
|
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus is remarkable. Despite numbering only about 10,000 neurons on each side of the third ventricle, the SCN is our principal circadian clock, directing the daily cycles of behaviour and physiology that set the tempo of our lives. When this nucleus is isolated in organotypic culture, its autonomous timing mechanism can persist indefinitely, with precision and robustness. The discovery of the cell-autonomous transcriptional and post-translational feedback loops that drive circadian activity in the SCN provided a powerful exemplar of the genetic specification of complex mammalian behaviours. However, the analysis of circadian time-keeping is moving beyond single cells. Technical and conceptual advances, including intersectional genetics, multidimensional imaging and network theory, are beginning to uncover the circuit-level mechanisms and emergent properties that make the SCN a uniquely precise and robust clock. However, much remains unknown about the SCN, not least the intrinsic properties of SCN neurons, its circuit topology and the neuronal computations that these circuits support. Moreover, the convention that the SCN is a neuronal clock has been overturned by the discovery that astrocytes are an integral part of the timepiece. As a test bed for examining the relationships between genes, cells and circuits in sculpting complex behaviours, the SCN continues to offer powerful lessons and opportunities for contemporary neuroscience.
Collapse
|
39
|
Ali AAH, Stahr A, Ingenwerth M, Theis M, Steinhäuser C, von Gall C. Connexin30 and Connexin43 show a time-of-day dependent expression in the mouse suprachiasmatic nucleus and modulate rhythmic locomotor activity in the context of chronodisruption. Cell Commun Signal 2019; 17:61. [PMID: 31186021 PMCID: PMC6560876 DOI: 10.1186/s12964-019-0370-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 05/14/2019] [Indexed: 11/15/2022] Open
Abstract
Background The astroglial connexins Cx30 and Cx43 contribute to many important CNS functions including cognitive behaviour, motoric capacity and regulation of the sleep-wake cycle. The sleep wake cycle, is controlled by the circadian system. The central circadian rhythm generator resides in the suprachiasmatic nucleus (SCN). SCN neurons are tightly coupled in order to generate a coherent circadian rhythm. The SCN receives excitatory glutamatergic input from the retina which mediates entrainment of the circadian system to the environmental light-dark cycle. Connexins play an important role in electric coupling of SCN neurons and astrocytic-neuronal signalling that regulates rhythmic SCN neuronal activity. However, little is known about the regulation of Cx30 and Cx43 expression in the SCN, and the role of these connexins in light entrainment of the circadian system and in circadian rhythm generation. Methods We analysed time-of-day dependent as well as circadian expression of Cx30 and Cx43 mRNA and protein in the mouse SCN by means of qPCR and immunohistochemistry. Moreover, we analysed rhythmic spontaneous locomotor activity in mice with a targeted deletion of Cx30 and astrocyte specific deletion of Cx43 (DKO) in different light regimes by means of on-cage infrared detectors. Results Fluctuation of Cx30 protein expression is strongly dependent on the light-dark cycle whereas fluctuation of Cx43 protein expression persisted in constant darkness. DKO mice entrained to the light-dark cycle. However, re-entrainment after a phase delay was slightly impaired in DKO mice. Surprisingly, DKO mice were more resilient to chronodisruption. Conclusion Circadian fluctuation of Cx30 and Cx43 protein expression in the SCN is differently regulated. Cx30 and astroglial Cx43 play a role in rhythm stability and re-entrainment under challenging conditions. Electronic supplementary material The online version of this article (10.1186/s12964-019-0370-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Amira A H Ali
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Merowinger Platz 1A, 40225, Düsseldorf, Germany
| | - Anna Stahr
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Merowinger Platz 1A, 40225, Düsseldorf, Germany
| | - Marc Ingenwerth
- Institute of Pathology, Medical Faculty, University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Martin Theis
- Institute of Cellular Neurosciences, Medical Faculty, University Bonn, Sigmund Freud Str. 25, 53105, Bonn, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University Bonn, Sigmund Freud Str. 25, 53105, Bonn, Germany
| | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Merowinger Platz 1A, 40225, Düsseldorf, Germany.
| |
Collapse
|
40
|
Michel S, Meijer JH. From clock to functional pacemaker. Eur J Neurosci 2019; 51:482-493. [PMID: 30793396 PMCID: PMC7027845 DOI: 10.1111/ejn.14388] [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: 11/09/2018] [Revised: 01/23/2019] [Accepted: 02/11/2019] [Indexed: 12/17/2022]
Abstract
In mammals, the central pacemaker that coordinates 24‐hr rhythms is located in the suprachiasmatic nucleus (SCN). Individual neurons of the SCN have a molecular basis for rhythm generation and hence, they function as cell autonomous oscillators. Communication and synchronization among these neurons are crucial for obtaining a coherent rhythm at the population level, that can serve as a pace making signal for brain and body. Hence, the ability of single SCN neurons to produce circadian rhythms is equally important as the ability of these neurons to synchronize one another, to obtain a bona fide pacemaker at the SCN tissue level. In this chapter we will discuss the mechanisms underlying synchronization, and plasticity herein, which allows adaptation to changes in day length. Furthermore, we will discuss deterioration in synchronization among SCN neurons in aging, and gain in synchronization by voluntary physical activity or exercise.
Collapse
Affiliation(s)
- Stephan Michel
- Group Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Johanna H Meijer
- Group Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
41
|
Lundberg L, Sienkiewicz Z, Anthony DC, Broom KA. Effects of 50 Hz magnetic fields on circadian rhythm control in mice. Bioelectromagnetics 2019; 40:250-259. [PMID: 30945762 PMCID: PMC6617993 DOI: 10.1002/bem.22188] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 03/12/2019] [Indexed: 12/27/2022]
Abstract
Artificial light and power frequency magnetic fields are ubiquitous in the built environment. Light is a potent zeitgeber but it is unclear whether power frequency magnetic fields can influence circadian rhythm control. To study this possibility, 8-12-week-old male C57BL/6J mice were exposed for 30 min starting at zeitgeber time 14 (ZT14, 2 h into the dark period of the day) to 50 Hz magnetic fields at 580 μT using a pair of Helmholtz coils and/or a blue LED light at 700 lux or neither. Our experiments revealed an acute adrenal response to blue light, in terms of increased adrenal per1 gene expression, increased serum corticosterone levels, increased time spent sleeping, and decreased locomotor activity (in all cases, P < 0.0001) compared to an unexposed control group. There appeared to be no modulating effect of the magnetic fields on the response to light, and there was also no effect of the magnetic fields alone (in both cases, P > 0.05) except for a decrease in locomotor activity (P < 0.03). Gene expression of the cryptochromes cry1 and cry2 in the adrenals, liver, and hippocampus was also not affected by exposures (in all cases, P > 0.05). In conclusion, these results suggest that 50 Hz magnetic fields do not significantly affect the acute light response to a degree that can be detected in the adrenal response. Bioelectromagnetics. 2019;9999:XX-XX. © 2019 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Louise Lundberg
- Public Health EnglandChiltonUnited Kingdom
- Department of PharmacologyUniversity of OxfordOxfordUnited Kingdom
| | | | | | | |
Collapse
|
42
|
Sládek M, Sumová A. Modulation of NMDA-Mediated Clock Resetting in the Suprachiasmatic Nuclei of mPer2 Luc Mouse by Endocannabinoids. Front Physiol 2019; 10:361. [PMID: 30984034 PMCID: PMC6450388 DOI: 10.3389/fphys.2019.00361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/14/2019] [Indexed: 11/29/2022] Open
Abstract
Light entrains the master circadian clock in the suprachiasmatic nucleus (SCN) predominantly through glutamatergic signaling via NMDA receptors. The magnitude and the direction of resulting phase shifts depend on timing of the photic stimulus. Previous reports based on behavioral and electrophysiological data suggested that endocannabinoids (EC) might reduce the ability of the SCN clock to respond to light. However, there is little direct evidence for the involvement of EC in entrainment of the rhythmic clock gene expression in the SCN. We have used luminescence recording of cultured SCN slices from mPer2Luc mice to construct a complete phase response curve (PRC) for NMDA receptor activation. The results demonstrated that NMDA administration phase-shifts the PER2 rhythm in a time-specific manner. A stable “singularity,” in the course of which the clock seemingly stops while the overall phase is caught between delays and advances, can occur in response to NMDA at a narrow interval during the PER2 level decrease. NMDA-induced phase delays were affected neither by the agonist (WIN 55,212-2 mesylate) nor by the antagonist (rimonabant hydrochloride) of EC receptors. However, the agonist significantly reduced the NMDA-induced phase advance of the clock, while the antagonist enhanced the phase advance, causing a shift in the sensitivity window of the SCN to NMDA. The modulation of EC signaling in the SCN had no effect by itself on the phase of the PER2 rhythm. The results provide evidence for a modulatory role of EC in photic entrainment of the circadian clock in the SCN.
Collapse
Affiliation(s)
- Martin Sládek
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Alena Sumová
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czechia
| |
Collapse
|
43
|
Hore PJ. Upper bound on the biological effects of 50/60 Hz magnetic fields mediated by radical pairs. eLife 2019; 8:44179. [PMID: 30801245 PMCID: PMC6417859 DOI: 10.7554/elife.44179] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/02/2019] [Indexed: 01/02/2023] Open
Abstract
Prolonged exposure to weak (~1 μT) extremely-low-frequency (ELF, 50/60 Hz) magnetic fields has been associated with an increased risk of childhood leukaemia. One of the few biophysical mechanisms that might account for this link involves short-lived chemical reaction intermediates known as radical pairs. In this report, we use spin dynamics simulations to derive an upper bound of 10 parts per million on the effect of a 1 μT ELF magnetic field on the yield of a radical pair reaction. By comparing this figure with the corresponding effects of changes in the strength of the Earth’s magnetic field, we conclude that if exposure to such weak 50/60 Hz magnetic fields has any effect on human biology, and results from a radical pair mechanism, then the risk should be no greater than travelling a few kilometres towards or away from the geomagnetic north or south pole.
Collapse
Affiliation(s)
- P J Hore
- Department of Chemistry, Physical & Theoretical Chemistry Laboratory, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
44
|
Overexpression of Prokineticin 2 in Transgenic Mice Leads to Reduced Circadian Behavioral Rhythmicity and Altered Molecular Rhythms in the Suprachiasmatic Clock. J Circadian Rhythms 2018; 16:13. [PMID: 30473715 PMCID: PMC6234414 DOI: 10.5334/jcr.170] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In mammals, the master pacemaker driving circadian rhythms is thought to reside in the suprachiasmatic nuclei (SCN) of the anterior hypothalamus. A clear view of molecular clock mechanisms within the SCN neurons has been elucidated. In contrast, much less is known about the output mechanism by which the SCN circadian pacemaker sends timing information for eventual control of physiological and behavioral rhythms. Two secreted molecules, prokineticin 2 (PK2) and vasopressin, that are encoded by respective clock-controlled genes, have been indicated as candidate SCN output molecules. Several lines of evidence have emerged that support the role of PK2 as an output signal for the SCN circadian clock, including the reduced circadian rhythms in mice that are deficient in PK2 or its receptor, PKR2. In the current study, transgenic mice with the overexpression of PK2 have been generated. These transgenic mice displayed reduced oscillation of the PK2 expression in the SCN and decreased amplitude of circadian locomotor rhythm, supporting the important signaling role of PK2 in the regulation of circadian rhythms. Altered molecular rhythms were also observed in the SCN in the transgenic mice, indicating that PK2 signaling also regulates the operation of core clockwork. This conclusion is consistent with recent reports showing the likely signaling role of PK2 from the intrinsically photosensitive retinal ganglion cells to SCN neurons. Thus, PK2 signaling plays roles in both the input and the output pathways of the SCN circadian clock.
Collapse
|
45
|
Connectome of the Suprachiasmatic Nucleus: New Evidence of the Core-Shell Relationship. eNeuro 2018; 5:eN-NWR-0205-18. [PMID: 30283813 PMCID: PMC6168316 DOI: 10.1523/eneuro.0205-18.2018] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 09/05/2018] [Accepted: 09/07/2018] [Indexed: 11/21/2022] Open
Abstract
A brain clock, constituted of ∼20,000 peptidergically heterogeneous neurons, is located in the hypothalamic suprachiasmatic nucleus (SCN). While many peptidergic cell types have been identified, little is known about the connections among these neurons in mice. We first sought to identify contacts among major peptidergic cell types in the SCN using triple-label fluorescent immunocytochemistry (ICC). To this end, contacts among vasoactive intestinal polypeptide (VIP), gastrin-releasing peptide (GRP), and calretinin (CALR) cells of the core, and arginine vasopressin (AVP) and met-enkephalin (ENK) cells of the shell were analyzed. Some core-to-shell and shell-to-core communications are specialized. We found that in wild-type (WT) mice, AVP fibers make extremely sparse contacts onto VIP neurons but contacts in the reverse direction are numerous. In contrast, AVP fibers make more contacts onto GRP neurons than conversely. For the other cell types tested, largely reciprocal connections are made. These results point to peptidergic cell type-specific communications between core and shell SCN neurons. To further understand the impact of VIP-to-AVP communication, we next explored the SCN in VIP-deficient mice (VIP-KO). In these animals, AVP expression is markedly reduced in the SCN, but it is not altered in the paraventricular nucleus (PVN) and supraoptic nucleus (SON). Surprisingly, in VIP-KO mice, the number of AVP appositions onto other peptidergic cell types is not different from controls. Colchicine administration, which blocks AVP transport, restored the numbers of AVP neurons in VIP-KO to that of WT littermates. The results indicate that VIP has an important role in modulating AVP expression levels in the SCN in this mouse.
Collapse
|
46
|
Wheaton KL, Hansen KF, Aten S, Sullivan KA, Yoon H, Hoyt KR, Obrietan K. The Phosphorylation of CREB at Serine 133 Is a Key Event for Circadian Clock Timing and Entrainment in the Suprachiasmatic Nucleus. J Biol Rhythms 2018; 33:497-514. [PMID: 30175684 DOI: 10.1177/0748730418791713] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Within the suprachiasmatic nucleus (SCN)-the locus of the master circadian clock- transcriptional regulation via the CREB/CRE pathway is implicated in the functioning of the molecular clock timing process, and is a key conduit through which photic input entrains the oscillator. One event driving CRE-mediated transcription is the phosphorylation of CREB at serine 133 (Ser133). Indeed, numerous reporter gene assays have shown that an alanine point mutation in Ser133 reduces CREB-mediated transcription. Here, we sought to examine the contribution of Ser133 phosphorylation to the functional role of CREB in SCN clock physiology in vivo. To this end, we used a CREB knock-in mouse strain, in which Ser133 was mutated to alanine (S/A CREB). Under a standard 12 h light-dark cycle, S/A CREB mice exhibited a marked alteration in clock-regulated wheel running activity. Relative to WT mice, S/A CREB mice had highly fragmented bouts of locomotor activity during the night phase, elevated daytime activity, and a delayed phase angle of entrainment. Further, under free-running conditions, S/A CREB mice had a significantly longer tau than WT mice and reduced activity amplitude. In S/A CREB mice, light-evoked clock entrainment, using both Aschoff type 1 and 6 h "jet lag" paradigms, was markedly reduced relative to WT mice. S/A CREB mice exhibited attenuated transcriptional drive, as assessed by examining both clock-gated and light-evoked gene expression. Finally, SCN slice culture imaging detected a marked disruption in cellular clock phase synchrony following a phase-resetting stimulus in S/A CREB mice. Together, these data indicate that signaling through CREB phosphorylation at Ser133 is critical for the functional fidelity of both SCN timing and entrainment.
Collapse
Affiliation(s)
- Kelin L Wheaton
- Division of Pharmaceutics and Pharmaceutical Chemistry, Ohio State University, Columbus, OH
| | | | - Sydney Aten
- Department of Neuroscience, Ohio State University, Columbus, OH
| | - Kyle A Sullivan
- Department of Neuroscience, Ohio State University, Columbus, OH
| | - Hyojung Yoon
- Department of Neuroscience, Ohio State University, Columbus, OH
| | - Kari R Hoyt
- Division of Pharmaceutics and Pharmaceutical Chemistry, Ohio State University, Columbus, OH
| | - Karl Obrietan
- Department of Neuroscience, Ohio State University, Columbus, OH
| |
Collapse
|
47
|
Mazuski C, Abel JH, Chen SP, Hermanstyne TO, Jones JR, Simon T, Doyle FJ, Herzog ED. Entrainment of Circadian Rhythms Depends on Firing Rates and Neuropeptide Release of VIP SCN Neurons. Neuron 2018; 99:555-563.e5. [PMID: 30017392 PMCID: PMC6085153 DOI: 10.1016/j.neuron.2018.06.029] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 05/13/2018] [Accepted: 06/15/2018] [Indexed: 01/23/2023]
Abstract
The mammalian suprachiasmatic nucleus (SCN) functions as a master circadian pacemaker, integrating environmental input to align physiological and behavioral rhythms to local time cues. Approximately 10% of SCN neurons express vasoactive intestinal polypeptide (VIP); however, it is unknown how firing activity of VIP neurons releases VIP to entrain circadian rhythms. To identify physiologically relevant firing patterns, we optically tagged VIP neurons and characterized spontaneous firing over 3 days. VIP neurons had circadian rhythms in firing rate and exhibited two classes of instantaneous firing activity. We next tested whether physiologically relevant firing affected circadian rhythms through VIP release. We found that VIP neuron stimulation with high, but not low, frequencies shifted gene expression rhythms in vitro through VIP signaling. In vivo, high-frequency VIP neuron activation rapidly entrained circadian locomotor rhythms. Thus, increases in VIP neuronal firing frequency release VIP and entrain molecular and behavioral circadian rhythms. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Cristina Mazuski
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - John H Abel
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Samantha P Chen
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Tracey O Hermanstyne
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Jeff R Jones
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Tatiana Simon
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Francis J Doyle
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Erik D Herzog
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA.
| |
Collapse
|
48
|
Calcium and cAMP directly modulate the speed of the Drosophila circadian clock. PLoS Genet 2018; 14:e1007433. [PMID: 29879123 PMCID: PMC6007936 DOI: 10.1371/journal.pgen.1007433] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/19/2018] [Accepted: 05/18/2018] [Indexed: 01/08/2023] Open
Abstract
Circadian clocks impose daily periodicities to animal behavior and physiology. At their core, circadian rhythms are produced by intracellular transcriptional/translational feedback loops (TTFL). TTFLs may be altered by extracellular signals whose actions are mediated intracellularly by calcium and cAMP. In mammals these messengers act directly on TTFLs via the calcium/cAMP-dependent transcription factor, CREB. In the fruit fly, Drosophila melanogaster, calcium and cAMP also regulate the periodicity of circadian locomotor activity rhythmicity, but whether this is due to direct actions on the TTFLs themselves or are a consequence of changes induced to the complex interrelationship between different classes of central pacemaker neurons is unclear. Here we investigated this question focusing on the peripheral clock housed in the non-neuronal prothoracic gland (PG), which, together with the central pacemaker in the brain, controls the timing of adult emergence. We show that genetic manipulations that increased and decreased the levels of calcium and cAMP in the PG caused, respectively, a shortening and a lengthening of the periodicity of emergence. Importantly, knockdown of CREB in the PG caused an arrhythmic pattern of eclosion. Interestingly, the same manipulations directed at central pacemaker neurons caused arrhythmicity of eclosion and of adult locomotor activity, suggesting a common mechanism. Our results reveal that the calcium and cAMP pathways can alter the functioning of the clock itself. In the PG, these messengers, acting as outputs of the clock or as second messengers for stimuli external to the PG, could also contribute to the circadian gating of adult emergence. Circadian clocks impose daily periodicities to animal behavior and physiology. At their core, circadian rhythms are produced by intracellular transcriptional/translational feedback loops (TTFL). TTFLs may be altered by extracellular signals whose actions are mediated intracellularly by calcium and cAMP. In Drosophila, calcium and cAMP levels affect the periodicity of Drosophila circadian rhythms, but whether this is due to direct actions on the TTFLs themselves or is a consequence of changes induced to the complex interrelationship between different classes of central pacemaker neurons is unclear. Here we used the non-neuronal circadian clock located in the prothoracic gland (PG) to show that these messengers affect the speed of the circadian clock that controls the timing of adult emergence and suggest that these actions are mediated by CREB. Importantly, since calcium and cAMP are also output signals of the clock, they may contribute to the mechanism that imposes a circadian gating to the timing of adult emergence.
Collapse
|
49
|
Cooper JM, Halter KA, Prosser RA. Circadian rhythm and sleep-wake systems share the dynamic extracellular synaptic milieu. Neurobiol Sleep Circadian Rhythms 2018; 5:15-36. [PMID: 31236509 PMCID: PMC6584685 DOI: 10.1016/j.nbscr.2018.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/06/2018] [Accepted: 04/10/2018] [Indexed: 01/23/2023] Open
Abstract
The mammalian circadian and sleep-wake systems are closely aligned through their coordinated regulation of daily activity patterns. Although they differ in their anatomical organization and physiological processes, they utilize overlapping regulatory mechanisms that include an assortment of proteins and molecules interacting within the extracellular space. These extracellular factors include proteases that interact with soluble proteins, membrane-attached receptors and the extracellular matrix; and cell adhesion molecules that can form complex scaffolds connecting adjacent neurons, astrocytes and their respective intracellular cytoskeletal elements. Astrocytes also participate in the dynamic regulation of both systems through modulating neuronal appositions, the extracellular space and/or through release of gliotransmitters that can further contribute to the extracellular signaling processes. Together, these extracellular elements create a system that integrates rapid neurotransmitter signaling across longer time scales and thereby adjust neuronal signaling to reflect the daily fluctuations fundamental to both systems. Here we review what is known about these extracellular processes, focusing specifically on areas of overlap between the two systems. We also highlight questions that still need to be addressed. Although we know many of the extracellular players, far more research is needed to understand the mechanisms through which they modulate the circadian and sleep-wake systems.
Collapse
Key Words
- ADAM, A disintegrin and metalloproteinase
- AMPAR, AMPA receptor
- Astrocytes
- BDNF, brain-derived neurotrophic factor
- BMAL1, Brain and muscle Arnt-like-1 protein
- Bmal1, Brain and muscle Arnt-like-1 gene
- CAM, cell adhesion molecules
- CRY, cryptochrome protein
- Cell adhesion molecules
- Circadian rhythms
- Cry, cryptochrome gene
- DD, dark-dark
- ECM, extracellular matrix
- ECS, extracellular space
- EEG, electroencephalogram
- Endo N, endoneuraminidase N
- Extracellular proteases
- GFAP, glial fibrillary acidic protein
- IL, interleukin
- Ig, immunoglobulin
- LC, locus coeruleus
- LD, light-dark
- LH, lateral hypothalamus
- LRP-1, low density lipoprotein receptor-related protein 1
- LTP, long-term potentiation
- MMP, matrix metalloproteinases
- NCAM, neural cell adhesion molecule protein
- NMDAR, NMDA receptor
- NO, nitric oxide
- NST, nucleus of the solitary tract
- Ncam, neural cell adhesion molecule gene
- Nrl, neuroligin gene
- Nrx, neurexin gene
- P2, purine type 2 receptor
- PAI-1, plasminogen activator inhibitor-1
- PER, period protein
- PPT, peduculopontine tegmental nucleus
- PSA, polysialic acid
- Per, period gene
- REMS, rapid eye movement sleep
- RSD, REM sleep disruption
- SCN, suprachiasmatic nucleus
- SWS, slow wave sleep
- Sleep-wake system
- Suprachiasmatic nucleus
- TNF, tumor necrosis factor
- TTFL, transcriptional-translational negative feedback loop
- VIP, vasoactive intestinal polypeptide
- VLPO, ventrolateral preoptic
- VP, vasopressin
- VTA, ventral tegmental area
- dNlg4, drosophila neuroligin-4 gene
- nNOS, neuronal nitric oxide synthase gene
- nNOS, neuronal nitric oxide synthase protein
- tPA, tissue-type plasminogen activator
- uPA, urokinase-type plasminogen activator
- uPAR, uPA receptor
Collapse
|
50
|
Cheng PC, Wang YC, Chen YS, Cheng RC, Yang JJ, Huang RC. Differential regulation of nimodipine-sensitive and -insensitive Ca 2+ influx by the Na +/Ca 2+ exchanger and mitochondria in the rat suprachiasmatic nucleus neurons. J Biomed Sci 2018; 25:44. [PMID: 29788971 PMCID: PMC5964920 DOI: 10.1186/s12929-018-0447-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 05/14/2018] [Indexed: 11/25/2022] Open
Abstract
Background Transmembrane Ca2+ influx is critical for molecular rhythmicity, metabolic activity, and neuropeptide release in the central clock of the suprachiasmatic nucleus (SCN). We previously reported that both the Na+/Ca2+ exchanger (NCX) and mitochondria play a role in regulating intracellular Ca2+ homeostasis in the rat SCN neurons. Here we present evidence to show differential regulation by NCX and mitochondria of nimodipine-sensitive and -insensitive Ca2+ influx. Methods Ratiometric Ca2+ imaging was used to measure change in [Ca2+]i and patch clamp recordings to study spontaneous firing, membrane potential, and voltage-dependent Ca2+ channels in neurons from reduced SCN slice preparations. Immunofluorescent staining was used to determine the distribution pattern of CaV1.2 and CaV1.3 and their colocalization with NCX1. Results Ratiometric Ca2+ imaging indicates that nimodipine (2 μM) blocked most of 20 (mM) K+-induced, but less so of 50 K+-induced, Ca2+ rise. The nimodipine-sensitive 50 K+-induced Ca2+ transient rose more rapidly but decayed similarly with the nimodipine-insensitive component, suggesting both components were extruded by NCX. Immunofluorescent stains showed the expression of both CaV1.2 and CaV1.3 and their colocalization with NCX1, whereas functional studies suggest that CaV1.2 mediated most of the nimodipine-sensitive Ca2+ rise but had insignificant effect on spontaneous firing. After normalization relative to the Ca2+-free solution, nimodipine reduced ~ 65% of basal Ca2+ influx, and TTX lowered it by ~ 35%, leaving ~ 25% basal Ca2+ influx in the combined presence of TTX and nimodipine. With the mitochondrial uncoupler carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP) to inhibit mitochondrial Ca2+ uptake, 20 K+-induced Ca2+ transients became larger and slower, both in the absence and presence of nimodipine. FCCP markedly enhanced nimodipine-insensitive, but not nimodipine-sensitive, Ca2+ transients, suggesting that mitochondria preferentially buffer nimodipine-insensitive Ca2+ influx. Results from using CaV2 channel blockers further indicate that FCCP enhanced Ca2+ transients mediated by N-, P/Q-, and the blocker cocktail-insensitive Ca2+ channels. Conclusions The differential regulation of transmembrane Ca2+ influx by NCX and mitochondria suggests that Ca2+ entry via different sources may be regulated differently to play different roles in SCN physiology.
Collapse
Affiliation(s)
- Pi-Cheng Cheng
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-San, Tao-Yuan, 33302, Taiwan
| | - Yi-Chi Wang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-San, Tao-Yuan, 33302, Taiwan
| | - Ya-Shuan Chen
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-San, Tao-Yuan, 33302, Taiwan
| | - Ruo-Ciao Cheng
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-San, Tao-Yuan, 33302, Taiwan
| | - Jyh-Jeen Yang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-San, Tao-Yuan, 33302, Taiwan
| | - Rong-Chi Huang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-San, Tao-Yuan, 33302, Taiwan. .,Healthy Aging Research Center, Chang Gung University, Tao-Yuan, 33302, Taiwan. .,Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Tao-Yuan, 33305, Taiwan.
| |
Collapse
|