1
|
Ma C, Li H, Li W, Yang G, Chen L. Adaptive Differences in Cellular and Behavioral Responses to Circadian Disruption between C57BL/6 and BALB/c Strains. Int J Mol Sci 2024; 25:10404. [PMID: 39408733 PMCID: PMC11476807 DOI: 10.3390/ijms251910404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
The regulation of the mammalian circadian clock is largely dependent on heredity. In model animals for circadian rhythm studies, C57BL/6 and BALB/c mice exhibit considerable differences in their adaptation to circadian disruption, yet deeper comparisons remain unexplored. Here, we have established embryonic fibroblast cells derived from C57BL/6 mice (MEF) and BALB/c (BALB/3T3) mice, which have been transfected with the Bmal1 promoter-driven luciferase (Bmal1-Luc) reporter gene. Next, dexamethasone was applied for various cyclic stimulations, which revealed that Bmal1 bioluminescence of MEF cells was entrained to 24 to 26 h cycles, whereas BALB/3T3 cells have a wider range (22 to 28 h) with lower amplitudes. Behaviorally, BALB/c mice swiftly adapted to a 6-h advance light/dark cycle, unlike C57BL/6 mice. Furthermore, we found the expression of the circadian rhythm gene Npas2 in BALB/c mice is significantly lower than that in C57BL/6 mice. This observation is consistent with the differentially expressed genes (DEGs) in the intestine and lung tissues of C57BL/6 and BALB/c mice, based on the RNA-seq datasets downloaded from the Gene Expression Omnibus (GEO). In summary, our study uncovers that BALB/c mice possess greater resilience in circadian rhythm than C57BL/6 mice, both cellular and behaviorally, identifying potential genes underlying this difference.
Collapse
Affiliation(s)
- Changxiao Ma
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (C.M.); (H.L.)
- Health Science Center, East China Normal University, Shanghai 200241, China;
| | - Haonan Li
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China; (C.M.); (H.L.)
| | - Wenyu Li
- Health Science Center, East China Normal University, Shanghai 200241, China;
| | - Guangrui Yang
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China
| | - Lihong Chen
- Health Science Center, East China Normal University, Shanghai 200241, China;
| |
Collapse
|
2
|
Qi D, Huang D, Ba M, Xuan S, Si H, Lu D, Pei X, Zhang W, Huang S, Li Z. Long-term high fructose intake reprograms the circadian transcriptome and disrupts homeostasis in mouse extra-orbital lacrimal glands. Exp Eye Res 2024; 246:110008. [PMID: 39025460 DOI: 10.1016/j.exer.2024.110008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
This study aims to explore the effects of long-term high fructose intake (LHFI) on the structure, functionality, and physiological homeostasis of mouse extra-orbital lacrimal glands (ELGs), a critical component of ocular health. Our findings reveal significant reprogramming of the circadian transcriptome in ELGs following LHFI, alongside the activation of specific inflammatory pathways, as well as metabolic and neural pathways. Notably, LHFI resulted in increased inflammatory infiltration, enhanced lipid deposition, and reduced nerve fiber density in ELGs compared to controls. Functional assessments indicated a marked reduction in lacrimal secretion following cholinergic stimulation in LHFI-treated mice, suggesting impaired gland function. Overall, our results suggest that LHFI disrupts lacrimal gland homeostasis, potentially leading to dry eye disease by altering its structure and secretory function. These insights underscore the profound impact of dietary choices on ocular health and highlight the need for strategies to mitigate these risks.
Collapse
Affiliation(s)
- Di Qi
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Duliurui Huang
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Mengru Ba
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Shuting Xuan
- Department of Ophthalmology, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Hongli Si
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Wenxiao Zhang
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450000, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Zhijie Li
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China.
| |
Collapse
|
3
|
Yamaguchi Y. Arginine vasopressin: Critical regulator of circadian homeostasis. Peptides 2024; 177:171229. [PMID: 38663583 DOI: 10.1016/j.peptides.2024.171229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 04/30/2024]
Abstract
Circadian rhythms optimally regulate numerous physiological processes in an organism and synchronize them with the external environment. The suprachiasmatic nucleus (SCN), the center of the circadian clock in mammals, is composed of multiple cell types that form a network that provides the basis for the remarkable stability of the circadian clock. Among the neuropeptides expressed in the SCN, arginine vasopressin (AVP) has attracted much attention because of its deep involvement in the function of circadian rhythms, as elucidated in particular by studies using genetically engineered mice. This review briefly summarizes the current knowledge on the peptidergic distribution and topographic neuronal organization in the SCN, the molecular mechanisms of the clock genes, and the relationship between the SCN and peripheral clocks. With respect to the physiological roles of AVP and AVP-expressing neurons, in addition to a sex-dependent action of AVP in the SCN, studies using AVP receptor knockout mice and mice genetically manipulated to alter the clock properties of AVP neurons are summarized here, highlighting its importance in maintaining circadian homeostasis and its potential as a target for therapeutic interventions.
Collapse
Affiliation(s)
- Yoshiaki Yamaguchi
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Japan.
| |
Collapse
|
4
|
Schmal C. The seasons within: a theoretical perspective on photoperiodic entrainment and encoding. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2024; 210:549-564. [PMID: 37659985 PMCID: PMC11226496 DOI: 10.1007/s00359-023-01669-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 09/04/2023]
Abstract
Circadian clocks are internal timing devices that have evolved as an adaption to the omnipresent natural 24 h rhythmicity of daylight intensity. Properties of the circadian system are photoperiod dependent. The phase of entrainment varies systematically with season. Plastic photoperiod-dependent re-arrangements in the mammalian circadian core pacemaker yield an internal representation of season. Output pathways of the circadian clock regulate photoperiodic responses such as flowering time in plants or hibernation in mammals. Here, we review the concepts of seasonal entrainment and photoperiodic encoding. We introduce conceptual phase oscillator models as their high level of abstraction, but, yet, intuitive interpretation of underlying parameters allows for a straightforward analysis of principles that determine entrainment characteristics. Results from this class of models are related and discussed in the context of more complex conceptual amplitude-phase oscillators as well as contextual molecular models that take into account organism, tissue, and cell-type-specific details.
Collapse
Affiliation(s)
- Christoph Schmal
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Philippstr. 13, 10115, Berlin, Germany.
| |
Collapse
|
5
|
Roy RK, Yao Y, Green IK, Aitken AV, Biancardi VC, Silver R, Stern JE. Blood flows from the SCN toward the OVLT within a new brain vascular portal pathway. SCIENCE ADVANCES 2024; 10:eadn8350. [PMID: 38905332 PMCID: PMC11192075 DOI: 10.1126/sciadv.adn8350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/16/2024] [Indexed: 06/23/2024]
Abstract
The suprachiasmatic nucleus (SCN) sets the phase of oscillation throughout the brain and body. Anatomical evidence reveals a portal system linking the SCN and the organum vasculosum of the lamina terminalis (OVLT), begging the question of the direction of blood flow and the nature of diffusible signals that flow in this specialized vasculature. Using a combination of anatomical and in vivo two-photon imaging approaches, we unequivocally show that blood flows unidirectionally from the SCN to the OVLT, that blood flow rate displays daily oscillations with a higher rate at night than in the day, and that circulating vasopressin can access portal vessels. These findings highlight a previously unknown central nervous system communication pathway, which, like that of the pituitary portal system, could allow neurosecretions to reach nearby target sites in OVLT, avoiding dilution in the systemic blood. In both of these brain portal pathways, the target sites relay signals broadly to both the brain and the rest of the body.
Collapse
Affiliation(s)
- Ranjan K. Roy
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
| | - Yifan Yao
- Department of Psychology, Columbia University, New York, NY, USA
| | | | - Andrew V. Aitken
- Department of Anatomy, Physiology and Pharmacology, Auburn University, Auburn, AL, USA
| | - Vinicia C. Biancardi
- Department of Anatomy, Physiology and Pharmacology, Auburn University, Auburn, AL, USA
| | - Rae Silver
- Department of Psychology, Columbia University, New York, NY, USA
- Department of Neuroscience and Behavior, Barnard College, New York, NY, USA
- Department of Pathology and Cell Biology Graduate Program, Columbia University, New York, NY, USA
- Zukerman Institute Affiliate, Columbia University, New York, NY, USA
| | - Javier E. Stern
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
6
|
Otsuka K, Beaty LA, Sato M, Shitakura K, Kikuchi T, Okajima K, Terada S, Cornelissen G. Chronobioethics: Symphony of biological clocks observed by 7-day/24-hour ambulatory blood pressure monitoring and cardiovascular health. Biomed J 2024:100753. [PMID: 38906327 DOI: 10.1016/j.bj.2024.100753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND The high prevalence of desynchronized biological rhythms is becoming a primary public health concern. We assess complex and diverse inter-modulations among multi-frequency rhythms present in blood pressure (BP) and heart rate (HR). SUBJECTS and Methods: We performed 7-day/24-hour Ambulatory BP Monitoring in 220 (133 women) residents (23 to 74 years) of a rural Japanese town in Kochi Prefecture under everyday life conditions. RESULTS A symphony of biological clocks contributes to the preservation of a synchronized circadian system. (1) Citizens with an average 12.02-h period had fewer vascular variability disorders than those with shorter (11.37-h) or longer (12.88-h) periods (P<0.05), suggesting that the circasemidian rhythm is potentially important for human health. (2) An appropriate BP-HR coupling promoted healthier circadian profiles than a phase-advanced BP: lower 7-day nighttime SBP (106.8 vs. 112.9 mmHg, P=0.0469), deeper nocturnal SBP dip (20.5% vs. 16.8%, P=0.0101), and less frequent incidence of masked non-dipping (0.53 vs. 0.86, P=0.0378), identifying the night as an important time window. CONCLUSION Adaptation to irregular schedules in everyday life occurs unconsciously at night, probably initiated from the brain default mode network, in coordination with the biological clock system, including a reinforced about 12-hour clock, as "a biological clock-guided core integration system".
Collapse
Affiliation(s)
- Kuniaki Otsuka
- Department of Chronomics and Gerontology, Tokyo Women's Medical University, Tokyo, Japan; Halberg Chronobiology Center, University of Minnesota, Minneapolis, MN, USA.
| | - Larry A Beaty
- Halberg Chronobiology Center, University of Minnesota, Minneapolis, MN, USA
| | - Madoka Sato
- Department of Medicine, Jyoban Hospital, Fukushima, Japan
| | - Kazunobu Shitakura
- Cardiovascular Internal Medicine, Higashi Omiya General Hospital, Saitama, Japan
| | - Tomoko Kikuchi
- Cardiovascular Internal Medicine, Higashi Omiya General Hospital, Saitama, Japan
| | - Kiyotaka Okajima
- Cardiovascular Internal Medicine, Higashi Omiya General Hospital, Saitama, Japan
| | - Shigehiko Terada
- Advanced Medical Center, Shonan Kamukura General Hospital, Kanagawa, Japan
| | | |
Collapse
|
7
|
Bonnefont X. Cell Signaling in the Circadian Pacemaker: New Insights from in vivo Imaging. Neuroendocrinology 2024:1-8. [PMID: 38754404 DOI: 10.1159/000539344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/12/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND "One for all, and all for one," the famous rallying cry of the Three Musketeers, in Alexandre Dumas's popular novel, certainly applies to the 20,000 cells composing the suprachiasmatic nuclei (SCN). These cells work together to form the central clock that coordinates body rhythms in tune with the day-night cycle. Like virtually every body cell, individual SCN cells exhibit autonomous circadian oscillations, but this rhythmicity only reaches a high level of precision and robustness when the cells are coupled with their neighbors. Therefore, understanding the functional network organization of SCN cells beyond their core rhythmicity is an important issue in circadian biology. SUMMARY The present review summarizes the main results from our recent study demonstrating the feasibility of recording SCN cells in freely moving mice and the significance of variations in intracellular calcium over several timescales. KEY MESSAGE We discuss how in vivo imaging at the cell level will be pivotal to interrogate the mammalian master clock, in an integrated context that preserves the SCN network organization, with intact inputs and outputs.
Collapse
Affiliation(s)
- Xavier Bonnefont
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- BioCampus Montpellier, Université de Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
8
|
del Olmo M, Schmal C, Mizaikoff C, Grabe S, Gabriel C, Kramer A, Herzel H. Are circadian amplitudes and periods correlated? A new twist in the story. F1000Res 2024; 12:1077. [PMID: 37771612 PMCID: PMC10526121 DOI: 10.12688/f1000research.135533.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/14/2024] [Indexed: 09/30/2023] Open
Abstract
Three parameters are important to characterize a circadian and in general any biological clock: period, phase and amplitude. While circadian periods have been shown to correlate with entrainment phases, and clock amplitude influences the phase response of an oscillator to pulse-like zeitgeber signals, the co-modulations of amplitude and periods, which we term twist, have not been studied in detail. In this paper we define two concepts: parametric twist refers to amplitude-period correlations arising in ensembles of self-sustained, limit cycle clocks in the absence of external inputs, and phase space twist refers to the co-modulation of an individual clock's amplitude and period in response to external zeitgebers. Our findings show that twist influences the interaction of oscillators with the environment, facilitating entrainment, speeding upfastening recovery to pulse-like perturbations or modifying the response of an individual clock to coupling. This theoretical framework might be applied to understand the emerging properties of other oscillating systems.
Collapse
Affiliation(s)
- Marta del Olmo
- Institute for Theoretical Biology, Humboldt Universität zu Berlin and Charité Universitätsmedizin Berlin, Philippstr. 13, 10115 Berlin, Germany
| | - Christoph Schmal
- Institute for Theoretical Biology, Humboldt Universität zu Berlin and Charité Universitätsmedizin Berlin, Philippstr. 13, 10115 Berlin, Germany
| | - Camillo Mizaikoff
- Institute for Theoretical Biology, Humboldt Universität zu Berlin and Charité Universitätsmedizin Berlin, Philippstr. 13, 10115 Berlin, Germany
| | - Saskia Grabe
- Institute for Theoretical Biology, Humboldt Universität zu Berlin and Charité Universitätsmedizin Berlin, Philippstr. 13, 10115 Berlin, Germany
| | - Christian Gabriel
- Laboratory of Chronobiology, Institute for Medical Immunology, Charite Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Achim Kramer
- Laboratory of Chronobiology, Institute for Medical Immunology, Charite Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Hanspeter Herzel
- Institute for Theoretical Biology, Humboldt Universität zu Berlin and Charité Universitätsmedizin Berlin, Philippstr. 13, 10115 Berlin, Germany
| |
Collapse
|
9
|
Lokossou HA, Rabuffo G, Bernard M, Bernard C, Viola A, Perles-Barbacaru TA. Impact of the day/night cycle on functional connectome in ageing male and female mice. Neuroimage 2024; 290:120576. [PMID: 38490583 DOI: 10.1016/j.neuroimage.2024.120576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024] Open
Abstract
To elucidate how time of day, sex, and age affect functional connectivity (FC) in mice, we aimed to examine whether the mouse functional connectome varied with the day/night cycle and whether it depended on sex and age. We explored C57Bl6/J mice (6♀ and 6♂) at mature age (5 ± 1 months) and middle-age (14 ± 1 months). Each mouse underwent Blood Oxygen-Level-Dependent (BOLD) resting-state functional MRI (rs-fMRI) on a 7T scanner at four different times of the day, two under the light condition and two under the dark condition. Data processing consisted of group independent component analysis (ICA) and region-level analysis using resting-state networks (RSNs) derived from literature. Linear mixed-effect models (LMEM) were used to assess the effects of sex, lighting condition and their interactions for each RSN obtained with group-ICA (RSNs-GICA) and six bilateral RSNs adapted from literature (RSNs-LIT). Our study highlighted new RSNs in mice related to day/night alternation in addition to other networks already reported in the literature. In mature mice, we found sex-related differences in brain activation only in one RSNs-GICA comprising the cortical, hippocampal, midbrain and cerebellar regions of the right hemisphere. In males, brain activity was significantly higher in the left hippocampus, the retrosplenial cortex, the superior colliculus, and the cerebellum regardless of lighting condition; consistent with the role of these structures in memory formation and integration, sleep, and sex-differences in memory processing. Experimental constraints limited the analysis to the impact of light/dark cycle on the RSNs for middle-aged females. We detected significant activation in the pineal gland during the dark condition, a finding in line with the nocturnal activity of this gland. For the analysis of RSNs-LIT, new variables "sexage" (sex and age combined) and "edges" (pairs of RSNs) were introduced. FC was calculated as the Pearson correlation between two RSNs. LMEM revealed no effect of sexage or lighting condition. The FC depended on the edges, but there were no interaction effects between sexage, lighting condition and edges. Interaction effects were detected between i) sex and lighting condition, with higher FC in males under the dark condition, ii) sexage and edges with higher FC in male brain regions related to vision, memory, and motor action. We conclude that time of day and sex should be taken into account when designing, analyzing, and interpreting functional imaging studies in rodents.
Collapse
Affiliation(s)
- Houéfa Armelle Lokossou
- Centre for Magnetic Resonance in Biology and Medicine, CRMBM UMR 7339, Aix-Marseille University-CNRS, Marseille, France; Institute of Systems Neuroscience, INS UMR 1106, Aix-Marseille University-INSERM, Marseille, France.
| | - Giovanni Rabuffo
- Institute of Systems Neuroscience, INS UMR 1106, Aix-Marseille University-INSERM, Marseille, France
| | - Monique Bernard
- Centre for Magnetic Resonance in Biology and Medicine, CRMBM UMR 7339, Aix-Marseille University-CNRS, Marseille, France
| | - Christophe Bernard
- Institute of Systems Neuroscience, INS UMR 1106, Aix-Marseille University-INSERM, Marseille, France.
| | - Angèle Viola
- Centre for Magnetic Resonance in Biology and Medicine, CRMBM UMR 7339, Aix-Marseille University-CNRS, Marseille, France
| | | |
Collapse
|
10
|
Smyllie NJ, Hastings MH, Patton AP. Neuron-Astrocyte Interactions and Circadian Timekeeping in Mammals. Neuroscientist 2024:10738584241245307. [PMID: 38602223 DOI: 10.1177/10738584241245307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Almost every facet of our behavior and physiology varies predictably over the course of day and night, anticipating and adapting us to their associated opportunities and challenges. These rhythms are driven by endogenous biological clocks that, when deprived of environmental cues, can continue to oscillate within a period of approximately 1 day, hence circa-dian. Normally, retinal signals synchronize them to the cycle of light and darkness, but disruption of circadian organization, a common feature of modern lifestyles, carries considerable costs to health. Circadian timekeeping pivots around a cell-autonomous molecular clock, widely expressed across tissues. These cellular timers are in turn synchronized by the principal circadian clock of the brain: the hypothalamic suprachiasmatic nucleus (SCN). Intercellular signals make the SCN network a very powerful pacemaker. Previously, neurons were considered the sole SCN timekeepers, with glial cells playing supportive roles. New discoveries have revealed, however, that astrocytes are active partners in SCN network timekeeping, with their cell-autonomous clock regulating extracellular glutamate and GABA concentrations to control circadian cycles of SCN neuronal activity. Here, we introduce circadian timekeeping at the cellular and SCN network levels before focusing on the contributions of astrocytes and their mutual interaction with neurons in circadian control in the brain.
Collapse
Affiliation(s)
- Nicola J Smyllie
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | | | - Andrew P Patton
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| |
Collapse
|
11
|
Ono D, Weaver DR, Hastings MH, Honma KI, Honma S, Silver R. The Suprachiasmatic Nucleus at 50: Looking Back, Then Looking Forward. J Biol Rhythms 2024; 39:135-165. [PMID: 38366616 PMCID: PMC7615910 DOI: 10.1177/07487304231225706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
It has been 50 years since the suprachiasmatic nucleus (SCN) was first identified as the central circadian clock and 25 years since the last overview of developments in the field was published in the Journal of Biological Rhythms. Here, we explore new mechanisms and concepts that have emerged in the subsequent 25 years. Since 1997, methodological developments, such as luminescent and fluorescent reporter techniques, have revealed intricate relationships between cellular and network-level mechanisms. In particular, specific neuropeptides such as arginine vasopressin, vasoactive intestinal peptide, and gastrin-releasing peptide have been identified as key players in the synchronization of cellular circadian rhythms within the SCN. The discovery of multiple oscillators governing behavioral and physiological rhythms has significantly advanced our understanding of the circadian clock. The interaction between neurons and glial cells has been found to play a crucial role in regulating these circadian rhythms within the SCN. Furthermore, the properties of the SCN network vary across ontogenetic stages. The application of cell type-specific genetic manipulations has revealed components of the functional input-output system of the SCN and their correlation with physiological functions. This review concludes with the high-risk effort of identifying open questions and challenges that lie ahead.
Collapse
Affiliation(s)
- Daisuke Ono
- Stress Recognition and Response, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - David R Weaver
- Department of Neurobiology and NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Michael H Hastings
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Ken-Ichi Honma
- Research and Education Center for Brain Science, Hokkaido University, Sapporo, Japan
- Center for Sleep and Circadian Rhythm Disorders, Sapporo Hanazono Hospital, Sapporo, Japan
| | - Sato Honma
- Research and Education Center for Brain Science, Hokkaido University, Sapporo, Japan
- Center for Sleep and Circadian Rhythm Disorders, Sapporo Hanazono Hospital, Sapporo, Japan
| | - Rae Silver
- Stress Recognition and Response, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Neuroscience & Behavior, Barnard College and Department of Psychology, Columbia University, New York City, New York, USA
| |
Collapse
|
12
|
Perisic M, Woolcock K, Hering A, Mendel H, Muttenthaler M. Oxytocin and vasopressin signaling in health and disease. Trends Biochem Sci 2024; 49:361-377. [PMID: 38418338 DOI: 10.1016/j.tibs.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 03/01/2024]
Abstract
Neurohypophysial peptides are ancient and evolutionarily highly conserved neuropeptides that regulate many crucial physiological functions in vertebrates and invertebrates. The human neurohypophysial oxytocin/vasopressin (OT/VP) signaling system with its four receptors has become an attractive drug target for a variety of diseases, including cancer, pain, cardiovascular indications, and neurological disorders. Despite its promise, drug development faces hurdles, including signaling complexity, selectivity and off-target concerns, translational interspecies differences, and inefficient drug delivery. In this review we dive into the complexity of the OT/VP signaling system in health and disease, provide an overview of relevant pharmacological probes, and discuss the latest trends in therapeutic lead discovery and drug development.
Collapse
Affiliation(s)
- Monika Perisic
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Vienna Doctoral School in Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Katrina Woolcock
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Anke Hering
- Institute for Molecular Bioscience, The University of Queensland, 4072 Brisbane, Australia
| | - Helen Mendel
- Institute for Molecular Bioscience, The University of Queensland, 4072 Brisbane, Australia
| | - Markus Muttenthaler
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Institute for Molecular Bioscience, The University of Queensland, 4072 Brisbane, Australia.
| |
Collapse
|
13
|
Del Olmo M, Kalashnikov A, Schmal C, Kramer A, Herzel H. Coupling allows robust mammalian redox circadian rhythms despite heterogeneity and noise. Heliyon 2024; 10:e24773. [PMID: 38312577 PMCID: PMC10835301 DOI: 10.1016/j.heliyon.2024.e24773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 12/06/2023] [Accepted: 01/14/2024] [Indexed: 02/06/2024] Open
Abstract
Circadian clocks are endogenous oscillators present in almost all cells that drive daily rhythms in physiology and behavior. There are two mechanisms that have been proposed to explain how circadian rhythms are generated in mammalian cells: through a transcription-translation feedback loop (TTFL) and based on oxidation/reduction reactions, both of which are intrinsically stochastic and heterogeneous at the single cell level. In order to explore the emerging properties of stochastic and heterogeneous redox oscillators, we simplify a recently developed kinetic model of redox oscillations to an amplitude-phase oscillator with 'twist' (period-amplitude correlation) and subject to Gaussian noise. We show that noise and heterogeneity alone lead to fast desynchronization, and that coupling between noisy oscillators can establish robust and synchronized rhythms with amplitude expansions and tuning of the period due to twist. Coupling a network of redox oscillators to a simple model of the TTFL also contributes to synchronization, large amplitudes and fine-tuning of the period for appropriate interaction strengths. These results provide insights into how the circadian clock compensates randomness from intracellular sources and highlight the importance of noise, heterogeneity and coupling in the context of circadian oscillators.
Collapse
Affiliation(s)
- Marta Del Olmo
- Institute for Theoretical Biology - Humboldt Universität zu Berlin and Charité Universitätsmedizin Berlin, Philippstraße 13, 10115 Berlin, Germany
| | - Anton Kalashnikov
- Institute for Theoretical Biology - Humboldt Universität zu Berlin and Charité Universitätsmedizin Berlin, Philippstraße 13, 10115 Berlin, Germany
| | - Christoph Schmal
- Institute for Theoretical Biology - Humboldt Universität zu Berlin and Charité Universitätsmedizin Berlin, Philippstraße 13, 10115 Berlin, Germany
| | - Achim Kramer
- Institute for Medical Immunology - Laboratory of Chronobiology, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Hanspeter Herzel
- Institute for Theoretical Biology - Humboldt Universität zu Berlin and Charité Universitätsmedizin Berlin, Philippstraße 13, 10115 Berlin, Germany
| |
Collapse
|
14
|
Riedel CS, Georg B, Hannibal J. Phenotyping of light-activated neurons in the mouse SCN based on the expression of FOS and EGR1. Front Physiol 2024; 14:1321007. [PMID: 38317846 PMCID: PMC10839010 DOI: 10.3389/fphys.2023.1321007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/26/2023] [Indexed: 02/07/2024] Open
Abstract
Light-sensitive neurons are located in the ventral and central core of the suprachiasmatic nucleus (SCN), whereas stably oscillating clock neurons are found mainly in the dorsal shell. Signals between the SCN core and shell are believed to play an important role in light entrainment. Core neurons express vasoactive intestinal polypeptide (VIP), gastrin-releasing peptide (GRP), and Neuroglobin (Ngb), whereas the shell neurons express vasopressin (AVP), prokineticin 2, and the VIP type 2 (VPAC2) receptor. In rodents, light has a phase-shifting capacity at night, which induces rapid and transient expression of the EGR1 and FOS in the SCN. Methods: The present study used immunohistochemical staining of FOS, EGR1, and phenotypical markers of SCN neurons (VIP, AVP, Ngb) to identify subtypes/populations of light-responsive neurons at early night. Results: Double immunohistochemistry and cell counting were used to evaluate the number of SCN neurons expressing FOS and EGR1 in the SCN. The number of neurons expressing either EGR1 or FOS was higher than the total number of neurons co-storing EGR1 and FOS. Of the total number of light-responsive cells, 42% expressed only EGR1, 43% expressed only FOS, and 15% expressed both EGR1 and FOS. Light-responsive VIP neurons represented only 31% of all VIP neurons, and EGR1 represents the largest group of light-responsive VIP neurons (18%). VIP neurons expressing only FOS represented 1% of the total light-responsive VIP neurons. 81% of the Ngb neurons in the mouse SCN were light-responsive, and of these neurons expressing only EGR1 after light stimulation represented 44%, whereas 24% expressed FOS. Although most light-responsive neurons are found in the core of the SCN, 29% of the AVP neurons in the shell were light-responsive, of which 8% expressed EGR1, 10% expressed FOS, and 11% co-expressed both EGR1 and FOS after light stimulation. Discussion: Our analysis revealed cell-specific differences in light responsiveness between different peptidergic and Ngb-expressing neurons in different compartments of the mouse SCN, indicating that light activates diverse neuronal networks in the SCN, some of which participate in photoentrainment.
Collapse
Affiliation(s)
| | | | - Jens Hannibal
- Department of Clinical Biochemistry, Faculty of Health Sciences, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Miyake T, Inoue Y, Maekawa Y, Doi M. Circadian Clock and Body Temperature. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1461:177-188. [PMID: 39289281 DOI: 10.1007/978-981-97-4584-5_12] [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 circadian fluctuation of body temperature is one of the most prominent and stable outputs of the circadian clock and plays an important role in maintaining optimal day-night energy homeostasis. The body temperature of homothermic animals is not strictly constant, but it shows daily oscillation within a range of 1-3 °C, which is sufficient to synchronize the clocks of peripheral tissues throughout the body. The thermal entrainment mechanisms of the clock are partly mediated by the action of the heat shock transcription factor and cold-inducible RNA-binding protein-both have the ability to affect clock gene expression. Body temperature in the poikilotherms is not completely passive to the ambient temperature change; they can travel to the place of preferred temperature in a manner depending on the time of their endogenous clock. Based on this behavior-level thermoregulation, flies exhibit a clear body temperature cycle. Noticeably, flies and mice share the same molecular circuit for the controlled body temperature; in both species, the calcitonin receptors participate in the formation of body temperature rhythms during the active phase and exhibit rather specific expression in subsets of clock neurons in the brain. We summarize knowledge on mutual relationships between body temperature regulation and the circadian clock.
Collapse
Affiliation(s)
- Takahito Miyake
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Yuichi Inoue
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Yota Maekawa
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Masao Doi
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan.
| |
Collapse
|
16
|
Hunyara JL, Daly KM, Torres K, Yurgel ME, Komal R, Hattar S, Kolodkin AL. Teneurin-3 regulates the generation of non-image-forming visual circuitry and responsiveness to light in the suprachiasmatic nucleus. PLoS Biol 2023; 21:e3002412. [PMID: 38048352 PMCID: PMC10729976 DOI: 10.1371/journal.pbio.3002412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 12/19/2023] [Accepted: 10/31/2023] [Indexed: 12/06/2023] Open
Abstract
Visual system function depends upon the elaboration of precise connections between retinal ganglion cell (RGC) axons and their central targets in the brain. Though some progress has been made in defining the molecules that regulate RGC connectivity required for the assembly and function of image-forming circuitry, surprisingly little is known about factors required for intrinsically photosensitive RGCs (ipRGCs) to target a principal component of the non-image-forming circuitry: the suprachiasmatic nucleus (SCN). Furthermore, the molecules required for forming circuits critical for circadian behaviors within the SCN are not known. We observe here that the adhesion molecule teneurin-3 (Tenm3) is highly expressed in vasoactive intestinal peptide (VIP) neurons located in the core region of the SCN. Since Tenm3 is required for other aspects of mammalian visual system development, we investigate roles for Tenm3 in regulating ipRGC-SCN connectivity and function. Our results show that Tenm3 negatively regulates association between VIP and arginine vasopressin (AVP) neurons within the SCN and is essential for M1 ipRGC axon innervation to the SCN. Specifically, in Tenm3-/- mice, we find a reduction in ventro-medial innervation to the SCN. Despite this reduction, Tenm3-/- mice have higher sensitivity to light and faster re-entrainment to phase advances, probably due to the increased association between VIP and AVP neurons. These data show that Tenm3 plays key roles in elaborating non-image-forming visual system circuitry and that it influences murine responses to phase-advancing light stimuli.
Collapse
Affiliation(s)
- John L. Hunyara
- The Solomon H. Snyder Department of Neuroscience, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - K. M. Daly
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Katherine Torres
- The Solomon H. Snyder Department of Neuroscience, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Maria E. Yurgel
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ruchi Komal
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Samer Hattar
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alex L. Kolodkin
- The Solomon H. Snyder Department of Neuroscience, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
17
|
Xu W, Li X. Special Issue: Circadian Rhythms and Age Related Disorder: How Does Aging Impact Mammalian Circadian Organization? Adv Biol (Weinh) 2023; 7:e2200219. [PMID: 36449746 DOI: 10.1002/adbi.202200219] [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: 08/07/2022] [Revised: 11/15/2022] [Indexed: 12/03/2022]
Abstract
Aging significantly impacts circadian timing in mammals. The amplitude and precision of behavioral, endocrine, and metabolic rhythms decline with age. This is accompanied with an age-related decline in the amplitude of central pacemaker output, although the molecular clock in the suprachiasmatic nucleus exhibit robust oscillation. Peripheral clocks also exhibit robust oscillation during aging, when extensive reprogramming of other genes' expression rhythms occurs in peripheral tissues. The age-related dissociation between the molecular clock and downstream rhythms in both central and peripheral tissues indicates that mechanisms other than the molecular clock are involved in mediating the impact the aging on circadian organization. In this article, findings are reviewed on the impact of aging on circadian timing functions, and the potential role of increased inflammatory response in age-related changes in circadian organization is highlighted.
Collapse
Affiliation(s)
- Wei Xu
- College of Life Sciences, Wuhan University, Hubei Province, 430072, P. R. China
| | - Xiaodong Li
- College of Life Sciences, Wuhan University, Hubei Province, 430072, P. R. China
| |
Collapse
|
18
|
Yamaguchi Y, Maekawa Y, Kabashima K, Mizuno T, Tainaka M, Suzuki T, Dojo K, Tominaga T, Kuroiwa S, Masubuchi S, Doi M, Tominaga K, Kobayashi K, Yamagata S, Itoi K, Abe M, Schwartz WJ, Sakimura K, Okamura H. An intact pituitary vasopressin system is critical for building a robust circadian clock in the suprachiasmatic nucleus. Proc Natl Acad Sci U S A 2023; 120:e2308489120. [PMID: 37844254 PMCID: PMC10614613 DOI: 10.1073/pnas.2308489120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/12/2023] [Indexed: 10/18/2023] Open
Abstract
The circadian clock is a biological timekeeping system that oscillates with a circa-24-h period, reset by environmental timing cues, especially light, to the 24-h day-night cycle. In mammals, a "central" clock in the hypothalamic suprachiasmatic nucleus (SCN) synchronizes "peripheral" clocks throughout the body to regulate behavior, metabolism, and physiology. A key feature of the clock's oscillation is resistance to abrupt perturbations, but the mechanisms underlying such robustness are not well understood. Here, we probe clock robustness to unexpected photic perturbation by measuring the speed of reentrainment of the murine locomotor rhythm after an abrupt advance of the light-dark cycle. Using an intersectional genetic approach, we implicate a critical role for arginine vasopressin pathways, both central within the SCN and peripheral from the anterior pituitary.
Collapse
Grants
- 18H04015 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 15H05642 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 22K06594 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 22K18384 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 20K20864 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 18002016 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 16H06276 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JPMJCR14W3 MEXT | JST | Core Research for Evolutional Science and Technology (CREST)
- BR220401 MEXT | Japan Society for the Promotion of Science (JSPS)
Collapse
Affiliation(s)
- Yoshiaki Yamaguchi
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto606-8501, Japan
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita564-8680, Japan
| | - Yota Maekawa
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto606-8501, Japan
| | - Kyohei Kabashima
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto606-8501, Japan
| | - Takanobu Mizuno
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto606-8501, Japan
| | - Motomi Tainaka
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto606-8501, Japan
| | - Toru Suzuki
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto606-8501, Japan
| | - Kumiko Dojo
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto606-8501, Japan
| | - Takeichiro Tominaga
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto606-8501, Japan
| | - Sayaka Kuroiwa
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto606-8501, Japan
| | - Satoru Masubuchi
- Department of Physiology, School of Medicine, Aichi Medical University, Nagakute480-1195, Japan
| | - Masao Doi
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto606-8501, Japan
| | - Keiko Tominaga
- Graduate School of Frontier Biosciences, Osaka University, Suita565-0871, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima960-1295, Japan
| | - Satoshi Yamagata
- Graduate School of Information Sciences, Tohoku University, Sendai980-0845, Japan
| | - Keiichi Itoi
- Graduate School of Information Sciences, Tohoku University, Sendai980-0845, Japan
- Department of Nursing, Faculty of Health Sciences, Tohoku Fukushi University, Sendai981-8522, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata951-8585, Japan
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata951-8585, Japan
| | - William J. Schwartz
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX78712
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata951-8585, Japan
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata951-8585, Japan
| | - Hitoshi Okamura
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto606-8501, Japan
- Department of Neuroscience, Graduate School of Medicine, Kyoto University, Kyoto606-8501, Japan
| |
Collapse
|
19
|
Pan D, Wang Z, Chen Y, Cao J. Melanopsin-mediated optical entrainment regulates circadian rhythms in vertebrates. Commun Biol 2023; 6:1054. [PMID: 37853054 PMCID: PMC10584931 DOI: 10.1038/s42003-023-05432-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/09/2023] [Indexed: 10/20/2023] Open
Abstract
Melanopsin (OPN4) is a light-sensitive protein that plays a vital role in the regulation of circadian rhythms and other nonvisual functions. Current research on OPN4 has focused on mammals; more evidence is needed from non-mammalian vertebrates to fully assess the significance of the non-visual photosensitization of OPN4 for circadian rhythm regulation. There are species differences in the regulatory mechanisms of OPN4 for vertebrate circadian rhythms, which may be due to the differences in the cutting variants, tissue localization, and photosensitive activation pathway of OPN4. We here summarize the distribution of OPN4 in mammals, birds, and teleost fish, and the classical excitation mode for the non-visual photosensitive function of OPN4 in mammals is discussed. In addition, the role of OPN4-expressing cells in regulating circadian rhythm in different vertebrates is highlighted, and the potential rhythmic regulatory effects of various neuropeptides or neurotransmitters expressed in mammalian OPN4-expressing ganglion cells are summarized among them.
Collapse
Affiliation(s)
- Deng Pan
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Zixu Wang
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Yaoxing Chen
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Jing Cao
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China.
| |
Collapse
|
20
|
Peng Z, Liang Y, Liu X, Shao J, Hu N, Zhang X. New insights into the mechanisms of diabetic kidney disease: Role of circadian rhythm and Bmal1. Biomed Pharmacother 2023; 166:115422. [PMID: 37660646 DOI: 10.1016/j.biopha.2023.115422] [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: 06/24/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023] Open
Abstract
It is common for diabetic kidney disease (DKD) to be complicated by abnormal blood glucose, blood lipids, and blood pressure rhythms. Thus, it is essential to examine diagnostic and treatment plans from the perspective of circadian disruption. This brief review discusses the clinical relevance of circadian rhythms in DKD and how the core clock gene encoding brain and muscle arnt-like protein 1 (BMAL1) functions owing to the importance of circadian rhythm disruption processes, including the excretion of urinary protein and irregular blood pressure, which occur in DKD. Exploring Bmal1 and its potential mechanisms and signaling pathways in DKD following contact with Sirt1 and NF-κB is novel and important. Finally, potential pharmacological and behavioral intervention strategies for DKD circadian rhythm disturbance are outlined. This review aids in unveiling novel, potential molecular targets for DKD based on circadian rhythms.
Collapse
Affiliation(s)
- Zhimei Peng
- Department of Nephrology, The Second Clinical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China; Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
| | - Yanting Liang
- Department of Nephrology, The Second Clinical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China.
| | - Xueying Liu
- Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
| | - Jie Shao
- Department of Nephrology, The Second Clinical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China.
| | - Nan Hu
- Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
| | - Xinzhou Zhang
- Department of Nephrology, The Second Clinical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China; Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
| |
Collapse
|
21
|
Zhang T, Liu Y, Yang L. Amplitude response and singularity behavior of circadian clock to external stimuli. NPJ Syst Biol Appl 2023; 9:39. [PMID: 37573374 PMCID: PMC10423250 DOI: 10.1038/s41540-023-00300-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/31/2023] [Indexed: 08/14/2023] Open
Abstract
Amplitude changes caused by environmental cues are universal in the circadian clock and associated with various diseases. Singularity behavior, characterized by the disruption of circadian rhythms due to critical stimuli, has been observed across various species. Several mathematical models of the circadian clock have replicated this phenomenon. A comprehensive understanding of the amplitude response remains elusive due to experimental limitations. In this study, we address this question by utilizing a simple normal form model that accurately fits previous experimental data, thereby presenting a general mechanism. We employ a geometric framework to illustrate the dynamics in different stimuli of light-induced transcription (LIT) and light-induced degradation (LID), highlighting the core role of invisible instability in amplitude response. Our model systematically elucidates how stimulus mode, phase, and strength determine amplitude responses. The results show that external stimuli induce alterations in both the amplitudes of individual oscillators and the synchronization among oscillators, collectively influencing the overall amplitude response. While experimental methods impose constraints resulting in limited outcomes under specific conditions, our model provides a comprehensive and three-dimensional mechanistic explanation. A comparison with existing experimental findings demonstrates the consistency of our proposed mechanism. Considering the response direction, the framework enables the identification of phases that lead to increased circadian amplitude. Based on this mechanism derived from the framework, stimulus strategies for resetting circadian rhythms with reduced side effects could be designed. Our results demonstrate that the framework has great potential for understanding and applying stimulus responses in the circadian clock and other limit cycle oscillations.
Collapse
Affiliation(s)
- Tao Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, Jiangsu, China
| | - Yu Liu
- School of Mathematical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Ling Yang
- School of Mathematical Sciences, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
22
|
Li R, Masuda K, Ono D, Kanbayashi T, Hirano A, Sakurai T. Aripiprazole disrupts cellular synchrony in the suprachiasmatic nucleus and enhances entrainment to environmental light-dark cycles in mice. Front Neurosci 2023; 17:1201137. [PMID: 37621713 PMCID: PMC10445652 DOI: 10.3389/fnins.2023.1201137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023] Open
Abstract
Many patients with psychiatric conditions, such as bipolar disorder and major depressive disorder, frequently experience disruptions in their sleep-wake cycles. Several case studies and clinical trials have shown that the administration of aripiprazole, a commonly prescribed antipsychotic drug, alleviates the symptoms of circadian sleep disorders in these patients. This improvement may be attributed to the effects of aripiprazole on the circadian central clock, specifically the hypothalamic suprachiasmatic nucleus (SCN), which regulates various circadian physiological rhythms, including the sleep-wake cycle, in mammals. To examine whether aripiprazole facilitates adaptation to changes in the light-dark cycle, we orally administered aripiprazole to mice and subjected them to jet-lag experiments. Mice receiving aripiprazole were more rapidly entrained to 6 h advanced light-dark cycles. Moreover, we examined the effect of aripiprazole on the cellular rhythms of SCN slice cultures and found that aripiprazole disrupted cellular synchronization in the SCN, thereby accelerating the damping of the SCN rhythm at the population level. Adenosine 3'5' monophosphate (cAMP) assay using a bioluminescence indicator revealed that intracellular cAMP level in the SCN increased following aripiprazole treatment. However, this increase was blocked by pre-treatment with the serotonin 1A receptor (5-HT1AR) antagonist. Based on these findings, we propose that aripiprazole modulates intracellular signaling, including 5-HT1AR-mediated cAMP signaling, and desynchronizes SCN neurons, ultimately leading to enhanced entrainment to phase advanced light-dark cycles in mice. These findings indicate that the improvement in sleep symptoms reported in patients with psychiatric disorders receiving aripiprazole may be due to modulation of the circadian clock. Our study provides novel insights into the potential clinical applications of aripiprazole in patients with various circadian sleep disorders.
Collapse
Affiliation(s)
- Ruoshi Li
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kosaku Masuda
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Takashi Kanbayashi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Ibaraki Prefectural Medical Center of Psychiatry, Kasama, Japan
| | - Arisa Hirano
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takeshi Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
23
|
Tsuno Y, Peng Y, Horike SI, Wang M, Matsui A, Yamagata K, Sugiyama M, Nakamura TJ, Daikoku T, Maejima T, Mieda M. In vivo recording of suprachiasmatic nucleus dynamics reveals a dominant role of arginine vasopressin neurons in circadian pacesetting. PLoS Biol 2023; 21:e3002281. [PMID: 37643163 PMCID: PMC10465001 DOI: 10.1371/journal.pbio.3002281] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 07/28/2023] [Indexed: 08/31/2023] Open
Abstract
The central circadian clock of the suprachiasmatic nucleus (SCN) is a network consisting of various types of neurons and glial cells. Individual cells have the autonomous molecular machinery of a cellular clock, but their intrinsic periods vary considerably. Here, we show that arginine vasopressin (AVP) neurons set the ensemble period of the SCN network in vivo to control the circadian behavior rhythm. Artificial lengthening of cellular periods by deleting casein kinase 1 delta (CK1δ) in the whole SCN lengthened the free-running period of behavior rhythm to an extent similar to CK1δ deletion specific to AVP neurons. However, in SCN slices, PER2::LUC reporter rhythms of these mice only partially and transiently recapitulated the period lengthening, showing a dissociation between the SCN shell and core with a period instability in the shell. In contrast, in vivo calcium rhythms of both AVP and vasoactive intestinal peptide (VIP) neurons in the SCN of freely moving mice demonstrated stably lengthened periods similar to the behavioral rhythm upon AVP neuron-specific CK1δ deletion, without changing the phase relationships between each other. Furthermore, optogenetic activation of AVP neurons acutely induced calcium increase in VIP neurons in vivo. These results indicate that AVP neurons regulate other SCN neurons, such as VIP neurons, in vivo and thus act as a primary determinant of the SCN ensemble period.
Collapse
Affiliation(s)
- Yusuke Tsuno
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yubo Peng
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Shin-ichi Horike
- Division of Integrated Omics Research, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Mohan Wang
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Ayako Matsui
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kanato Yamagata
- Child Brain Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Mizuki Sugiyama
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Takahiro J. Nakamura
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Takiko Daikoku
- Division of Animal Disease Model, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Takashi Maejima
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
24
|
Calligaro H, Shoghi A, Chen X, Kim KY, Yu HL, Khov B, Finander B, Le H, Ellisman MH, Panda S. Ultrastructure of Synaptic Connectivity within Subregions of the Suprachiasmatic Nucleus Revealed by a Genetically Encoded Tag and Serial Blockface Electron Microscopy. eNeuro 2023; 10:ENEURO.0227-23.2023. [PMID: 37500494 PMCID: PMC10449486 DOI: 10.1523/eneuro.0227-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/01/2023] [Indexed: 07/29/2023] Open
Abstract
The hypothalamic suprachiasmatic nucleus (SCN) is the central circadian pacemaker in vertebrates. The SCN receives photic information exclusively through melanopsin-expressing retinal ganglion cells (mRGCs) to synchronize circadian rhythms with the environmental light cycles. The SCN is composed of two major peptidergic neuron types in the core and shell regions of the SCN. Determining how mRGCs interact with the network of synaptic connections onto and between SCN neurons is key to understand how light regulates the circadian clock and to elucidate the relevant local circuits within the SCN. To map these connections, we used a newly developed Cre-dependent electron microscopy (EM) reporter, APEX2, to label the mitochondria of mRGC axons. Serial blockface scanning electron microscopy was then used to resolve the fine 3D structure of mRGC axons and synaptic boutons in the SCN of a male mouse. The resulting maps reveal patterns of connectomic organization in the core and shell of the SCN. We show that these regions are composed of different neuronal subtypes and differ with regard to the pattern of mRGC input, as the shell receives denser mRGC synaptic input compared with the core. This finding challenges the present view that photic information coming directly from the retina is received primarily by the core region of the SCN.
Collapse
Affiliation(s)
- Hugo Calligaro
- Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Azarin Shoghi
- Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Xinyue Chen
- Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Keun-Young Kim
- Department of Neurosciences, University of California at San Diego School of Medicine, La Jolla, CA 92161
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92161
| | - Hsin Liu Yu
- Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Brian Khov
- Salk Institute for Biological Studies, La Jolla, CA 92037
| | | | - Hiep Le
- Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Mark H. Ellisman
- Department of Neurosciences, University of California at San Diego School of Medicine, La Jolla, CA 92161
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92161
| | | |
Collapse
|
25
|
Tu HQ, Li S, Xu YL, Zhang YC, Li PY, Liang LY, Song GP, Jian XX, Wu M, Song ZQ, Li TT, Hu HB, Yuan JF, Shen XL, Li JN, Han QY, Wang K, Zhang T, Zhou T, Li AL, Zhang XM, Li HY. Rhythmic cilia changes support SCN neuron coherence in circadian clock. Science 2023; 380:972-979. [PMID: 37262147 DOI: 10.1126/science.abm1962] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 04/13/2023] [Indexed: 06/03/2023]
Abstract
The suprachiasmatic nucleus (SCN) drives circadian clock coherence through intercellular coupling, which is resistant to environmental perturbations. We report that primary cilia are required for intercellular coupling among SCN neurons to maintain the robustness of the internal clock in mice. Cilia in neuromedin S-producing (NMS) neurons exhibit pronounced circadian rhythmicity in abundance and length. Genetic ablation of ciliogenesis in NMS neurons enabled a rapid phase shift of the internal clock under jet-lag conditions. The circadian rhythms of individual neurons in cilia-deficient SCN slices lost their coherence after external perturbations. Rhythmic cilia changes drive oscillations of Sonic Hedgehog (Shh) signaling and clock gene expression. Inactivation of Shh signaling in NMS neurons phenocopied the effects of cilia ablation. Thus, cilia-Shh signaling in the SCN aids intercellular coupling.
Collapse
Affiliation(s)
- Hai-Qing Tu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Sen Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Ling Xu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Cheng Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Pei-Yao Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Li-Yun Liang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Guang-Ping Song
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Xiao-Xiao Jian
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Min Wu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Zeng-Qing Song
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Ting-Ting Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Huai-Bin Hu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Jin-Feng Yuan
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Xiao-Lin Shen
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Jia-Ning Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Qiu-Ying Han
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Kai Wang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Tao Zhang
- Laboratory Animal Center, Academy of Military Medical Sciences, Beijing, China
| | - Tao Zhou
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
| | - Ai-Ling Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xue-Min Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hui-Yan Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, China
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Patton AP, Morris EL, McManus D, Wang H, Li Y, Chin JW, Hastings MH. Astrocytic control of extracellular GABA drives circadian timekeeping in the suprachiasmatic nucleus. Proc Natl Acad Sci U S A 2023; 120:e2301330120. [PMID: 37186824 PMCID: PMC10214171 DOI: 10.1073/pnas.2301330120] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
The hypothalamic suprachiasmatic nucleus (SCN) is the master mammalian circadian clock. Its cell-autonomous timing mechanism, a transcriptional/translational feedback loop (TTFL), drives daily peaks of neuronal electrical activity, which in turn control circadian behavior. Intercellular signals, mediated by neuropeptides, synchronize and amplify TTFL and electrical rhythms across the circuit. SCN neurons are GABAergic, but the role of GABA in circuit-level timekeeping is unclear. How can a GABAergic circuit sustain circadian cycles of electrical activity, when such increased neuronal firing should become inhibitory to the network? To explore this paradox, we show that SCN slices expressing the GABA sensor iGABASnFR demonstrate a circadian oscillation of extracellular GABA ([GABA]e) that, counterintuitively, runs in antiphase to neuronal activity, with a prolonged peak in circadian night and a pronounced trough in circadian day. Resolving this unexpected relationship, we found that [GABA]e is regulated by GABA transporters (GATs), with uptake peaking during circadian day, hence the daytime trough and nighttime peak. This uptake is mediated by the astrocytically expressed transporter GAT3 (Slc6a11), expression of which is circadian-regulated, being elevated in daytime. Clearance of [GABA]e in circadian day facilitates neuronal firing and is necessary for circadian release of the neuropeptide vasoactive intestinal peptide, a critical regulator of TTFL and circuit-level rhythmicity. Finally, we show that genetic complementation of the astrocytic TTFL alone, in otherwise clockless SCN, is sufficient to drive [GABA]e rhythms and control network timekeeping. Thus, astrocytic clocks maintain the SCN circadian clockwork by temporally controlling GABAergic inhibition of SCN neurons.
Collapse
Affiliation(s)
- Andrew P. Patton
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Emma L. Morris
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - David McManus
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Huan Wang
- State Key Laboratory of Membrane Biology, Peking University, School of Life Sciences, 100871Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University, School of Life Sciences, 100871Beijing, China
| | - Jason W. Chin
- PNAC Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Michael H. Hastings
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| |
Collapse
|
27
|
Li Z, Fu B, Wei A, Wu Y, Huang M, Zhang E, Cui B, Wang B, Peng H. d-Glucosamine induces circadian phase delay by promoting BMAL1 degradation through AMPK/mTOR pathway. Life Sci 2023; 325:121765. [PMID: 37169147 DOI: 10.1016/j.lfs.2023.121765] [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: 03/05/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/13/2023]
Abstract
Circadian rhythms are closely linked to the metabolic network through circadian feedback regulation. The hexosamine biosynthetic pathway (HBP) is a branch of glucose metabolism that affects circadian rhythms through the O-linked N-acetylglucosamine modification (O-GlcNAcylation) of clock proteins. Here, we found out that, among the downstream metabolites regulated by d-glucosamine (GlcN) in HBP salvage pathway, only GlcN is able to induce circadian phase delay both in vitro and in vivo. Mechanistic studies indicated that the phase-shift induced by GlcN is independent of O-GlcNAcylation. Instead, GlcN selectively up-regulates p-AMPK activity, leading to the inhibition of mTOR signaling pathway, and thus down-regulation of p-BMAL1 both in human cell line and mouse tissues. Moreover, GlcN promoted BMAL1 degradation via proteasome pathway. These findings reveal a novel molecular mechanism of GlcN in regulating clock phase and suggest the therapeutic potential of GlcN as new use for an old drug in the future treatment of shift work and circadian misalignment.
Collapse
Affiliation(s)
- Zeqi Li
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Bo Fu
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Aili Wei
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Yanchen Wu
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ming Huang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Enhao Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Bo Cui
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Bo Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Hui Peng
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China; School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
28
|
Xie L, Xiong Y, Ma D, Shi K, Chen J, Yang Q, Yan J. Cholecystokinin neurons in mouse suprachiasmatic nucleus regulate the robustness of circadian clock. Neuron 2023:S0896-6273(23)00301-X. [PMID: 37172583 DOI: 10.1016/j.neuron.2023.04.016] [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: 01/06/2023] [Revised: 03/09/2023] [Accepted: 04/14/2023] [Indexed: 05/15/2023]
Abstract
The suprachiasmatic nucleus (SCN) can generate robust circadian behaviors in mammals under different environments, but the underlying neural mechanisms remained unclear. Here, we showed that the activities of cholecystokinin (CCK) neurons in the mouse SCN preceded the onset of behavioral activities under different photoperiods. CCK-neuron-deficient mice displayed shortened free-running periods, failed to compress their activities under a long photoperiod, and developed rapid splitting or became arrhythmic under constant light. Furthermore, unlike vasoactive intestinal polypeptide (VIP) neurons, CCK neurons are not directly light sensitive, but their activation can elicit phase advance and counter light-induced phase delay mediated by VIP neurons. Under long photoperiods, the impact of CCK neurons on SCN dominates over that of VIP neurons. Finally, we found that the slow-responding CCK neurons control the rate of recovery during jet lag. Together, our results demonstrated that SCN CCK neurons are crucial for the robustness and plasticity of the mammalian circadian clock.
Collapse
Affiliation(s)
- Lucheng Xie
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yangyang Xiong
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Danyi Ma
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kaiwen Shi
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiu Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiaoqiao Yang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jun Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 101408, China.
| |
Collapse
|
29
|
Weigel TK, Guo CL, Güler AD, Ferris HA. Altered circadian behavior and light sensing in mouse models of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.02.539086. [PMID: 37205532 PMCID: PMC10187209 DOI: 10.1101/2023.05.02.539086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Circadian symptoms have long been observed in Alzheimer's disease (AD) and often appear before cognitive symptoms, but the mechanisms underlying circadian alterations in AD are poorly understood. We studied circadian re-entrainment in AD model mice using a "jet lag" paradigm, observing their behavior on a running wheel after a six hour advance in the light:dark cycle. Female 3xTg mice, which carry mutations producing progressive amyloid beta and tau pathology, re-entrained following jet lag more rapidly than age-matched wild type controls at both 8 and 13 months of age. This re-entrainment phenotype has not been previously reported in a murine AD model. Because microglia are activated in AD and in AD models, and inflammation can affect circadian rhythms, we hypothesized that microglia contribute to this re-entrainment phenotype. To test this, we used the colony stimulating factor 1 receptor (CSF1R) inhibitor PLX3397, which rapidly depletes microglia from the brain. Microglia depletion did not alter re-entrainment in either wild type or 3xTg mice, demonstrating that microglia activation is not acutely responsible for the re-entrainment phenotype. To test whether mutant tau pathology is necessary for this behavioral phenotype, we repeated the jet lag behavioral test with the 5xFAD mouse model, which develops amyloid plaques, but not neurofibrillary tangles. As with 3xTg mice, 7-month-old female 5xFAD mice re-entrained more rapidly than controls, demonstrating that mutant tau is not necessary for the re-entrainment phenotype. Because AD pathology affects the retina, we tested whether differences in light sensing may contribute to altered entrainment behavior. 3xTg mice demonstrated heightened negative masking, an SCN-independent circadian behavior measuring responses to different levels of light, and re-entrained dramatically faster than WT mice in a jet lag experiment performed in dim light. 3xTg mice show a heightened sensitivity to light as a circadian cue that may contribute to accelerated photic re-entrainment. Together, these experiments demonstrate novel circadian behavioral phenotypes with heightened responses to photic cues in AD model mice which are not dependent on tauopathy or microglia.
Collapse
|
30
|
Zeman M, Okuliarova M, Rumanova VS. Disturbances of Hormonal Circadian Rhythms by Light Pollution. Int J Mol Sci 2023; 24:ijms24087255. [PMID: 37108420 PMCID: PMC10138516 DOI: 10.3390/ijms24087255] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The circadian rhythms evolved to anticipate and cope with cyclic changes in environmental conditions. This adaptive function is currently compromised by increasing levels of artificial light at night (ALAN), which can represent a risk for the development of diseases of civilisation. The causal links are not completely understood, and this featured review focuses on the chronodisruption of the neuroendocrine control of physiology and behaviour by dim ALAN. The published data indicate that low levels of ALAN (2-5 lux) can attenuate the molecular mechanisms generating circadian rhythms in the central oscillator, eliminate the rhythmic changes in dominant hormonal signals, such as melatonin, testosterone and vasopressin, and interfere with the circadian rhythm of the dominant glucocorticoid corticosterone in rodents. These changes are associated with a disturbed daily pattern of metabolic changes and behavioural rhythms in activity and food and water intake. The increasing levels of ALAN require the identification of the pathways mediating possible negative consequences on health to design effective mitigation strategies to eliminate or minimise the effects of light pollution.
Collapse
Affiliation(s)
- Michal Zeman
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia
| | - Monika Okuliarova
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia
| | - Valentina Sophia Rumanova
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia
| |
Collapse
|
31
|
Murakami A, Tsuji K, Isoda M, Matsuo M, Abe Y, Yasui M, Okamura H, Tominaga K. Prolonged Light Exposure Induces Circadian Impairment in Aquaporin-4-Knockout Mice. J Biol Rhythms 2023; 38:208-214. [PMID: 36694941 DOI: 10.1177/07487304221146242] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Astrocytes are densely present in the suprachiasmatic nucleus (SCN), which is the master circadian oscillator in mammals, and are presumed to play a key role in circadian oscillation. However, specific astrocytic molecules that regulate the circadian clock are not yet well understood. In our study, we found that the water channel aquaporin-4 (AQP4) was abundantly expressed in SCN astrocytes, and we further examined its circadian role using AQP4-knockout mice. There was no prominent difference in circadian behavioral rhythms between Aqp4-/- and Aqp4+/+ mice subjected to light-dark cycles and constant dark conditions. However, exposure to constant light induced a greater decrease in the Aqp4-/- mice rhythmicity. Although the damped rhythm in long-term constant light recovered after transfer to constant dark conditions in both genotypes, the period until the reappearance of original rhythmicity was severely prolonged in Aqp4-/- mice. In conclusion, AQP4 absence exacerbates the prolonged light-induced impairment of circadian oscillations and delays their recovery to normal rhythmicity.
Collapse
Affiliation(s)
- Atsumi Murakami
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Kouki Tsuji
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Japan
| | - Minako Isoda
- Graduate School of Science, Kyoto University, Sakyo-ku, Japan
| | - Masahiro Matsuo
- Department of Psychiatry, Shiga University Graduate School of Medicine, Otsu, Japan
| | - Yoichiro Abe
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
- Keio University Global Research Institute, Center for Water Biology and Medicine, Tokyo, Japan
| | - Masato Yasui
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
- Keio University Global Research Institute, Center for Water Biology and Medicine, Tokyo, Japan
| | - Hitoshi Okamura
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Japan
- Department of Neurobiology, Graduate School of Medicine, Kyoto University, Sakyō-ku, Japan
| | - Keiko Tominaga
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
32
|
Tsuchiya H, Fujinoki M, Azuma M, Koshimizu TA. Vasopressin V1a receptor and oxytocin receptor regulate murine sperm motility differently. Life Sci Alliance 2023; 6:e202201488. [PMID: 36650057 PMCID: PMC9846835 DOI: 10.26508/lsa.202201488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 01/06/2023] [Accepted: 01/06/2023] [Indexed: 01/19/2023] Open
Abstract
Specific receptors for the neurohypophyseal hormones, arginine vasopressin (AVP) and oxytocin, are present in the male reproductive organs. However, their exact roles remain unknown. To elucidate the physiological functions of pituitary hormones in male reproduction, this study first focused on the distribution and function of one of the AVP receptors, V1a. In situ hybridization analysis revealed high expression of the Avpr1a in Leydig cells of the testes and narrow/clear cells in the epididymis, with the expression pattern differing from that of the oxytocin receptor (OTR). Notably, persistent motility and highly proportional hyperactivation were observed in spermatozoa from V1a receptor-deficient mice. In contrast, OTR blocking by antagonist atosiban decreased hyperactivation rate. Furthermore, AVP stimulation could alter the extracellular pH mediated by the V1a receptor. The results highlight the crucial role of neurohypophyseal hormones in male reproductive physiology, with potential contradicting roles of V1a and OTR in sperm maturation. Our findings suggest that V1a receptor antagonists are potential therapeutic drugs for male infertility.
Collapse
Affiliation(s)
- Hiroyoshi Tsuchiya
- Division of Molecular Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke, Japan
| | - Masakatsu Fujinoki
- Research Center for Laboratory Animals, Comprehensive Research Facilities for Advanced Medical Science, School of Medicine, Dokkyo Medical University, Mibu, Japan
| | - Morio Azuma
- Division of Molecular Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke, Japan
| | - Taka-Aki Koshimizu
- Division of Molecular Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke, Japan
| |
Collapse
|
33
|
An extended Hilbert transform method for reconstructing the phase from an oscillatory signal. Sci Rep 2023; 13:3535. [PMID: 36864108 PMCID: PMC9981592 DOI: 10.1038/s41598-023-30405-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
Rhythmic activity is ubiquitous in biological systems from the cellular to organism level. Reconstructing the instantaneous phase is the first step in analyzing the essential mechanism leading to a synchronization state from the observed signals. A popular method of phase reconstruction is based on the Hilbert transform, which can only reconstruct the interpretable phase from a limited class of signals, e.g., narrow band signals. To address this issue, we propose an extended Hilbert transform method that accurately reconstructs the phase from various oscillatory signals. The proposed method is developed by analyzing the reconstruction error of the Hilbert transform method with the aid of Bedrosian's theorem. We validate the proposed method using synthetic data and show its systematically improved performance compared with the conventional Hilbert transform method with respect to accurately reconstructing the phase. Finally, we demonstrate that the proposed method is potentially useful for detecting the phase shift in an observed signal. The proposed method is expected to facilitate the study of synchronization phenomena from experimental data.
Collapse
|
34
|
Rigney N, de Vries GJ, Petrulis A. Modulation of social behavior by distinct vasopressin sources. Front Endocrinol (Lausanne) 2023; 14:1127792. [PMID: 36860367 PMCID: PMC9968743 DOI: 10.3389/fendo.2023.1127792] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
The neuropeptide arginine-vasopressin (AVP) is well known for its peripheral effects on blood pressure and antidiuresis. However, AVP also modulates various social and anxiety-related behaviors by its actions in the brain, often sex-specifically, with effects typically being stronger in males than in females. AVP in the nervous system originates from several distinct sources which are, in turn, regulated by different inputs and regulatory factors. Based on both direct and indirect evidence, we can begin to define the specific role of AVP cell populations in social behavior, such as, social recognition, affiliation, pair bonding, parental behavior, mate competition, aggression, and social stress. Sex differences in function may be apparent in both sexually-dimorphic structures as well as ones without prominent structural differences within the hypothalamus. The understanding of how AVP systems are organized and function may ultimately lead to better therapeutic interventions for psychiatric disorders characterized by social deficits.
Collapse
Affiliation(s)
- Nicole Rigney
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | | | | |
Collapse
|
35
|
Fujioka A, Nagano M, Ikegami K, Masumoto KH, Yoshikawa T, Koinuma S, Nakahama KI, Shigeyoshi Y. Circadian expression and specific localization of synaptotagmin17 in the suprachiasmatic nucleus, the master circadian oscillator in mammals. Brain Res 2023; 1798:148129. [PMID: 36332665 DOI: 10.1016/j.brainres.2022.148129] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/12/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
The localization and function of synaptotagmin (syt)17 in the suprachiasmatic nucleus (SCN) of the brain, which is the master circadian oscillator, were investigated. The Syt17 mRNA-containing neurons were mainly situated in the shell region while SYT17 immunoreactive cell bodies and neural fibers were detected in the core and shell of the SCN and the subparaventricular zone (SPZ). Further, electron microscopy analysis revealed SYT17 in the rough endoplasmic reticulum (rER), Golgi apparatus (G), and large and small vesicles of neurons. Syt17 mRNA expression in the SCN showed a circadian rhythm, and light exposure at night suppressed its expression. In addition, the free running period of locomotor activity rhythm was shortened in Syt17-deletion mutant mice. These findings suggest that SYT17 is involved in the regulation of circadian rhythms.
Collapse
Affiliation(s)
- Atsuko Fujioka
- Department of Anatomy and Neurobiology, Kindai University, Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan
| | - Mamoru Nagano
- Department of Anatomy and Neurobiology, Kindai University, Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan
| | - Keisuke Ikegami
- Department of Anatomy and Neurobiology, Kindai University, Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan
| | - Koh-Hei Masumoto
- Department of Anatomy and Neurobiology, Kindai University, Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan
| | - Tomoko Yoshikawa
- Department of Anatomy and Neurobiology, Kindai University, Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan
| | - Satoshi Koinuma
- Department of Anatomy and Neurobiology, Kindai University, Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan
| | - Ken-Ichi Nakahama
- Department of Cellular Physiological Chemistry, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yasufumi Shigeyoshi
- Department of Anatomy and Neurobiology, Kindai University, Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan.
| |
Collapse
|
36
|
Vasopressin as a Possible Link between Sleep-Disturbances and Memory Problems. Int J Mol Sci 2022; 23:ijms232415467. [PMID: 36555107 PMCID: PMC9778878 DOI: 10.3390/ijms232415467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/18/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Normal biological rhythms, including sleep, are very important for a healthy life and their disturbance may induce-among other issues-memory impairment, which is a key problem of many psychiatric pathologies. The major brain center of circadian regulation is the suprachiasmatic nucleus, and vasopressin (AVP), which is one of its main neurotransmitters, also plays a key role in memory formation. In this review paper, we aimed to summarize our knowledge on the vasopressinergic connection between sleep and memory with the help of the AVP-deficient Brattleboro rat strain. These animals have EEG disturbances with reduced sleep and impaired memory-boosting theta oscillation and show memory impairment in parallel. Based upon human and animal data measuring AVP levels, haplotypes, and the administration of AVP or its agonist or antagonist via different routes (subcutaneous, intraperitoneal, intracerebroventricular, or intranasal), V1a receptors (especially of hippocampal origin) were implicated in the sleep-memory interaction. All in all, the presented data confirm the possible connective role of AVP between biological rhythms and memory formation, thus, supporting the importance of AVP in several psychopathological conditions.
Collapse
|
37
|
Sassenbach L. Identification of novel proteins involved in P2X7-mediated signaling cascades. Purinergic Signal 2022; 18:495-498. [PMID: 35960424 PMCID: PMC9832184 DOI: 10.1007/s11302-022-09893-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 07/28/2022] [Indexed: 01/14/2023] Open
Abstract
High concentration of extracellular ATP acts as a danger signal that is sensed by the P2X7 receptor (P2X7R). This ATP-gated ion channel has been shown to induce multiple metabotropic events such as changes in plasma membrane composition and morphology, ectodomain shedding, activation of lipases, kinases, and transcription factors as well as cytokine release. The specific signaling pathways and molecular mechanisms remain largely obscure. Using an unbiased genome-scale CRISPR/Cas9 screening approach in a murine T cell line, Ryoden et al. (2022, 2020) identified three proteins involved in P2X7 regulation and signaling: Essential for Reactive Oxygen Species (EROS) is essential for P2X7 folding and maturation, and Xk and Vsp13a are required for P2X7-mediated phosphatidyl serine exposure and cell lysis. They further provide evidence for an interaction of Xk and Vsp13a at the plasma membrane and confirm the role of Xk in ATP-induced cytolysis in primary CD25+CD4+ T cells from Xk-/- mice.
Collapse
Affiliation(s)
- Lukas Sassenbach
- Walther-Straub-Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany.
| |
Collapse
|
38
|
Galinde AAS, Al-Mughales F, Oster H, Heyde I. Different levels of circadian (de)synchrony -- where does it hurt? F1000Res 2022; 11:1323. [PMID: 37125019 PMCID: PMC10130703 DOI: 10.12688/f1000research.127234.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
A network of cellular timers ensures the maintenance of homeostasis by temporal modulation of physiological processes across the day. These so-called circadian clocks are synchronized to geophysical time by external time cues (or zeitgebers). In modern societies, natural environmental cycles are disrupted by artificial lighting, around-the-clock availability of food or shiftwork. Such contradictory zeitgeber input promotes chronodisruption, i.e., the perturbation of internal circadian rhythms, resulting in adverse health outcomes. While this phenomenon is well described, it is still poorly understood at which level of organization perturbed rhythms impact on health and wellbeing. In this review, we discuss different levels of chronodisruption and what is known about their health effects. We summarize the results of disrupted phase coherence between external and internal time vs. misalignment of tissue clocks amongst each other, i.e., internal desynchrony. Last, phase incoherence can also occur at the tissue level itself. Here, alterations in phase coordination can emerge between cellular clocks of the same tissue or between different clock genes within the single cell. A better understanding of the mechanisms of circadian misalignment and its effects on physiology will help to find effective tools to prevent or treat disorders arising from modern-day chronodisruptive environments.
Collapse
Affiliation(s)
- Ankita AS. Galinde
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, 23562, Germany
| | - Faheem Al-Mughales
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, 23562, Germany
- Biochemistry Department, Faculty of Medicine and Health Sciences, Taiz University, Taiz, Yemen
| | - Henrik Oster
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, 23562, Germany
| | - Isabel Heyde
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, 23562, Germany
| |
Collapse
|
39
|
Galinde AAS, Al-Mughales F, Oster H, Heyde I. Different levels of circadian (de)synchrony -- where does it hurt? F1000Res 2022; 11:1323. [PMID: 37125019 PMCID: PMC10130703 DOI: 10.12688/f1000research.127234.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 04/05/2023] Open
Abstract
A network of cellular timers ensures the maintenance of homeostasis by temporal modulation of physiological processes across the day. These so-called circadian clocks are synchronized to geophysical time by external time cues (or zeitgebers). In modern societies, natural environmental cycles are disrupted by artificial lighting, around-the-clock availability of food or shift work. Such contradictory zeitgeber input promotes chronodisruption, i.e., the perturbation of internal circadian rhythms, resulting in adverse health outcomes. While this phenomenon is well described, it is still poorly understood at which level of organization perturbed rhythms impact on health and wellbeing. In this review, we discuss different levels of chronodisruption and what is known about their health effects. We summarize the results of disrupted phase coherence between external and internal time vs. misalignment of tissue clocks amongst each other, i.e., internal desynchrony. Last, phase incoherence can also occur at the tissue level itself. Here, alterations in phase coordination can emerge between cellular clocks of the same tissue or between different clock genes within the single cell. A better understanding of the mechanisms of circadian misalignment and its effects on physiology will help to find effective tools to prevent or treat disorders arising from modern-day chronodisruptive environments.
Collapse
Affiliation(s)
- Ankita AS. Galinde
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, 23562, Germany
| | - Faheem Al-Mughales
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, 23562, Germany
- Biochemistry Department, Faculty of Medicine and Health Sciences, Taiz University, Taiz, Yemen
| | - Henrik Oster
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, 23562, Germany
| | - Isabel Heyde
- Institute of Neurobiology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, 23562, Germany
| |
Collapse
|
40
|
Morimoto T, Yoshikawa T, Nagano M, Shigeyoshi Y. Regionality of short and long period oscillators in the suprachiasmatic nucleus and their manner of synchronization. PLoS One 2022; 17:e0276372. [PMID: 36256675 PMCID: PMC9578605 DOI: 10.1371/journal.pone.0276372] [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: 06/06/2022] [Accepted: 10/05/2022] [Indexed: 11/18/2022] Open
Abstract
In mammals, the center of the circadian clock is located in the suprachiasmatic nucleus (SCN) of the hypothalamus. Many studies have suggested that there are multiple regions generating different circadian periods within the SCN, but the exact localization of the regions has not been elucidated. In this study, using a transgenic rat carrying a destabilized luciferase reporter gene driven by a regulatory element of Per2 gene (Per2::dLuc), we investigated the regional variation of period lengths in horizontal slices of the SCN. We revealed a distinct caudal medial region (short period region, SPR) and a rostro-lateral region (long period region, LPR) that generate circadian rhythms with periods shorter than and longer than 24 hours, respectively. We also found that the core region of the SCN marked by dense VIP (vasoactive intestinal peptide) mRNA-expressing neurons covered a part of LPR, and that the shell region of the SCN contains both SPR and the rest of the LPR. Furthermore, we observed how synchronization is achieved between regions generating distinct circadian periods in the SCN. We found that the longer circadian rhythm of the rostral region appears to entrain the circadian rhythm in the caudal region. Our findings clarify the localization of regionality of circadian periods and the mechanism by which the integrated circadian rhythm is formed in the SCN.
Collapse
Affiliation(s)
- Tadamitsu Morimoto
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Kindai University, Osaka-Sayama, Osaka, Japan
| | - Tomoko Yoshikawa
- Organization for International Education and Exchange, University of Toyama, Toyama, Japan
| | - Mamoru Nagano
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Kindai University, Osaka-Sayama, Osaka, Japan
| | - Yasufumi Shigeyoshi
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Kindai University, Osaka-Sayama, Osaka, Japan,* E-mail:
| |
Collapse
|
41
|
Costello HM, Johnston JG, Juffre A, Crislip GR, Gumz ML. Circadian clocks of the kidney: function, mechanism, and regulation. Physiol Rev 2022; 102:1669-1701. [PMID: 35575250 PMCID: PMC9273266 DOI: 10.1152/physrev.00045.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/03/2022] [Accepted: 05/07/2022] [Indexed: 11/22/2022] Open
Abstract
An intrinsic cellular circadian clock is located in nearly every cell of the body. The peripheral circadian clocks within the cells of the kidney contribute to the regulation of a variety of renal processes. In this review, we summarize what is currently known regarding the function, mechanism, and regulation of kidney clocks. Additionally, the effect of extrarenal physiological processes, such as endocrine and neuronal signals, on kidney function is also reviewed. Circadian rhythms in renal function are an integral part of kidney physiology, underscoring the importance of considering time of day as a key biological variable. The field of circadian renal physiology is of tremendous relevance, but with limited physiological and mechanistic information on the kidney clocks this is an area in need of extensive investigation.
Collapse
Affiliation(s)
- Hannah M Costello
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Jermaine G Johnston
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida
| | - Alexandria Juffre
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
| | - G Ryan Crislip
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
| | - Michelle L Gumz
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida
- North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, Florida
| |
Collapse
|
42
|
Roberts NT, MacDonald CR, Mohammadpour H, Antoch MP, Repasky EA. Circadian Rhythm Disruption Increases Tumor Growth Rate and Accumulation of Myeloid-Derived Suppressor Cells. Adv Biol (Weinh) 2022; 6:e2200031. [PMID: 35652494 PMCID: PMC9474681 DOI: 10.1002/adbi.202200031] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/05/2022] [Indexed: 01/28/2023]
Abstract
Circadian rhythm disruption is implicated in the initiation and progression of many diseases, including cancer. External stimuli, such as sunlight, serve to synchronize physiological processes and cellular functions to a 24-h cycle. The immune system is controlled by circadian rhythms, and perturbation of these rhythms can potentially alter the immune response to infections and tumors. The effect of circadian rhythm disruption on the immune response to tumors remains unclear. Specifically, the effects of circadian disruption (CD) on immunosuppressive cell types within the tumor, such as myeloid-derived suppressor cells (MDSCs), are unknown. In this study, a shifting lighting schedule is used to disrupt the circadian rhythm of mice. After acclimation to lighting schedules, mice are inoculated with 4T1 or B16-F10 tumors. Tumor growth is increased in mice housed under circadian disrupting lighting conditions compared to standard lighting conditions. Analysis of immune populations within the spleen and tumor shows an increased accumulation of MDSCs within these tissues, suggesting that MDSC mediated immunosuppression plays a role in the enhanced tumor growth caused by circadian disruption. This paves the way for future studies of the effects of CD on immunosuppression in cancer.
Collapse
Affiliation(s)
- Nathan T. Roberts
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Cameron R. MacDonald
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Hemn Mohammadpour
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Marina P. Antoch
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, 665 Elm St, Buffalo, NY 14203, USA
| | - Elizabeth A. Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| |
Collapse
|
43
|
Singla R, Mishra A, Cao R. The trilateral interactions between mammalian target of rapamycin (mTOR) signaling, the circadian clock, and psychiatric disorders: an emerging model. Transl Psychiatry 2022; 12:355. [PMID: 36045116 PMCID: PMC9433414 DOI: 10.1038/s41398-022-02120-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 08/08/2022] [Accepted: 08/12/2022] [Indexed: 02/07/2023] Open
Abstract
Circadian (~24 h) rhythms in physiology and behavior are evolutionarily conserved and found in almost all living organisms. The rhythms are endogenously driven by daily oscillatory activities of so-called "clock genes/proteins", which are widely distributed throughout the mammalian brain. Mammalian (mechanistic) target of rapamycin (mTOR) signaling is a fundamental intracellular signal transduction cascade that controls important neuronal processes including neurodevelopment, synaptic plasticity, metabolism, and aging. Dysregulation of the mTOR pathway is associated with psychiatric disorders including autism spectrum disorders (ASD) and mood disorders (MD), in which patients often exhibit disrupted daily physiological rhythms and abnormal circadian gene expression in the brain. Recent work has found that the activities of mTOR signaling are temporally controlled by the circadian clock and exhibit robust circadian oscillations in multiple systems. In the meantime, mTOR signaling regulates fundamental properties of the central and peripheral circadian clocks, including period length, entrainment, and synchronization. Whereas the underlying mechanisms remain to be fully elucidated, increasing clinical and preclinical evidence support significant crosstalk between mTOR signaling, the circadian clock, and psychiatric disorders. Here, we review recent progress in understanding the trilateral interactions and propose an "interaction triangle" model between mTOR signaling, the circadian clock, and psychiatric disorders (focusing on ASD and MD).
Collapse
Affiliation(s)
- Rubal Singla
- grid.17635.360000000419368657Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812 USA
| | - Abhishek Mishra
- grid.17635.360000000419368657Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812 USA
| | - Ruifeng Cao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA. .,Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
| |
Collapse
|
44
|
The duper mutation reveals previously unsuspected functions of Cryptochrome 1 in circadian entrainment and heart disease. Proc Natl Acad Sci U S A 2022; 119:e2121883119. [PMID: 35930669 PMCID: PMC9371649 DOI: 10.1073/pnas.2121883119] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The Cryptochrome 1 (Cry1)-deficient duper mutant hamster has a short free-running period in constant darkness (τDD) and shows large phase shifts in response to brief light pulses. We tested whether this measure of the lability of the circadian phase is a general characteristic of Cry1-null animals and whether it indicates resistance to jet lag. Upon advance of the light:dark (LD) cycle, both duper hamsters and Cry1-/- mice re-entrained locomotor rhythms three times as fast as wild types. However, accelerated re-entrainment was dissociated from the amplified phase-response curve (PRC): unlike duper hamsters, Cry1-/- mice show no amplification of the phase response to 15' light pulses. Neither the amplified acute shifts nor the increased rate of re-entrainment in duper mutants is due to acceleration of the circadian clock: when mutants drank heavy water to lengthen the period, these aspects of the phenotype persisted. In light of the health consequences of circadian misalignment, we examined effects of duper and phase shifts on a hamster model of heart disease previously shown to be aggravated by repeated phase shifts. The mutation shortened the lifespan of cardiomyopathic hamsters relative to wild types, but this effect was eliminated when mutants experienced 8-h phase shifts every second week, to which they rapidly re-entrained. Our results reveal previously unsuspected roles of Cry1 in phase shifting and longevity in the face of heart disease. The duper mutant offers new opportunities to understand the basis of circadian disruption and jet lag.
Collapse
|
45
|
Circadian protection against bacterial skin infection by epidermal CXCL14-mediated innate immunity. Proc Natl Acad Sci U S A 2022; 119:e2116027119. [PMID: 35704759 DOI: 10.1073/pnas.2116027119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The epidermis is the outermost layer of the skin and the body's primary barrier to external pathogens; however, the early epidermal immune response remains to be mechanistically understood. We show that the chemokine CXCL14, produced by epidermal keratinocytes, exhibits robust circadian fluctuations and initiates innate immunity. Clearance of the skin pathogen Staphylococcus aureus in nocturnal mice was associated with CXCL14 expression, which was high during subjective daytime and low at night. In contrast, in marmosets, a diurnal primate, circadian CXCL14 expression was reversed. Rhythmically expressed CXCL14 binds to S. aureus DNA and induces inflammatory cytokine production by activating Toll-like receptor (TLR)9-dependent innate pathways in dendritic cells and macrophages underneath the epidermis. CXCL14 also promoted phagocytosis by macrophages in a TLR9-independent manner. These data indicate that circadian production of the epidermal chemokine CXCL14 rhythmically suppresses skin bacterial proliferation in mammals by activating the innate immune system.
Collapse
|
46
|
Kim H, Min C, Jeong B, Lee KJ. Deciphering clock cell network morphology within the biological master clock, suprachiasmatic nucleus: From the perspective of circadian wave dynamics. PLoS Comput Biol 2022; 18:e1010213. [PMID: 35666776 PMCID: PMC9203024 DOI: 10.1371/journal.pcbi.1010213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 06/16/2022] [Accepted: 05/16/2022] [Indexed: 11/18/2022] Open
Abstract
The biological master clock, suprachiasmatic nucleus (of rat and mouse), is composed of ~10,000 clock cells which are heterogeneous with respect to their circadian periods. Despite this inhomogeneity, an intact SCN maintains a very good degree of circadian phase (time) coherence which is vital for sustaining various circadian rhythmic activities, and it is supposedly achieved by not just one but a few different cell-to-cell coupling mechanisms, among which action potential (AP)-mediated connectivity is known to be essential. But, due to technical difficulties and limitations in experiments, so far very little information is available about the morphology of the connectivity at a cellular scale. Building upon this limited amount of information, here we exhaustively and systematically explore a large pool (~25,000) of various network morphologies to come up with some plausible network features of SCN networks. All candidates under consideration reflect an experimentally obtained 'indegree distribution' as well as a 'physical range distribution of afferent clock cells.' Then, importantly, with a set of multitude criteria based on the properties of SCN circadian phase waves in extrinsically perturbed as well as in their natural states, we select out appropriate model networks: Some important measures are, 1) level of phase dispersal and direction of wave propagation, 2) phase-resetting ability of the model networks subject to external circadian forcing, and 3) decay rate of perturbation induced "phase-singularities." The successful, realistic networks have several common features: 1) "indegree" and "outdegree" should have a positive correlation; 2) the cells in the SCN ventrolateral region (core) have a much larger total degree than that of the dorsal medial region (shell); 3) The number of intra-core edges is about 7.5 times that of intra-shell edges; and 4) the distance probability density function for the afferent connections fits well to a beta function. We believe that these newly identified network features would be a useful guide for future explorations on the very much unknown AP-mediated clock cell connectome within the SCN.
Collapse
Affiliation(s)
- Hyun Kim
- Department of Physics, Korea University, Seoul, Korea
| | - Cheolhong Min
- Department of Physics, Korea University, Seoul, Korea
| | - Byeongha Jeong
- University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Kyoung J. Lee
- Department of Physics, Korea University, Seoul, Korea
| |
Collapse
|
47
|
Huang S, Si H, Liu J, Qi D, Pei X, Lu D, Zou S, Li Z. Sleep Loss Causes Dysfunction in Murine Extraorbital Lacrimal Glands. Invest Ophthalmol Vis Sci 2022; 63:19. [PMID: 35731510 PMCID: PMC9233287 DOI: 10.1167/iovs.63.6.19] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Purpose Sleep loss markedly affects the structure and function of the lacrimal gland and may cause ocular surface disease as a common public health problem. This study aims to investigate the circadian disturbance caused by sleep loss leading to dysfunction of extraorbital lacrimal glands (ELGs). Methods A mouse sleep deprivation (SD) model for sleep loss studies was built in C57BL/6J male mice. After four weeks, the ELGs were collected at three-hour intervals during a 24-hour period. The Jonckheere-Terpstra-Kendall algorithm was used to determine the composition, phase, and rhythmicity of transcriptomic profiles in ELGs. Furthermore, we compared the non-sleep-deprived and SD-treated mouse ELG (i) reactive oxygen species (ROS) by fluorescein staining, (ii) DNA damage by immunostaining for γ-H2Ax, and (iii) circadian migration of immune cells by immunostaining for CD4, CD8, γδ-TCR, CD64, and CX3CR1. Finally, we also evaluated (i) the locomotor activity and core body temperature rhythm of mice and (ii) the mass, cell size, and tear secretion of the ELGs. Results SD dramatically altered the composition and phase-associated functional enrichment of the circadian transcriptome, immune cell trafficking, metabolism, cell differentiation, and neural secretory activities of mouse ELGs. Additionally, SD caused the ROS accumulation and consequent DNA damage in the ELGs, and the ELG dysfunction caused by SD was irreversible. Conclusions SD damages the structure, function, and diurnal oscillations of ELGs. These results highlight comprehensive characterization of insufficient sleep–affected ELG circadian transcriptome that may provide a new therapeutic approach to counteract the effects of SD on ELG function.
Collapse
Affiliation(s)
- Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongli Si
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiangman Liu
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Di Qi
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Sen Zou
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhijie Li
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
48
|
A light-induced small G-protein gem limits the circadian clock phase-shift magnitude by inhibiting voltage-dependent calcium channels. Cell Rep 2022; 39:110844. [PMID: 35613591 DOI: 10.1016/j.celrep.2022.110844] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/09/2022] [Accepted: 04/28/2022] [Indexed: 11/23/2022] Open
Abstract
Calcium signaling is pivotal to the circadian clockwork in the suprachiasmatic nucleus (SCN), particularly in rhythm entrainment to environmental light-dark cycles. Here, we show that a small G-protein Gem, an endogenous inhibitor of high-voltage-activated voltage-dependent calcium channels (VDCCs), is rapidly induced by light in SCN neurons via the calcium (Ca2+)-mediated CREB/CRE transcriptional pathway. Gem attenuates light-induced calcium signaling through its interaction with VDCCs. The phase-shift magnitude of locomotor activity rhythms by light, at night, increases in Gem-deficient (Gem-/-) mice. Similarly, in SCN slices from Gem-/- mice, depolarizing stimuli induce larger phase shifts of clock gene transcription rhythms that are normalized by the application of an L-type VDCC blocker, nifedipine. Voltage-clamp recordings from SCN neurons reveal that Ca2+ currents through L-type channels increase in Gem-/- mice. Our findings suggest that transcriptionally activated Gem feeds back to suppress excessive light-evoked L-type VDCC activation, adjusting the light-induced phase-shift magnitude to an appropriate level in mammals.
Collapse
|
49
|
Yalçin M, Mundorf A, Thiel F, Amatriain-Fernández S, Kalthoff IS, Beucke JC, Budde H, Garthus-Niegel S, Peterburs J, Relógio A. It's About Time: The Circadian Network as Time-Keeper for Cognitive Functioning, Locomotor Activity and Mental Health. Front Physiol 2022; 13:873237. [PMID: 35547585 PMCID: PMC9081535 DOI: 10.3389/fphys.2022.873237] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/08/2022] [Indexed: 12/24/2022] Open
Abstract
A variety of organisms including mammals have evolved a 24h, self-sustained timekeeping machinery known as the circadian clock (biological clock), which enables to anticipate, respond, and adapt to environmental influences such as the daily light and dark cycles. Proper functioning of the clock plays a pivotal role in the temporal regulation of a wide range of cellular, physiological, and behavioural processes. The disruption of circadian rhythms was found to be associated with the onset and progression of several pathologies including sleep and mental disorders, cancer, and neurodegeneration. Thus, the role of the circadian clock in health and disease, and its clinical applications, have gained increasing attention, but the exact mechanisms underlying temporal regulation require further work and the integration of evidence from different research fields. In this review, we address the current knowledge regarding the functioning of molecular circuits as generators of circadian rhythms and the essential role of circadian synchrony in a healthy organism. In particular, we discuss the role of circadian regulation in the context of behaviour and cognitive functioning, delineating how the loss of this tight interplay is linked to pathological development with a focus on mental disorders and neurodegeneration. We further describe emerging new aspects on the link between the circadian clock and physical exercise-induced cognitive functioning, and its current usage as circadian activator with a positive impact in delaying the progression of certain pathologies including neurodegeneration and brain-related disorders. Finally, we discuss recent epidemiological evidence pointing to an important role of the circadian clock in mental health.
Collapse
Affiliation(s)
- Müge Yalçin
- Institute for Theoretical Biology (ITB), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Annakarina Mundorf
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Freya Thiel
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
- Institute and Policlinic of Occupational and Social Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sandra Amatriain-Fernández
- Institute for Systems Medicine and Faculty of Human Sciences, MSH Medical School Hamburg, Hamburg, Germany
| | - Ida Schulze Kalthoff
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Jan-Carl Beucke
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Henning Budde
- Institute for Systems Medicine and Faculty of Human Sciences, MSH Medical School Hamburg, Hamburg, Germany
| | - Susan Garthus-Niegel
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
- Institute and Policlinic of Occupational and Social Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Department of Child Health and Development, Norwegian Institute of Public Health, Oslo, Norway
| | - Jutta Peterburs
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Angela Relógio
- Institute for Theoretical Biology (ITB), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| |
Collapse
|
50
|
UBR4/POE facilitates secretory trafficking to maintain circadian clock synchrony. Nat Commun 2022; 13:1594. [PMID: 35332162 PMCID: PMC8948264 DOI: 10.1038/s41467-022-29244-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 03/02/2022] [Indexed: 11/08/2022] Open
Abstract
Ubiquitin ligases control the degradation of core clock proteins to govern the speed and resetting properties of the circadian pacemaker. However, few studies have addressed their potential to regulate other cellular events within clock neurons beyond clock protein turnover. Here, we report that the ubiquitin ligase, UBR4/POE, strengthens the central pacemaker by facilitating neuropeptide trafficking in clock neurons and promoting network synchrony. Ubr4-deficient mice are resistant to jetlag, whereas poe knockdown flies are prone to arrhythmicity, behaviors reflective of the reduced axonal trafficking of circadian neuropeptides. At the cellular level, Ubr4 ablation impairs the export of secreted proteins from the Golgi apparatus by reducing the expression of Coronin 7, which is required for budding of Golgi-derived transport vesicles. In summary, UBR4/POE fulfills a conserved and unexpected role in the vesicular trafficking of neuropeptides, a function that has important implications for circadian clock synchrony and circuit-level signal processing. Although ubiquitin ligases are known to control clock protein degradation, their other roles in clock neurons are unclear. Here the authors report that UBR4 promotes export of neuropeptides from the Golgi for axonal trafficking, which is important for circadian clock synchrony in mice and flies.
Collapse
|