1
|
Ness N, Díaz-Clavero S, Hoekstra MMB, Brancaccio M. Rhythmic astrocytic GABA production synchronizes neuronal circadian timekeeping in the suprachiasmatic nucleus. EMBO J 2025; 44:356-381. [PMID: 39623138 PMCID: PMC11731042 DOI: 10.1038/s44318-024-00324-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 10/18/2024] [Accepted: 11/04/2024] [Indexed: 01/15/2025] Open
Abstract
Astrocytes of the suprachiasmatic nucleus (SCN) can regulate sleep-wake cycles in mammals. However, the nature of the information provided by astrocytes to control circadian patterns of behavior is unclear. Neuronal circadian activity across the SCN is organized into spatiotemporal waves that govern seasonal adaptations and timely engagement of behavioral outputs. Here, we show that astrocytes across the mouse SCN exhibit instead a highly uniform, pulse-like nighttime activity. We find that rhythmic astrocytic GABA production via polyamine degradation provides an inhibitory nighttime tone required for SCN circuit synchrony, thereby acting as an internal astrocyte zeitgeber (or "astrozeit"). We further identify synaptic GABA and astrocytic GABA as two key players underpinning coherent spatiotemporal circadian patterns of SCN neuronal activity. In describing a new mechanism by which astrocytes contribute to circadian timekeeping, our work provides a general blueprint for understanding how astrocytes encode temporal information underlying complex behaviors in mammals.
Collapse
Affiliation(s)
- Natalie Ness
- Department of Brain Science, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Sandra Díaz-Clavero
- Department of Brain Science, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Marieke M B Hoekstra
- Department of Brain Science, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Marco Brancaccio
- Department of Brain Science, Imperial College London, London, UK.
- UK Dementia Research Institute at Imperial College London, London, UK.
| |
Collapse
|
2
|
Klett N, Gompf HS, Allen CN, Cravetchi O, Hablitz LM, Gunesch AN, Irwin RP, Todd WD, Saper CB, Fuller PM. GABAergic signalling in the suprachiasmatic nucleus is required for coherent circadian rhythmicity. Eur J Neurosci 2024; 60:6652-6667. [PMID: 39558544 PMCID: PMC11612841 DOI: 10.1111/ejn.16582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 09/11/2024] [Accepted: 10/07/2024] [Indexed: 11/20/2024]
Abstract
The suprachiasmatic nucleus is the circadian pacemaker of the mammalian brain. Suprachiasmatic nucleus neurons display synchronization of their firing frequency on a circadian timescale, which is required for the pacemaker function of the suprachiasmatic nucleus. However, the mechanisms by which suprachiasmatic nucleus neurons remain synchronized in vivo are poorly understood, although synaptic communication is considered indispensable. Suprachiasmatic nucleus neurons contain the neurotransmitter GABA and express GABA receptors. This has inspired the hypothesis that GABA signalling may play a central role in network synchronization, although this remains untested in vivo. Here, using local genetic deletion, we show that disruption of GABA synaptic transmission within the suprachiasmatic nucleus of adult mice results in the eventual deterioration of physiological and behavioural rhythmicity in vivo and concomitant cellular desynchrony in vitro. These findings suggest that intercellular GABA signalling is essential for behavioural rhythmicity and cellular synchrony of the suprachiasmatic nucleus neural network.
Collapse
Affiliation(s)
- Nathan Klett
- Oregon Institute for Occupational Health SciencesUSA
- Neuroscience Graduate ProgramUSA
| | - Heinrich S. Gompf
- Department of Neurological SurgeryUniversity of California, DavisDavisCAUSA
| | - Charles N. Allen
- Oregon Institute for Occupational Health SciencesUSA
- Department of Behavioral NeuroscienceOregon Health & Science UniversityPortlandORUSA
| | | | - Lauren M. Hablitz
- Oregon Institute for Occupational Health SciencesUSA
- Department of Behavioral NeuroscienceOregon Health & Science UniversityPortlandORUSA
- Present address:
Center for Translational NeuromedicineUniversity of Rochester Medical CenterRochesterNYUSA
| | | | | | - William D. Todd
- Department of Neurology, Division of Sleep Medicine, and Program in NeuroscienceBeth Israel Deaconess Medical Center, Harvard Medical School BostonMAUSA
- Present address:
Department of Zoology and PhysiologyUniversity of WyomingLaramieWYUSA
| | - Clifford B. Saper
- Department of Neurology, Division of Sleep Medicine, and Program in NeuroscienceBeth Israel Deaconess Medical Center, Harvard Medical School BostonMAUSA
| | - Patrick M. Fuller
- Department of Neurological SurgeryUniversity of California, DavisDavisCAUSA
| |
Collapse
|
3
|
Thompson WA, Vijayan MM. Zygotic Exposure to Venlafaxine Disrupts the Circadian Locomotor Activity Behaviour in Zebrafish Larvae. J Pineal Res 2024; 76:e12984. [PMID: 38874070 DOI: 10.1111/jpi.12984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/15/2024]
Abstract
The antidepressant venlafaxine, a selective serotonin and norepinephrine reuptake inhibitor, is commonly prescribed to treat major depressive disorder and is found at high concentrations in the aquatic environment. Concerns have been raised related to the health of aquatic organisms in response to this nontargeted pharmaceutical exposure. For instance, we previously demonstrated that exposure to venlafaxine perturbs neurodevelopment, leading to behavioural alterations in zebrafish (Danio rerio). We also observed disruption in serotonin expression in the pineal and raphe, regions critical in regulating circadian rhythms, leading us to hypothesize that zygotic exposure to venlafaxine disrupts the circadian locomotor rhythm in larval zebrafish. To test this, we microinjected zebrafish embryos with venlafaxine (1 or 10 ng) and recorded the locomotor activity in 5-day-old larvae over a 24-h period. Venlafaxine deposition reduced larval locomotor activity during the light phase, but not during the dark phase of the diurnal cycle. The melatonin levels were higher in the dark compared to during the light photoperiod and this was not affected by embryonic venlafaxine deposition. Venlafaxine exposure also did not affect the transcript abundance of clock genes, including clock1a, bmal2, cry1a and per2, which showed a clear day/night rhythmicity. A notable finding was that exposure to luzindole, a melatonin receptor antagonist, decreased the locomotor activity in the control group in light, whereas the activity was higher in larvae raised from the venlafaxine-deposited embryos. Overall, zygotic exposure to venlafaxine disrupts the locomotor activity of larval zebrafish fish during the day, demonstrating the capacity of antidepressants to disrupt the circadian rhythms in behaviour. Our results suggest that disruption in melatonin signalling may be playing a role in the venlafaxine impact on circadian behaviour, but further investigation is required to elucidate the possible mechanisms in larval zebrafish.
Collapse
Affiliation(s)
- W Andrew Thompson
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | | |
Collapse
|
4
|
Granados-Fuentes D, Lambert P, Simon T, Mennerick S, Herzog ED. GABA A receptor subunit composition regulates circadian rhythms in rest-wake and synchrony among cells in the suprachiasmatic nucleus. Proc Natl Acad Sci U S A 2024; 121:e2400339121. [PMID: 39047036 PMCID: PMC11295074 DOI: 10.1073/pnas.2400339121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
The mammalian circadian clock located in the suprachiasmatic nucleus (SCN) produces robust daily rhythms including rest-wake. SCN neurons synthesize and respond to γ-aminobutyric acid (GABA), but its role remains unresolved. We tested the hypothesis that γ2- and δ-subunits of the GABAA receptor in the SCN differ in their regulation of synchrony among circadian cells. We used two approaches: 1) shRNA to knock-down (KD) the expression of either γ2 or δ subunits in the SCN or 2) knock-in mice harboring a point mutation in the M2 domains of the endogenous GABAA γ2 or δ subunits. KD of either γ2 or δ subunits in the SCN increased daytime running and reduced nocturnal running by reducing their circadian amplitude by a third. Similarly, δ subunit knock-in mice showed decreased circadian amplitude, increased duration of daily activity, and decreased total daily activity. Reduction, or mutation of either γ2 or δ subunits halved the synchrony among, and amplitude of, circadian SCN cells as measured by firing rate or expression of the PERIOD2 protein, in vitro. Surprisingly, overexpression of the γ2 subunit rescued these phenotypes following KD or mutation of the δ subunit, and overexpression of the δ subunit rescued deficiencies due to γ2 subunit KD or mutation. We conclude that γ2 and δ GABAA receptor subunits play similar roles in maintaining circadian synchrony in the SCN and amplitude of daily rest-wake rhythms, but that modulation of their relative densities can change the duration and amplitude of daily activities.
Collapse
Affiliation(s)
| | - Peter Lambert
- Department of Psychiatry, Washington University in St. Louis, MO63130-4899
| | - Tatiana Simon
- Department of Biology, Washington University in St. Louis, MO63130-4899
| | - Steven Mennerick
- Department of Psychiatry, Washington University in St. Louis, MO63130-4899
| | - Erik D. Herzog
- Department of Biology, Washington University in St. Louis, MO63130-4899
| |
Collapse
|
5
|
Rodan AR. Circadian Rhythm Regulation by Pacemaker Neuron Chloride Oscillation in Flies. Physiology (Bethesda) 2024; 39:0. [PMID: 38411570 PMCID: PMC11368518 DOI: 10.1152/physiol.00006.2024] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/28/2024] Open
Abstract
Circadian rhythms in physiology and behavior sync organisms to external environmental cycles. Here, circadian oscillation in intracellular chloride in central pacemaker neurons of the fly, Drosophila melanogaster, is reviewed. Intracellular chloride links SLC12 cation-coupled chloride transporter function with kinase signaling and the regulation of inwardly rectifying potassium channels.
Collapse
Affiliation(s)
- Aylin R Rodan
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah, United States
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, United States
- Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, United States
| |
Collapse
|
6
|
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
|
7
|
Naveed M, Chao OY, Hill JW, Yang YM, Huston JP, Cao R. Circadian neurogenetics and its implications in neurophysiology, behavior, and chronomedicine. Neurosci Biobehav Rev 2024; 157:105523. [PMID: 38142983 PMCID: PMC10872425 DOI: 10.1016/j.neubiorev.2023.105523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 12/26/2023]
Abstract
The circadian rhythm affects multiple physiological processes, and disruption of the circadian system can be involved in a range of disease-related pathways. The genetic underpinnings of the circadian rhythm have been well-studied in model organisms. Significant progress has been made in understanding how clock genes affect the physiological functions of the nervous system. In addition, circadian timing is becoming a key factor in improving drug efficacy and reducing drug toxicity. The circadian biology of the target cell determines how the organ responds to the drug at a specific time of day, thus regulating pharmacodynamics. The current review brings together recent advances that have begun to unravel the molecular mechanisms of how the circadian clock affects neurophysiological and behavioral processes associated with human brain diseases. We start with a brief description of how the ubiquitous circadian rhythms are regulated at the genetic, cellular, and neural circuit levels, based on knowledge derived from extensive research on model organisms. We then summarize the latest findings from genetic studies of human brain disorders, focusing on the role of human clock gene variants in these diseases. Lastly, we discuss the impact of common dietary factors and medications on human circadian rhythms and advocate for a broader application of the concept of chronomedicine.
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Owen Y Chao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Joseph P Huston
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Ruifeng Cao
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA; Department of Neurology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
8
|
Bussi IL, Neitz AF, Sanchez REA, Casiraghi LP, Moldavan M, Kunda D, Allen CN, Evans JA, de la Iglesia HO. Expression of the vesicular GABA transporter within neuromedin S + neurons sustains behavioral circadian rhythms. Proc Natl Acad Sci U S A 2023; 120:e2314857120. [PMID: 38019855 PMCID: PMC10710084 DOI: 10.1073/pnas.2314857120] [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: 08/30/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus is the site of a central circadian clock that orchestrates overt rhythms of physiology and behavior. Circadian timekeeping requires intercellular communication among SCN neurons, and multiple signaling pathways contribute to SCN network coupling. Gamma-aminobutyric acid (GABA) is produced by virtually all SCN neurons, and previous work demonstrates that this transmitter regulates coupling in the adult SCN but is not essential for the nucleus to sustain overt circadian rhythms. Here, we show that the deletion of the gene that codes for the GABA vesicular transporter Vgat from neuromedin-S (NMS)+ neurons-a subset of neurons critical for SCN function-causes arrhythmia of locomotor activity and sleep. Further, NMS-Vgat deletion impairs intrinsic clock gene rhythms in SCN explants cultured ex vivo. Although vasoactive intestinal polypeptide (VIP) is critical for SCN function, Vgat deletion from VIP-expressing neurons did not lead to circadian arrhythmia in locomotor activity rhythms. Likewise, adult SCN-specific deletion of Vgat led to mild impairment of behavioral rhythms. Our results suggest that while the removal of GABA release from the adult SCN does not affect the pacemaker's ability to sustain overt circadian rhythms, its removal from a critical subset of neurons within the SCN throughout development removes the nucleus ability to sustain circadian rhythms. Our findings support a model in which SCN GABA release is critical for the developmental establishment of intercellular network properties that define the SCN as a central pacemaker.
Collapse
Affiliation(s)
- Ivana L. Bussi
- Department of Biology, University of Washington, Seattle, WA98195-1800
| | - Alexandra F. Neitz
- Department of Biology, University of Washington, Seattle, WA98195-1800
- Molecular and Cellular Biology in Seattle, University of Washington and Fred Hutch, Seattle, WA98195-7275
| | - Raymond E. A. Sanchez
- Department of Biology, University of Washington, Seattle, WA98195-1800
- Graduate Program in Neuroscience, University of Washington, Seattle, WA98195
| | | | - Michael Moldavan
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR97239
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR97239
| | - Divya Kunda
- Department of Biology, University of Washington, Seattle, WA98195-1800
| | - Charles N. Allen
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR97239
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR97239
| | - Jennifer A. Evans
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI53233
| | - Horacio O. de la Iglesia
- Department of Biology, University of Washington, Seattle, WA98195-1800
- Molecular and Cellular Biology in Seattle, University of Washington and Fred Hutch, Seattle, WA98195-7275
- Graduate Program in Neuroscience, University of Washington, Seattle, WA98195
| |
Collapse
|
9
|
Qian L, Gu Y, Zhai Q, Xue Z, Liu Y, Li S, Zeng Y, Sun R, Zhang Q, Cai X, Ge W, Dong Z, Gao H, Zhou Y, Zhu Y, Xu Y, Guo T. Multitissue Circadian Proteome Atlas of WT and Per1 -/-/Per2 -/- Mice. Mol Cell Proteomics 2023; 22:100675. [PMID: 37940002 PMCID: PMC10750102 DOI: 10.1016/j.mcpro.2023.100675] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/22/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023] Open
Abstract
The molecular basis of circadian rhythm, driven by core clock genes such as Per1/2, has been investigated on the transcriptome level, but not comprehensively on the proteome level. Here we quantified over 11,000 proteins expressed in eight types of tissues over 46 h with an interval of 2 h, using WT and Per1/Per2 double knockout mouse models. The multitissue circadian proteome landscape of WT mice shows tissue-specific patterns and reflects circadian anticipatory phenomena, which are less obvious on the transcript level. In most peripheral tissues of double knockout mice, reduced protein cyclers are identified when compared with those in WT mice. In addition, PER1/2 contributes to controlling the anticipation of the circadian rhythm, modulating tissue-specific cyclers as well as key pathways including nucleotide excision repair. Severe intertissue temporal dissonance of circadian proteome has been observed in the absence of Per1 and Per2. The γ-aminobutyric acid might modulate some of these temporally correlated cyclers in WT mice. Our study deepens our understanding of rhythmic proteins across multiple tissues and provides valuable insights into chronochemotherapy. The data are accessible at https://prot-rhythm.prottalks.com/.
Collapse
Affiliation(s)
- Liujia Qian
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Yue Gu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou, Jiangsu Province, China
| | - Qiaocheng Zhai
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou, Jiangsu Province, China
| | - Zhangzhi Xue
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Youqi Liu
- Westlake Omics (Hangzhou) Biotechnology Co, Ltd, Hangzhou, Zhejiang Province, China
| | - Sainan Li
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Yizhun Zeng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou, Jiangsu Province, China
| | - Rui Sun
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Qiushi Zhang
- Westlake Omics (Hangzhou) Biotechnology Co, Ltd, Hangzhou, Zhejiang Province, China
| | - Xue Cai
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Weigang Ge
- Westlake Omics (Hangzhou) Biotechnology Co, Ltd, Hangzhou, Zhejiang Province, China
| | - Zhen Dong
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Huanhuan Gao
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Yan Zhou
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Yi Zhu
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China.
| | - Ying Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou, Jiangsu Province, China.
| | - Tiannan Guo
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|
10
|
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: 23] [Impact Index Per Article: 11.5] [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
|
11
|
Giantomasi L, Ribeiro JF, Barca-Mayo O, Malerba M, Miele E, De Pietri Tonelli D, Berdondini L. Astrocytes actively support long-range molecular clock synchronization of segregated neuronal populations. Sci Rep 2023; 13:4815. [PMID: 36964220 PMCID: PMC10038999 DOI: 10.1038/s41598-023-31966-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 03/20/2023] [Indexed: 03/26/2023] Open
Abstract
In mammals, the suprachiasmatic nucleus of the hypothalamus is the master circadian pacemaker that synchronizes the clocks in the central nervous system and periphery, thus orchestrating rhythms throughout the body. However, little is known about how so many cellular clocks within and across brain circuits can be effectively synchronized. In this work, we investigated the implication of two possible pathways: (i) astrocytes-mediated synchronization and (ii) neuronal paracrine factors-mediated synchronization. By taking advantage of a lab-on-a-chip microfluidic device developed in our laboratory, here we report that both pathways are involved. We found the paracrine factors-mediated synchronization of molecular clocks is diffusion-limited and, in our device, effective only in case of a short distance between neuronal populations. Interestingly, interconnecting astrocytes define an active signaling channel that can synchronize molecular clocks of neuronal populations also at longer distances. At mechanism level, we found that astrocytes-mediated synchronization involves both GABA and glutamate, while neuronal paracrine factors-mediated synchronization occurs through GABA signaling. These findings identify a previously unknown role of astrocytes as active cells that might distribute long-range signals to synchronize the brain clocks, thus further strengthening the importance of reciprocal interactions between glial and neuronal cells in the context of circadian circuitry.
Collapse
Affiliation(s)
- Lidia Giantomasi
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano Di Tecnologia (IIT), 16163, Genova, Italy
| | - João F Ribeiro
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano Di Tecnologia (IIT), 16163, Genova, Italy
| | - Olga Barca-Mayo
- Neurobiology of miRNA, Fondazione Istituto Italiano Di Tecnologia (IIT), 16163, Genova, Italy
- Circadian and Glial Biology Lab, Physiology Department, Molecular Medicine, and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Mario Malerba
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano Di Tecnologia (IIT), 16163, Genova, Italy
- Centre de Nanosciences et de Nanotechnologies (C2N), CNRS UMR 9001, Université Paris-Saclay, 91120, Palaiseau, France
| | - Ermanno Miele
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano Di Tecnologia (IIT), 16163, Genova, Italy
- NanoPhotonics Centre, Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | | | - Luca Berdondini
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano Di Tecnologia (IIT), 16163, Genova, Italy.
| |
Collapse
|
12
|
Schurhoff N, Toborek M. Circadian rhythms in the blood-brain barrier: impact on neurological disorders and stress responses. Mol Brain 2023; 16:5. [PMID: 36635730 PMCID: PMC9835375 DOI: 10.1186/s13041-023-00997-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023] Open
Abstract
Circadian disruption has become more prevalent in society due to the increase in shift work, sleep disruption, blue light exposure, and travel via different time zones. The circadian rhythm is a timed transcription-translation feedback loop with positive regulators, BMAL1 and CLOCK, that interact with negative regulators, CRY and PER, to regulate both the central and peripheral clocks. This review highlights the functions of the circadian rhythm, specifically in the blood-brain barrier (BBB), during both healthy and pathological states. The BBB is a highly selective dynamic interface composed of CNS endothelial cells, astrocytes, pericytes, neurons, and microglia that form the neurovascular unit (NVU). Circadian rhythms modulate BBB integrity through regulating oscillations of tight junction proteins, assisting in functions of the NVU, and modulating transporter functions. Circadian disruptions within the BBB have been observed in stress responses and several neurological disorders, including brain metastasis, epilepsy, Alzheimer's disease, and Parkinson's disease. Further understanding of these interactions may facilitate the development of improved treatment options and preventative measures.
Collapse
Affiliation(s)
- Nicolette Schurhoff
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Suite 528, 1011 NW 15th Street, Miami, FL, 33155, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Suite 528, 1011 NW 15th Street, Miami, FL, 33155, USA.
- Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, 40-065, Katowice, Poland.
| |
Collapse
|
13
|
Cruz-Sanabria F, Carmassi C, Bruno S, Bazzani A, Carli M, Scarselli M, Faraguna U. Melatonin as a Chronobiotic with Sleep-promoting Properties. Curr Neuropharmacol 2023; 21:951-987. [PMID: 35176989 PMCID: PMC10227911 DOI: 10.2174/1570159x20666220217152617] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/26/2022] [Accepted: 02/10/2022] [Indexed: 11/22/2022] Open
Abstract
The use of exogenous melatonin (exo-MEL) as a sleep-promoting drug has been under extensive debate due to the lack of consistency of its described effects. In this study, we conduct a systematic and comprehensive review of the literature on the chronobiotic, sleep-inducing, and overall sleep-promoting properties of exo-MEL. To this aim, we first describe the possible pharmacological mechanisms involved in the sleep-promoting properties and then report the corresponding effects of exo-MEL administration on clinical outcomes in: a) healthy subjects, b) circadian rhythm sleep disorders, c) primary insomnia. Timing of administration and doses of exo-MEL received particular attention in this work. The exo-MEL pharmacological effects are hereby interpreted in view of changes in the physiological properties and rhythmicity of endogenous melatonin. Finally, we discuss some translational implications for the personalized use of exo-MEL in the clinical practice.
Collapse
Affiliation(s)
- Francy Cruz-Sanabria
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa - Italy
| | - Claudia Carmassi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa - Italy
| | - Simone Bruno
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa - Italy
| | - Andrea Bazzani
- Institute of Management, Scuola Superiore Sant’Anna, Pisa – Italy
| | - Marco Carli
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa - Italy
| | - Marco Scarselli
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa - Italy
| | - Ugo Faraguna
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa - Italy
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, Pisa, Pisa, Italy
| |
Collapse
|
14
|
Lu Q, Kim JY. Mammalian circadian networks mediated by the suprachiasmatic nucleus. FEBS J 2022; 289:6589-6604. [PMID: 34657394 DOI: 10.1111/febs.16233] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/09/2021] [Accepted: 10/15/2021] [Indexed: 12/28/2022]
Abstract
The brain has a complex structure composed of hundreds of regions, forming networks to cooperate body functions. Therefore, understanding how various brain regions communicate with each other and with peripheral organs is important to understand human physiology. The suprachiasmatic nucleus (SCN) in the brain is the circadian pacemaker. The SCN receives photic information from the environment and conveys this to other parts of the brain and body to synchronize all circadian clocks. The circadian clock is an endogenous oscillator that generates daily rhythms in metabolism and physiology in almost all cells via a conserved transcriptional-translational negative feedback loop. So, the information flow from the environment to the SCN to other tissues synchronizes locally distributed circadian clocks to maintain homeostasis. Thus, understanding the circadian networks and how they adjust to environmental changes will better understand human physiology. This review will focus on circadian networks mediated by the SCN to understand how the environment, brain, and peripheral tissues form networks for cooperation.
Collapse
Affiliation(s)
- Qingqing Lu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Jin Young Kim
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Tung Foundation Biomedical Sciences Centre, Hong Kong, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| |
Collapse
|
15
|
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
|
16
|
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: 4.0] [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
|
17
|
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
|
18
|
Chaturvedi R, Stork T, Yuan C, Freeman MR, Emery P. Astrocytic GABA transporter controls sleep by modulating GABAergic signaling in Drosophila circadian neurons. Curr Biol 2022; 32:1895-1908.e5. [PMID: 35303417 PMCID: PMC9090989 DOI: 10.1016/j.cub.2022.02.066] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 01/11/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022]
Abstract
A precise balance between sleep and wakefulness is essential to sustain a good quality of life and optimal brain function. GABA is known to play a key and conserved role in sleep control, and GABAergic tone should, therefore, be tightly controlled in sleep circuits. Here, we examined the role of the astrocytic GABA transporter (GAT) in sleep regulation using Drosophila melanogaster. We found that a hypomorphic gat mutation (gat33-1) increased sleep amount, decreased sleep latency, and increased sleep consolidation at night. Interestingly, sleep defects were suppressed when gat33-1 was combined with a mutation disrupting wide-awake (wake), a gene that regulates the cell-surface levels of the GABAA receptor resistance to dieldrin (RDL) in the wake-promoting large ventral lateral neurons (l-LNvs). Moreover, RNAi knockdown of rdl and its modulators dnlg4 and wake in these circadian neurons also suppressed gat33-1 sleep phenotypes. Brain immunohistochemistry showed that GAT-expressing astrocytes were located near RDL-positive l-LNv cell bodies and dendritic processes. We concluded that astrocytic GAT decreases GABAergic tone and RDL activation in arousal-promoting LNvs, thus determining proper sleep amount and quality in Drosophila.
Collapse
Affiliation(s)
- Ratna Chaturvedi
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Tobias Stork
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Chunyan Yuan
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Marc R Freeman
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Patrick Emery
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
19
|
Jiménez A, Lu Y, Jambhekar A, Lahav G. Principles, mechanisms and functions of entrainment in biological oscillators. Interface Focus 2022; 12:20210088. [PMID: 35450280 PMCID: PMC9010850 DOI: 10.1098/rsfs.2021.0088] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Entrainment is a phenomenon in which two oscillators interact with each other, typically through physical or chemical means, to synchronize their oscillations. This phenomenon occurs in biology to coordinate processes from the molecular to organismal scale. Biological oscillators can be entrained within a single cell, between cells or to an external input. Using six illustrative examples of entrainable biological oscillators, we discuss the distinctions between entrainment and synchrony and explore features that contribute to a system's propensity to entrain. Entrainment can either enhance or reduce the heterogeneity of oscillations within a cell population, and we provide examples and mechanisms of each case. Finally, we discuss the known functions of entrainment and discuss potential functions from an evolutionary perspective.
Collapse
Affiliation(s)
- Alba Jiménez
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
| | - Ying Lu
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
| | - Ashwini Jambhekar
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
- Ludwig Center at Harvard, Boston, MA 02115, USA
| | - Galit Lahav
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
- Ludwig Center at Harvard, Boston, MA 02115, USA
| |
Collapse
|
20
|
Lee R, McGee A, Fernandez FX. Systematic review of drugs that modify the circadian system's phase-shifting responses to light exposure. Neuropsychopharmacology 2022; 47:866-879. [PMID: 34961774 PMCID: PMC8882192 DOI: 10.1038/s41386-021-01251-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/08/2021] [Accepted: 11/30/2021] [Indexed: 11/09/2022]
Abstract
We searched PubMed for primary research quantifying drug modification of light-induced circadian phase-shifting in rodents. This search, conducted for work published between 1960 and 2018, yielded a total of 146 papers reporting results from 901 studies. Relevant articles were those with any extractable data on phase resetting in wildtype (non-trait selected) rodents administered a drug, alongside a vehicle/control group, near or at the time of exposure. Most circadian pharmacology experiments were done using drugs thought to act directly on either the brain's central pacemaker, the suprachiasmatic nucleus (SCN), the SCN's primary relay, the retinohypothalamic tract, secondary pathways originating from the medial/dorsal raphe nuclei and intergeniculate leaflet, or the brain's sleep-arousal centers. While the neurotransmitter systems underlying these circuits were of particular interest, including those involving glutamate, gamma-aminobutyric acid, serotonin, and acetylcholine, other signaling modalities have also been assessed, including agonists and antagonists of receptors linked to dopamine, histamine, endocannabinoids, adenosine, opioids, and second-messenger pathways downstream of glutamate receptor activation. In an effort to identify drugs that unduly influence circadian responses to light, we quantified the net effects of each drug class by ratioing the size of the phase-shift observed after administration to that observed with vehicle in a given experiment. This allowed us to organize data across the literature, compare the relative efficacy of one mechanism versus another, and clarify which drugs might best suppress or potentiate phase resetting. Aggregation of the available data in this manner suggested that several candidates might be clinically relevant as auxiliary treatments to suppress ectopic light responses during shiftwork or amplify the circadian effects of timed bright light therapy. Future empirical research will be necessary to validate these possibilities.
Collapse
Affiliation(s)
- Robert Lee
- Department of Psychology, University of Arizona, Tucson, AZ, USA
| | - Austin McGee
- Department of Psychology, University of Arizona, Tucson, AZ, USA
| | - Fabian-Xosé Fernandez
- Department of Psychology, University of Arizona, Tucson, AZ, USA.
- Department of Neurology, University of Arizona, Tucson, AZ, USA.
- BIO5 and McKnight Brain Research Institutes, Tucson, AZ, USA.
| |
Collapse
|
21
|
Klett NJ, Cravetchi O, Allen CN. Long-Term Imaging Reveals a Circadian Rhythm of Intracellular Chloride in Neurons of the Suprachiasmatic Nucleus. J Biol Rhythms 2022; 37:110-123. [PMID: 34994231 PMCID: PMC9203244 DOI: 10.1177/07487304211059770] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Both inhibitory and excitatory GABA transmission exist in the mature suprachiasmatic nucleus (SCN), the master pacemaker of circadian physiology. Whether GABA is inhibitory or excitatory depends on the intracellular chloride concentration ([Cl-]i). Here, using the genetically encoded ratiometric probe Cl-Sensor, we investigated [Cl-]i in AVP and VIP-expressing SCN neurons for several days in culture. The chloride ratio (RCl) demonstrated circadian rhythmicity in AVP + neurons and VIP + neurons, but was not detected in GFAP + astrocytes. RCl peaked between ZT 7 and ZT 8 in both AVP + and VIP + neurons. RCl rhythmicity was not dependent on the activity of several transmembrane chloride carriers, action potential generation, or the L-type voltage-gated calcium channels, but was sensitive to GABA antagonists. We conclude that [Cl-]i is under circadian regulation in both AVP + and VIP + neurons.
Collapse
Affiliation(s)
- Nathan J. Klett
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR 97239
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239
| | - Olga Cravetchi
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239
| | - Charles N. Allen
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
| |
Collapse
|
22
|
Li Y, Androulakis IP. Light-induced synchronization of the SCN coupled oscillators and implications for entraining the HPA axis. Front Endocrinol (Lausanne) 2022; 13:960351. [PMID: 36387856 PMCID: PMC9648564 DOI: 10.3389/fendo.2022.960351] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
The suprachiasmatic nucleus (SCN) synchronizes the physiological rhythms to the external light-dark cycle and tunes the dynamics of circadian rhythms to photoperiod fluctuations. Changes in the neuronal network topologies are suggested to cause adaptation of the SCN in different photoperiods, resulting in the broader phase distribution of neuron activities in long photoperiods (LP) compared to short photoperiods (SP). Regulated by the SCN output, the level of glucocorticoids is elevated in short photoperiod, which is associated with peak disease incidence. The underlying coupling mechanisms of the SCN and the interplay between the SCN and the HPA axis have yet to be fully elucidated. In this work, we propose a mathematical model including a multiple-cellular SCN compartment and the HPA axis to investigate the properties of the circadian timing system under photoperiod changes. Our model predicts that the probability-dependent network is more energy-efficient than the distance-dependent network. Coupling the SCN network by intra-subpopulation and inter-subpopulation forces, we identified the negative correlation between robustness and plasticity of the oscillatory network. The HPA rhythms were predicted to be strongly entrained to the SCN rhythms with a pro-inflammatory high-amplitude glucocorticoid profile under SP. The fast temporal topology switch of the SCN network was predicted to enhance synchronization when the synchronization is not complete. These synchronization and circadian dynamics alterations might govern the seasonal variation of disease incidence and its symptom severity.
Collapse
Affiliation(s)
- Yannuo Li
- Chemical & Biochemical Engineering Department, Rutgers University, Piscataway, NJ, United States
| | - Ioannis P. Androulakis
- Chemical & Biochemical Engineering Department, Rutgers University, Piscataway, NJ, United States
- Biomedical Engineering Department, Rutgers University, Piscataway, NJ, United States
- *Correspondence: Ioannis P. Androulakis,
| |
Collapse
|
23
|
Gpr19 is a circadian clock-controlled orphan GPCR with a role in modulating free-running period and light resetting capacity of the circadian clock. Sci Rep 2021; 11:22406. [PMID: 34789778 PMCID: PMC8599615 DOI: 10.1038/s41598-021-01764-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/03/2021] [Indexed: 01/26/2023] Open
Abstract
Gpr19 encodes an evolutionarily conserved orphan G-protein-coupled receptor (GPCR) with currently no established physiological role in vivo. We characterized Gpr19 expression in the suprachiasmatic nucleus (SCN), the locus of the master circadian clock in the brain, and determined its role in the context of the circadian rhythm regulation. We found that Gpr19 is mainly expressed in the dorsal part of the SCN, with its expression fluctuating in a circadian fashion. A conserved cAMP-responsive element in the Gpr19 promoter was able to produce circadian transcription in the SCN. Gpr19−/− mice exhibited a prolonged circadian period and a delayed initiation of daily locomotor activity. Gpr19 deficiency caused the downregulation of several genes that normally peak during the night, including Bmal1 and Gpr176. In response to light exposure at night, Gpr19−/− mice had a reduced capacity for light-induced phase-delays, but not for phase-advances. This defect was accompanied by reduced response of c-Fos expression in the dorsal region of the SCN, while apparently normal in the ventral area of the SCN, in Gpr19−/− mice. Thus, our data demonstrate that Gpr19 is an SCN-enriched orphan GPCR with a distinct role in circadian regulation and may provide a potential target option for modulating the circadian clock.
Collapse
|
24
|
Astrocyte Gliotransmission in the Regulation of Systemic Metabolism. Metabolites 2021; 11:metabo11110732. [PMID: 34822390 PMCID: PMC8623475 DOI: 10.3390/metabo11110732] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 12/28/2022] Open
Abstract
Normal brain function highly relies on the appropriate functioning of astrocytes. These glial cells are strategically situated between blood vessels and neurons, provide significant substrate support to neuronal demand, and are sensitive to neuronal activity and energy-related molecules. Astrocytes respond to many metabolic conditions and regulate a wide array of physiological processes, including cerebral vascular remodeling, glucose sensing, feeding, and circadian rhythms for the control of systemic metabolism and behavior-related responses. This regulation ultimately elicits counterregulatory mechanisms in order to couple whole-body energy availability with brain function. Therefore, understanding the role of astrocyte crosstalk with neighboring cells via the release of molecules, e.g., gliotransmitters, into the parenchyma in response to metabolic and neuronal cues is of fundamental relevance to elucidate the distinct roles of these glial cells in the neuroendocrine control of metabolism. Here, we review the mechanisms underlying astrocyte-released gliotransmitters that have been reported to be crucial for maintaining homeostatic regulation of systemic metabolism.
Collapse
|
25
|
Osuna-Lopez F, Reyes-Mendez ME, Herrera-Zamora JM, Gongora-Alfaro JL, Moreno-Galindo EG, Alamilla J. GABA Neurotransmission of the Suprachiasmatic Nucleus Is Modified During Rat Postnatal Development. J Biol Rhythms 2021; 36:567-574. [PMID: 34643150 DOI: 10.1177/07487304211048052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus is the brain structure that controls circadian rhythms in mammals. The SCN is formed by two neuroanatomical regions: the ventral and dorsal. Gamma-aminobutyric acid (GABA) neurotransmission is important for the regulation of circadian rhythms. Excitatory GABA effects have been described in both SCN regions displaying a circadian variation. Moreover, the GABAergic system transfers photic information from the ventral to the dorsal SCN. However, there is almost no knowledge about GABA neurotransmission during the prenatal or postnatal development of the SCN. Here, we used whole-cell patch-clamp recordings to study spontaneous inhibitory postsynaptic currents (IPSCs) in the two SCN regions, at two zeitgeber times (day or night), and at four postnatal (P) ages: P3-5, P7-9, P12-15, and P20-25. The results herein show that the three analyzed parameters of the IPSCs, frequency, amplitude, and decay time, were significantly affected by the postnatal age: mostly, the IPSC frequency increased with age, principally in the ventral SCN in both day and night recordings; similarly, the amplitude of IPSCs augmented with age, especially at night, whereas the IPSC decay time was reduced (it was faster) with postnatal age, mainly during the day. Our findings first reveal that parameters of GABA neurotransmission are modified by postnatal development, implying that synaptic adjustments are required for an appropriate maturation of the GABAergic system in the SCN.
Collapse
Affiliation(s)
- Fernando Osuna-Lopez
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Mexico
| | - Miriam E Reyes-Mendez
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Mexico
| | | | - Jose Luis Gongora-Alfaro
- Departamento de Neurociencias, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi," Universidad Autónoma de Yucatán, Mérida, Mexico
| | - Eloy G Moreno-Galindo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Mexico
| | - Javier Alamilla
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Mexico.,Consejo Nacional de Ciencia y Tecnología, Universidad de Colima, Colima, Mexico
| |
Collapse
|
26
|
Massah A, Neupert S, Brodesser S, Homberg U, Stengl M. Distribution and daily oscillation of GABA in the circadian system of the cockroach Rhyparobia maderae. J Comp Neurol 2021; 530:770-791. [PMID: 34586642 DOI: 10.1002/cne.25244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 12/28/2022]
Abstract
Gamma-aminobutyric acid (GABA) is the prevalent inhibitory neurotransmitter in nervous systems promoting sleep in both mammals and insects. In the Madeira cockroach, sleep-wake cycles are controlled by a circadian clock network in the brain's optic lobes, centered in the accessory medulla (AME) with its innervating pigment-dispersing factor (PDF) expressing clock neurons at the anterior-ventral rim of the medulla. GABA is present in cell clusters that innervate different circuits of the cockroach's AME clock, without colocalizing in PDF clock neurons. Physiological, immunohistochemical, and behavioral assays provided evidence for a role of GABA in light entrainment, possibly via the distal tract that connects the AME's glomeruli to the medulla. Furthermore, GABA was implemented in clock outputs to multiple effector systems in optic lobe and midbrain. Here, GABAergic brain circuits were analyzed further, focusing on the circadian system in search for sleep/wake controlling brain circuits. All GABA-immunoreactive neurons of the cockroach brain were also stained with an antiserum against the GABA-synthesizing enzyme glutamic acid decarboxylase. We found strong overlap of the distribution of GABA-immunoreactive networks with PDF clock networks in optic lobes and midbrain. Neurons in five of the six soma groups that innervate the clock exhibited GABA immunoreactivity. The intensity of GABA immunoreactivity in the distal tract showed daily fluctuations with maximum staining intensity in the middle of the day and weakest staining at the end of the day. Quantification via enzyme-linked immunosorbent assay and quantitative liquid chromatography coupled to electrospray ionization tandem mass spectrometry, likewise, showed higher GABA levels in the optic lobe during the inactivity phase of the cockroach during the day and lower levels during its activity phase at dusk. Our data further support the hypothesis that light- and PDF-dependently the circadian clock network of the cockroach controls GABA levels and thereby promotes sleep during the day.
Collapse
Affiliation(s)
- Azar Massah
- Institute of Biology, Animal Physiology, University of Kassel, Kassel, Germany
| | - Susanne Neupert
- Institute of Biology, Animal Physiology, University of Kassel, Kassel, Germany
| | - Susanne Brodesser
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Uwe Homberg
- Department of Biology, Animal Physiology, Philipps-Universität Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| | - Monika Stengl
- Institute of Biology, Animal Physiology, University of Kassel, Kassel, Germany
| |
Collapse
|
27
|
Sueviriyapan N, Granados-Fuentes D, Simon T, Herzog ED, Henson MA. Modelling the functional roles of synaptic and extra-synaptic γ-aminobutyric acid receptor dynamics in circadian timekeeping. J R Soc Interface 2021; 18:20210454. [PMID: 34520693 PMCID: PMC8440032 DOI: 10.1098/rsif.2021.0454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/23/2021] [Indexed: 11/12/2022] Open
Abstract
In the suprachiasmatic nucleus (SCN), γ-aminobutyric acid (GABA) is a primary neurotransmitter. GABA can signal through two types of GABAA receptor subunits, often referred to as synaptic GABAA (gamma subunit) and extra-synaptic GABAA (delta subunit). To test the functional roles of these distinct GABAA in regulating circadian rhythms, we developed a multicellular SCN model where we could separately compare the effects of manipulating GABA neurotransmitter or receptor dynamics. Our model predicted that blocking GABA signalling modestly increased synchrony among circadian cells, consistent with published SCN pharmacology. Conversely, the model predicted that lowering GABAA receptor density reduced firing rate, circadian cell fraction, amplitude and synchrony among individual neurons. When we tested these predictions, we found that the knockdown of delta GABAA reduced the amplitude and synchrony of clock gene expression among cells in SCN explants. The model further predicted that increasing gamma GABAA densities could enhance synchrony, as opposed to increasing delta GABAA densities. Overall, our model reveals how blocking GABAA receptors can modestly increase synchrony, while increasing the relative density of gamma over delta subunits can dramatically increase synchrony. We hypothesize that increased gamma GABAA density in the winter could underlie the tighter phase relationships among SCN cells.
Collapse
Affiliation(s)
- Natthapong Sueviriyapan
- Department of Chemical Engineering and the Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| | | | - Tatiana Simon
- Department of Biology, Washington University in St Louis, Saint Louis, MO, USA
| | - Erik D. Herzog
- Department of Biology, Washington University in St Louis, Saint Louis, MO, USA
| | - Michael A. Henson
- Department of Chemical Engineering and the Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
28
|
Salehinejad MA, Wischnewski M, Ghanavati E, Mosayebi-Samani M, Kuo MF, Nitsche MA. Cognitive functions and underlying parameters of human brain physiology are associated with chronotype. Nat Commun 2021; 12:4672. [PMID: 34344864 PMCID: PMC8333420 DOI: 10.1038/s41467-021-24885-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/08/2021] [Indexed: 01/03/2023] Open
Abstract
Circadian rhythms have natural relative variations among humans known as chronotype. Chronotype or being a morning or evening person, has a specific physiological, behavioural, and also genetic manifestation. Whether and how chronotype modulates human brain physiology and cognition is, however, not well understood. Here we examine how cortical excitability, neuroplasticity, and cognition are associated with chronotype in early and late chronotype individuals. We monitor motor cortical excitability, brain stimulation-induced neuroplasticity, and examine motor learning and cognitive functions at circadian-preferred and non-preferred times of day in 32 individuals. Motor learning and cognitive performance (working memory, and attention) along with their electrophysiological components are significantly enhanced at the circadian-preferred, compared to the non-preferred time. This outperformance is associated with enhanced cortical excitability (prominent cortical facilitation, diminished cortical inhibition), and long-term potentiation/depression-like plasticity. Our data show convergent findings of how chronotype can modulate human brain functions from basic physiological mechanisms to behaviour and higher-order cognition.
Collapse
Affiliation(s)
- Mohammad Ali Salehinejad
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
- International Graduate School of Neuroscience, Ruhr-University Bochum, Bochum, Germany
| | - Miles Wischnewski
- Donders Institute for Brain, Cognition, and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Elham Ghanavati
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
- Department of Psychology, Ruhr-University Bochum, Bochum, Germany
| | - Mohsen Mosayebi-Samani
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Min-Fang Kuo
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Michael A Nitsche
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany.
- Department of Neurology, University Medical Hospital Bergmannsheil, Bochum, Germany.
| |
Collapse
|
29
|
Hughes ATL, Samuels RE, Baño-Otálora B, Belle MDC, Wegner S, Guilding C, Northeast RC, Loudon ASI, Gigg J, Piggins HD. Timed daily exercise remodels circadian rhythms in mice. Commun Biol 2021; 4:761. [PMID: 34145388 PMCID: PMC8213798 DOI: 10.1038/s42003-021-02239-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/18/2021] [Indexed: 01/26/2023] Open
Abstract
Regular exercise is important for physical and mental health. An underexplored and intriguing property of exercise is its actions on the body’s 24 h or circadian rhythms. Molecular clock cells in the brain’s suprachiasmatic nuclei (SCN) use electrical and chemical signals to orchestrate their activity and convey time of day information to the rest of the brain and body. To date, the long-lasting effects of regular physical exercise on SCN clock cell coordination and communication remain unresolved. Utilizing mouse models in which SCN intercellular neuropeptide signaling is impaired as well as those with intact SCN neurochemical signaling, we examined how daily scheduled voluntary exercise (SVE) influenced behavioral rhythms and SCN molecular and neuronal activities. We show that in mice with disrupted neuropeptide signaling, SVE promotes SCN clock cell synchrony and robust 24 h rhythms in behavior. Interestingly, in both intact and neuropeptide signaling deficient animals, SVE reduces SCN neural activity and alters GABAergic signaling. These findings illustrate the potential utility of regular exercise as a long-lasting and effective non-invasive intervention in the elderly or mentally ill where circadian rhythms can be blunted and poorly aligned to the external world. Using mice with disrupted neuropeptide signaling, Hughes et al. show that daily scheduled voluntary exercise (SVE) promotes suprachiasmatic nuclei (SCN) clock cell synchrony and robust 24 h rhythms in behavior. This study suggests the potential utility of regular exercise as a non-invasive intervention for the elderly or mentally ill, where circadian rhythms can be poorly aligned to the external world.
Collapse
Affiliation(s)
- Alun Thomas Lloyd Hughes
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,School of Biological and Environmental Sciences, Liverpool John Moores University, Liverpool, UK
| | - Rayna Eve Samuels
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Beatriz Baño-Otálora
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Mino David Charles Belle
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,University of Exeter Medical School, Exeter, UK
| | - Sven Wegner
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Clare Guilding
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,School of Medical Education, Newcastle University, Newcastle, UK
| | | | | | - John Gigg
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Hugh David Piggins
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK. .,School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, UK.
| |
Collapse
|
30
|
Cheng AH, Cheng HYM. Genesis of the Master Circadian Pacemaker in Mice. Front Neurosci 2021; 15:659974. [PMID: 33833665 PMCID: PMC8021851 DOI: 10.3389/fnins.2021.659974] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus is the central circadian clock of mammals. It is responsible for communicating temporal information to peripheral oscillators via humoral and endocrine signaling, ultimately controlling overt rhythms such as sleep-wake cycles, body temperature, and locomotor activity. Given the heterogeneity and complexity of the SCN, its genesis is tightly regulated by countless intrinsic and extrinsic factors. Here, we provide a brief overview of the development of the SCN, with special emphasis on the murine system.
Collapse
Affiliation(s)
- Arthur H. Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Hai-Ying Mary Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
31
|
Maejima T, Tsuno Y, Miyazaki S, Tsuneoka Y, Hasegawa E, Islam MT, Enoki R, Nakamura TJ, Mieda M. GABA from vasopressin neurons regulates the time at which suprachiasmatic nucleus molecular clocks enable circadian behavior. Proc Natl Acad Sci U S A 2021; 118:e2010168118. [PMID: 33526663 PMCID: PMC8017960 DOI: 10.1073/pnas.2010168118] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The suprachiasmatic nucleus (SCN), the central circadian pacemaker in mammals, is a network structure composed of multiple types of γ-aminobutyric acid (GABA)-ergic neurons and glial cells. However, the roles of GABA-mediated signaling in the SCN network remain controversial. Here, we report noticeable impairment of the circadian rhythm in mice with a specific deletion of the vesicular GABA transporter in arginine vasopressin (AVP)-producing neurons. These mice showed disturbed diurnal rhythms of GABAA receptor-mediated synaptic transmission in SCN neurons and marked lengthening of the activity time in circadian behavioral rhythms due to the extended interval between morning and evening locomotor activities. Synchrony of molecular circadian oscillations among SCN neurons did not significantly change, whereas the phase relationships between SCN molecular clocks and circadian morning/evening locomotor activities were altered significantly, as revealed by PER2::LUC imaging of SCN explants and in vivo recording of intracellular Ca2+ in SCN AVP neurons. In contrast, daily neuronal activity in SCN neurons in vivo clearly showed a bimodal pattern that correlated with dissociated morning/evening locomotor activities. Therefore, GABAergic transmission from AVP neurons regulates the timing of SCN neuronal firing to temporally restrict circadian behavior to appropriate time windows in SCN molecular clocks.
Collapse
Affiliation(s)
- Takashi Maejima
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 920-8640 Ishikawa, Japan
| | - Yusuke Tsuno
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 920-8640 Ishikawa, Japan
| | - Shota Miyazaki
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, 214-8571 Kanagawa, Japan
| | - Yousuke Tsuneoka
- Department of Anatomy, Faculty of Medicine, Toho University, 143-8540 Tokyo, Japan
| | - Emi Hasegawa
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 920-8640 Ishikawa, Japan
| | - Md Tarikul Islam
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 920-8640 Ishikawa, Japan
| | - Ryosuke Enoki
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 444-8787 Okazaki, Japan
- Division of Biophotonics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, 444-8787 Okazaki, Japan
| | - Takahiro J Nakamura
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, 214-8571 Kanagawa, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, 920-8640 Ishikawa, Japan;
| |
Collapse
|
32
|
Tamura EK, Oliveira-Silva KS, Ferreira-Moraes FA, Marinho EAV, Guerrero-Vargas NN. Circadian rhythms and substance use disorders: A bidirectional relationship. Pharmacol Biochem Behav 2021; 201:173105. [PMID: 33444601 DOI: 10.1016/j.pbb.2021.173105] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 01/23/2023]
Abstract
The circadian system organizes circadian rhythms (biological cycles that occur around 24 h) that couple environmental cues (zeitgebers) with internal functions of the organism. The misalignment between circadian rhythms and external cues is known as chronodisruption and contributes to the development of mental, metabolic and other disorders, including cancer, cardiovascular diseases and addictive disorders. Drug addiction represents a global public health concern and affects the health and well-being of individuals, families and communities. In this manuscript, we reviewed evidence indicating a bidirectional relationship between the circadian system and the development of addictive disorders. We provide information on the interaction between the circadian system and drug addiction for each drug or drug class (alcohol, cannabis, hallucinogens, psychostimulants and opioids). We also describe evidence showing that drug use follows a circadian pattern, which changes with the progression of addiction. Furthermore, clock gene expression is also altered during the development of drug addiction in many brain areas related to drug reward, drug seeking and relapse. The regulation of the glutamatergic and dopaminergic neurocircuitry by clock genes is postulated to be the main circadian mechanism underlying the escalation of drug addiction. The bidirectional interaction between the circadian system and drug addiction seems to be mediated by the effects caused by each drug or class of drugs of abuse. These studies provide new insights on the development of successful strategies aimed at restoring/stabilizing circadian rhythms to reduce the risk for addiction development and relapse.
Collapse
Affiliation(s)
- Eduardo K Tamura
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil.
| | - Kallyane S Oliveira-Silva
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Felipe A Ferreira-Moraes
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Eduardo A V Marinho
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Natalí N Guerrero-Vargas
- Department of Anatomy, Faculty of Medicine, Universidad Nacional Autonóma de México, Av Universidad 3000, Ciudad Universitaria, México City 04510, Mexico
| |
Collapse
|
33
|
Seasonality and light phase-resetting in the mammalian circadian rhythm. Sci Rep 2020; 10:19506. [PMID: 33177530 PMCID: PMC7658258 DOI: 10.1038/s41598-020-74002-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/18/2020] [Indexed: 11/13/2022] Open
Abstract
We study the impact of light on the mammalian circadian system using the theory of phase response curves. Using a recently developed ansatz we derive a low-dimensional macroscopic model for the core circadian clock in mammals. Significantly, the variables and parameters in our model have physiological interpretations and may be compared with experimental results. We focus on the effect of four key factors which help shape the mammalian phase response to light: heterogeneity in the population of oscillators, the structure of the typical light phase response curve, the fraction of oscillators which receive direct light input and changes in the coupling strengths associated with seasonal day-lengths. We find these factors can explain several experimental results and provide insight into the processing of light information in the mammalian circadian system. In particular, we find that the sensitivity of the circadian system to light may be modulated by changes in the relative coupling forces between the light sensing and non-sensing populations. Finally, we show how seasonal day-length, after-effects to light entrainment and seasonal variations in light sensitivity in the mammalian circadian clock are interrelated.
Collapse
|
34
|
Bioaminergic Responses in an In Vitro System Studying Human Gut Microbiota-Kiwifruit Interactions. Microorganisms 2020; 8:microorganisms8101582. [PMID: 33066564 PMCID: PMC7602194 DOI: 10.3390/microorganisms8101582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/09/2020] [Accepted: 10/13/2020] [Indexed: 01/04/2023] Open
Abstract
Whole kiwifruit ('Hayward' and 'Zesy002') were examined for their bioaminergic potential after being subjected to in vitro gastrointestinal digestion and colonic fermentation. Controls included the prebiotic inulin and water, a carbohydrate-free vehicle. The dopamine precursor l-dihydroxyphenylalanine (L-DOPA) and the serotonin precursor 5-hydroxytryptophan were increased in the kiwifruit gastrointestinal digesta ('Hayward' > 'Zesy002') in comparison to the water digesta. Fermentation of the digesta with human fecal bacteria for 18 h modulated the concentrations of bioamine metabolites. The most notable were the significant increases in L-DOPA ('Zesy002' > 'Hayward') and γ-aminobutyric acid (GABA) ('Hayward' > 'Zesy002'). Kiwifruit increased Bifidobacterium spp. and Veillonellaceae (correlating with L-DOPA increase), and Lachnospira spp. (correlating with GABA). The digesta and fermenta were incubated with Caco-2 cells for 3 h followed by gene expression analysis. Effects were seen on genes related to serotonin synthesis/re-uptake/conversion to melatonin, gut tight junction, inflammation and circadian rhythm with different digesta and fermenta from the four treatments. These indicate potential effects of the substrates and the microbially generated organic acid and bioamine metabolites on intestinal functions that have physiological relevance. Further studies are required to confirm the potential bioaminergic effects of gut microbiota-kiwifruit interactions.
Collapse
|
35
|
Roy U, Heredia-Muñoz MT, Stute L, Höfling C, Matysik J, Meijer JH, Roßner S, Alia A. Degeneration of the Suprachiasmatic Nucleus in an Alzheimer's Disease Mouse Model Monitored by in vivo Magnetic Resonance Relaxation Measurements and Immunohistochemistry. J Alzheimers Dis 2020; 69:363-375. [PMID: 30958376 DOI: 10.3233/jad-190037] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In Alzheimer's disease (AD), disturbances in the circadian rhythm and sleep-wake cycle are frequently observed. Both are controlled by the master clock: the suprachiasmatic nucleus (SCN), which was reported in postmortem studies of AD subjects to be compromised. However, the influence of age and gender on the biophysical integrity and subtle microstructural changes of SCN and mechanistic connections between SCN dysfunction and AD progression in vivo remain to be explored. In the present study, we utilized state-of-the-art in vivo magnetic resonance relaxation measurements in combination with immunohistochemistry to follow microstructural changes in SCN of the Tg2576 mouse model of AD. Longitudinal monitoring of in vivo T2 relaxation with age shows significant shortening of T2 values in the SCN of transgenic mice and more substantially in female transgenic than aged-matched controls. Multiexponential T2 analysis detected a unique long T2 component in SCN of transgenic mice which was absent in wild-type mice. Immunohistochemical examination revealed significantly elevated numbers of activated astrocytes and an increase in the astrocyte to neuron ratio in SCN of transgenic compared to wild-type mice. This increase was more substantial in female than in male transgenic mice. In addition, low GABA production in SCN of transgenic mice was detected. Our results offer a brief appraisal of SCN dysfunction in AD and demonstrate that inflammatory responses may be an underlying perpetrator for the changes in circadian rhythmicity and sleep disturbance in AD and could also be at the root of marked sex disparities observed in AD subjects.
Collapse
Affiliation(s)
- Upasana Roy
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany.,Institute of Analytical Chemistry, University of Leipzig, Leipzig, Germany
| | | | - Lara Stute
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany.,Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Corinna Höfling
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Jörg Matysik
- Institute of Analytical Chemistry, University of Leipzig, Leipzig, Germany
| | - Johanna H Meijer
- Department of Cell and Chemical Biology, Laboratory for Neurophysiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - A Alia
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany.,Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| |
Collapse
|
36
|
Todd WD. Potential Pathways for Circadian Dysfunction and Sundowning-Related Behavioral Aggression in Alzheimer's Disease and Related Dementias. Front Neurosci 2020; 14:910. [PMID: 33013301 PMCID: PMC7494756 DOI: 10.3389/fnins.2020.00910] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/06/2020] [Indexed: 12/19/2022] Open
Abstract
Patients with Alzheimer's disease (AD) and related dementias are commonly reported to exhibit aggressive behavior and other emotional behavioral disturbances, which create a tremendous caretaker burden. There has been an abundance of work highlighting the importance of circadian function on mood and emotional behavioral regulation, and recent evidence demonstrates that a specific hypothalamic pathway links the circadian system to neurons that modulate aggressive behavior, regulating the propensity for aggression across the day. Such shared circuitry may have important ramifications for clarifying the complex interactions underlying "sundowning syndrome," a poorly understood (and even controversial) clinical phenomenon in AD and dementia patients that is characterized by agitation, aggression, and delirium during the late afternoon and early evening hours. The goal of this review is to highlight the potential output and input pathways of the circadian system that may underlie circadian dysfunction and behavioral aggression associated with sundowning syndrome, and to discuss possible ways these pathways might inform specific interventions for treatment. Moreover, the apparent bidirectional relationship between chronic disruptions of circadian and sleep-wake regulation and the pathology and symptoms of AD suggest that understanding the role of these circuits in such neurobehavioral pathologies could lead to better diagnostic or even preventive measures.
Collapse
Affiliation(s)
- William D Todd
- Program in Neuroscience, Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
| |
Collapse
|
37
|
Suprachiasmatic VIP neurons are required for normal circadian rhythmicity and comprised of molecularly distinct subpopulations. Nat Commun 2020; 11:4410. [PMID: 32879310 PMCID: PMC7468160 DOI: 10.1038/s41467-020-17197-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 06/12/2020] [Indexed: 12/02/2022] Open
Abstract
The hypothalamic suprachiasmatic (SCN) clock contains several neurochemically defined cell groups that contribute to the genesis of circadian rhythms. Using cell-specific and genetically targeted approaches we have confirmed an indispensable role for vasoactive intestinal polypeptide-expressing SCN (SCNVIP) neurons, including their molecular clock, in generating the mammalian locomotor activity (LMA) circadian rhythm. Optogenetic-assisted circuit mapping revealed functional, di-synaptic connectivity between SCNVIP neurons and dorsomedial hypothalamic neurons, providing a circuit substrate by which SCNVIP neurons may regulate LMA rhythms. In vivo photometry revealed that while SCNVIP neurons are acutely responsive to light, their activity is otherwise behavioral state invariant. Single-nuclei RNA-sequencing revealed that SCNVIP neurons comprise two transcriptionally distinct subtypes, including putative pacemaker and non-pacemaker populations. Altogether, our work establishes necessity of SCNVIP neurons for the LMA circadian rhythm, elucidates organization of circadian outflow from and modulatory input to SCNVIP cells, and demonstrates a subpopulation-level molecular heterogeneity that suggests distinct functions for specific SCNVIP subtypes. Cell groups in the hypothalamic suprachiasmatic clock contribute to the genesis of circadian rhythms. The authors identified two populations of vasoactive intestinal polypeptide-expressing neurons in the suprachiasmatic nucleus which regulate locomotor circadian rhythm in mice.
Collapse
|
38
|
Olde Engberink AHO, Huisman J, Michel S, Meijer JH. Brief light exposure at dawn and dusk can encode day-length in the neuronal network of the mammalian circadian pacemaker. FASEB J 2020; 34:13685-13695. [PMID: 32869393 DOI: 10.1096/fj.202001133rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/23/2020] [Accepted: 07/29/2020] [Indexed: 11/11/2022]
Abstract
The central circadian pacemaker in mammals, the suprachiasmatic nucleus (SCN), is important for daily as well as seasonal rhythms. The SCN encodes seasonal changes in day length by adjusting phase distribution among oscillating neurons thereby shaping the output signal used for adaptation of physiology and behavior. It is well-established that brief light exposure at the beginning and end of the day, also referred to as "skeleton" light pulses, are sufficient to evoke the seasonal behavioral phenotype. However, the effect of skeleton light exposure on SCN network reorganization remains unknown. Therefore, we exposed mice to brief morning and evening light pulses that mark the time of dawn and dusk in a short winter- or a long summer day. Single-cell PER2::LUC recordings, electrophysiological recordings of SCN activity, and measurements of GABA response polarity revealed that skeleton light-regimes affected the SCN network to the same degree as full photoperiod. These results indicate the powerful, yet potentially harmful effects of even relatively short light exposures during the evening or night for nocturnal animals.
Collapse
Affiliation(s)
- Anneke H O Olde Engberink
- Department of Cellular and Chemical Biology, Laboratory for Neurophysiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Job Huisman
- Department of Cellular and Chemical Biology, Laboratory for Neurophysiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Stephan Michel
- Department of Cellular and Chemical Biology, Laboratory for Neurophysiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Johanna H Meijer
- Department of Cellular and Chemical Biology, Laboratory for Neurophysiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
39
|
Ono D, Honma KI, Honma S. GABAergic mechanisms in the suprachiasmatic nucleus that influence circadian rhythm. J Neurochem 2020; 157:31-41. [PMID: 32198942 DOI: 10.1111/jnc.15012] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 01/23/2023]
Abstract
The mammalian central circadian clock is located in the suprachiasmatic nucleus (SCN) of the hypothalamus. The SCN contains multiple circadian oscillators which synchronize with each other via several neurotransmitters. Importantly, an inhibitory neurotransmitter, γ-amino butyric acid (GABA), is expressed in almost all SCN neurons. In this review, we discuss how GABA influences circadian rhythms in the SCN. Excitatory and inhibitory effects of GABA may depend on intracellular Cl- concentration, in which several factors such as day-length, time of day, development, and region in the SCN may be involved. GABA also mediates oscillatory coupling of the circadian rhythms in the SCN. Recent genetic approaches reveal that GABA refines circadian output rhythms, but not circadian oscillations in the SCN. Since several efferent projections of the SCN have been suggested, GABA might work downstream of neuronal pathways from the SCN which regulate the temporal order of physiology and behavior.
Collapse
Affiliation(s)
- Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ken-Ichi Honma
- Research and Education Center for Brain Science, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Sato Honma
- Research and Education Center for Brain Science, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
40
|
Mieda M. The central circadian clock of the suprachiasmatic nucleus as an ensemble of multiple oscillatory neurons. Neurosci Res 2020; 156:24-31. [DOI: 10.1016/j.neures.2019.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 08/09/2019] [Indexed: 10/26/2022]
|
41
|
McNeill JK, Walton JC, Ryu V, Albers HE. The Excitatory Effects of GABA within the Suprachiasmatic Nucleus: Regulation of Na-K-2Cl Cotransporters (NKCCs) by Environmental Lighting Conditions. J Biol Rhythms 2020; 35:275-286. [PMID: 32406304 DOI: 10.1177/0748730420924271] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The suprachiasmatic nucleus (SCN) contains a pacemaker that generates circadian rhythms and entrains them with the 24-h light-dark cycle (LD). The SCN is composed of 16,000 to 20,000 heterogeneous neurons in bilaterally paired nuclei. γ-amino butyric acid (GABA) is the primary neurochemical signal within the SCN and plays a key role in regulating circadian function. While GABA is the primary inhibitory neurotransmitter in the brain, there is now evidence that GABA can also exert excitatory effects in the adult brain. Cation chloride cotransporters determine the effects of GABA on chloride equilibrium, thereby determining whether GABA produces hyperpolarizing or depolarizing actions following activation of GABAA receptors. The activity of Na-K-2Cl cotransporter1 (NKCC1), the most prevalent chloride influx cotransporter isoform in the brain, plays a critical role in determining whether GABA has depolarizing effects. In the present study, we tested the hypothesis that NKCC1 protein expression in the SCN is regulated by environmental lighting and displays daily and circadian changes in the intact circadian system of the Syrian hamster. In hamsters housed in constant light (LL), the overall NKCC1 immunoreactivity (NKCC1-ir) in the SCN was significantly greater than in hamsters housed in LD or constant darkness (DD), although NKCC1 protein levels in the SCN were not different between hamsters housed in LD and DD. In hamsters housed in LD cycles, no differences in NKCC1-ir within the SCN were observed over the 24-h cycle. NKCC1 protein in the SCN was found to vary significantly over the circadian cycle in hamsters housed in free-running conditions. Overall, NKCC1 protein was greater in the ventral SCN than in the dorsal SCN, although no significant differences were observed across lighting conditions or time of day in either subregion. These data support the hypothesis that NKCC1 protein expression can be regulated by environmental lighting and circadian mechanisms within the SCN.
Collapse
Affiliation(s)
- John K McNeill
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia
| | - James C Walton
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia
| | - Vitaly Ryu
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia
| | - H Elliott Albers
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia
| |
Collapse
|
42
|
Sanchez REA, Bussi IL, Ben-Hamo M, Caldart CS, Catterall WA, De La Iglesia HO. Circadian regulation of sleep in a pre-clinical model of Dravet syndrome: dynamics of sleep stage and siesta re-entrainment. Sleep 2020; 42:5539047. [PMID: 31346614 DOI: 10.1093/sleep/zsz173] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/24/2019] [Indexed: 12/11/2022] Open
Abstract
STUDY OBJECTIVES Sleep disturbances are common co-morbidities of epileptic disorders. Dravet syndrome (DS) is an intractable epilepsy accompanied by disturbed sleep. While there is evidence that daily sleep timing is disrupted in DS, the difficulty of chronically recording polysomnographic sleep from patients has left our understanding of the effect of DS on circadian sleep regulation incomplete. We aim to characterize circadian sleep regulation in a mouse model of DS. METHODS Here we exploit long-term electrocorticographic recordings of sleep in a mouse model of DS in which one copy of the Scn1a gene is deleted. This model both genocopies and phenocopies the disease in humans. We test the hypothesis that the deletion of Scn1a in DS mice is associated with impaired circadian regulation of sleep. RESULTS We find that DS mice show impairments in circadian sleep regulation, including a fragmented rhythm of non-rapid eye movement (NREM) sleep and an elongated circadian period of sleep. Next, we characterize re-entrainment of sleep stages and siesta following jet lag in the mouse. Strikingly, we find that re-entrainment of sleep following jet lag is normal in DS mice, in contrast to previous demonstrations of slowed re-entrainment of wheel-running activity. Finally, we report that DS mice are more likely to have an absent or altered daily "siesta". CONCLUSIONS Our findings support the hypothesis that the circadian regulation of sleep is altered in DS and highlight the value of long-term chronic polysomnographic recording in studying the role of the circadian clock on sleep/wake cycles in pre-clinical models of disease.
Collapse
Affiliation(s)
- Raymond E A Sanchez
- Department of Biology, University of Washington, Seattle, WA.,Graduate Program in Neuroscience, University of Washington, Seattle WA
| | - Ivana L Bussi
- Department of Biology, University of Washington, Seattle, WA
| | - Miriam Ben-Hamo
- Department of Biology, University of Washington, Seattle, WA
| | | | - William A Catterall
- Graduate Program in Neuroscience, University of Washington, Seattle WA.,Department of Pharmacology, University of Washington, Seattle WA
| | - Horacio O De La Iglesia
- Department of Biology, University of Washington, Seattle, WA.,Graduate Program in Neuroscience, University of Washington, Seattle WA
| |
Collapse
|
43
|
Han K, Mei L, Zhong R, Pang Y, Zhang EE, Huang Y. A microfluidic approach for experimentally modelling the intercellular coupling system of a mammalian circadian clock at single-cell level. LAB ON A CHIP 2020; 20:1204-1211. [PMID: 32149320 DOI: 10.1039/d0lc00140f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In mammals, it is believed that the intercellular coupling mechanism between neurons in the suprachiasmatic nucleus (SCN) confers robustness and distinguishes the central clock from peripheral circadian oscillators. Current in vitro culturing methods used in Petri dishes to study intercellular coupling by exogenous factors invariably cause perturbations, such as simple media changes. Here, we design a microfluidic device to quantitatively study the intercellular coupling mechanism of circadian clock at the single cell level, and demonstrate that vasoactive intestinal peptide (VIP) induced coupling in clock mutant Cry1-/- mouse adult fibroblasts engineered to express the VIP receptor, VPAC2, is sufficient to synchronize and maintain robust circadian oscillations. Our study provides a proof-of-concept platform to reconstitute the intercellular coupling system of the central clock using uncoupled, single fibroblast cells in vitro, to mimic SCN slice cultures ex vivo and mouse behavior in vivo phenotypically. Such a versatile microfluidic platform may greatly facilitate the studies of intercellular regulation networks, and provide new insights into the coupling mechanisms of the circadian clock.
Collapse
Affiliation(s)
- Kui Han
- Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics (ICG), College of Engineering, College of Chemistry, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.
| | | | | | | | | | | |
Collapse
|
44
|
Harvey JRM, Plante AE, Meredith AL. Ion Channels Controlling Circadian Rhythms in Suprachiasmatic Nucleus Excitability. Physiol Rev 2020; 100:1415-1454. [PMID: 32163720 DOI: 10.1152/physrev.00027.2019] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Animals synchronize to the environmental day-night cycle by means of an internal circadian clock in the brain. In mammals, this timekeeping mechanism is housed in the suprachiasmatic nucleus (SCN) of the hypothalamus and is entrained by light input from the retina. One output of the SCN is a neural code for circadian time, which arises from the collective activity of neurons within the SCN circuit and comprises two fundamental components: 1) periodic alterations in the spontaneous excitability of individual neurons that result in higher firing rates during the day and lower firing rates at night, and 2) synchronization of these cellular oscillations throughout the SCN. In this review, we summarize current evidence for the identity of ion channels in SCN neurons and the mechanisms by which they set the rhythmic parameters of the time code. During the day, voltage-dependent and independent Na+ and Ca2+ currents, as well as several K+ currents, contribute to increased membrane excitability and therefore higher firing frequency. At night, an increase in different K+ currents, including Ca2+-activated BK currents, contribute to membrane hyperpolarization and decreased firing. Layered on top of these intrinsically regulated changes in membrane excitability, more than a dozen neuromodulators influence action potential activity and rhythmicity in SCN neurons, facilitating both synchronization and plasticity of the neural code.
Collapse
Affiliation(s)
- Jenna R M Harvey
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Amber E Plante
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Andrea L Meredith
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
45
|
Bartman CM, Eckle T. Circadian-Hypoxia Link and its Potential for Treatment of Cardiovascular Disease. Curr Pharm Des 2020; 25:1075-1090. [PMID: 31096895 DOI: 10.2174/1381612825666190516081612] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/03/2019] [Indexed: 12/29/2022]
Abstract
Throughout the evolutionary time, all organisms and species on Earth evolved with an adaptation to consistent oscillations of sunlight and darkness, now recognized as 'circadian rhythm.' Single-cellular to multisystem organisms use circadian biology to synchronize to the external environment and provide predictive adaptation to changes in cellular homeostasis. Dysregulation of circadian biology has been implicated in numerous prevalent human diseases, and subsequently targeting the circadian machinery may provide innovative preventative or treatment strategies. Discovery of 'peripheral circadian clocks' unleashed widespread investigations into the potential roles of clock biology in cellular, tissue, and organ function in healthy and diseased states. Particularly, oxygen-sensing pathways (e.g. hypoxia inducible factor, HIF1), are critical for adaptation to changes in oxygen availability in diseases such as myocardial ischemia. Recent investigations have identified a connection between the circadian rhythm protein Period 2 (PER2) and HIF1A that may elucidate an evolutionarily conserved cellular network that can be targeted to manipulate metabolic function in stressed conditions like hypoxia or ischemia. Understanding the link between circadian and hypoxia pathways may provide insights and subsequent innovative therapeutic strategies for patients with myocardial ischemia. This review addresses our current understanding of the connection between light-sensing pathways (PER2), and oxygen-sensing pathways (HIF1A), in the context of myocardial ischemia and lays the groundwork for future studies to take advantage of these two evolutionarily conserved pathways in the treatment of myocardial ischemia.
Collapse
Affiliation(s)
- Colleen Marie Bartman
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, Graduate Training Program in Cell Biology, Stem Cells, and Development, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Tobias Eckle
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, Graduate Training Program in Cell Biology, Stem Cells, and Development, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
46
|
Abstract
The circadian clock is an endogenous, time-tracking system that directs multiple metabolic and physiological functions required for homeostasis. The master or central clock located within the suprachiasmatic nucleus in the hypothalamus governs peripheral clocks present in all systemic tissues, contributing to their alignment and ultimately to temporal coordination of physiology. Accumulating evidence reveals the presence of additional clocks in the brain and suggests the possibility that circadian circuits may feed back to these from the periphery. Here, we highlight recent advances in the communications between clocks and discuss how they relate to circadian physiology and metabolism.
Collapse
Affiliation(s)
- Carolina Magdalen Greco
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, School of Medicine, University of California, Irvine, CA, USA
| | - Paolo Sassone-Corsi
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, School of Medicine, University of California, Irvine, CA, USA.
| |
Collapse
|
47
|
Rohr KE, Pancholi H, Haider S, Karow C, Modert D, Raddatz NJ, Evans J. Seasonal plasticity in GABA A signaling is necessary for restoring phase synchrony in the master circadian clock network. eLife 2019; 8:49578. [PMID: 31746738 PMCID: PMC6867713 DOI: 10.7554/elife.49578] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 10/15/2019] [Indexed: 12/14/2022] Open
Abstract
Annual changes in the environment threaten survival, and numerous biological processes in mammals adjust to this challenge via seasonal encoding by the suprachiasmatic nucleus (SCN). To tune behavior according to day length, SCN neurons display unified rhythms with synchronous phasing when days are short, but will divide into two sub-clusters when days are long. The transition between SCN states is critical for maintaining behavioral responses to seasonal change, but the mechanisms regulating this form of neuroplasticity remain unclear. Here we identify that a switch in chloride transport and GABAA signaling is critical for maintaining state plasticity in the SCN network. Further, we reveal that blocking excitatory GABAA signaling locks the SCN into its long day state. Collectively, these data demonstrate that plasticity in GABAA signaling dictates how clock neurons interact to maintain environmental encoding. Further, this work highlights factors that may influence susceptibility to seasonal disorders in humans. In winter, as the days become shorter, millions of people find that their mood and energy levels start to drop. They crave carbohydrates, struggle with their weight, and find it harder to get out of bed in the mornings. These individuals are suffering from the ‘winter blues’ or seasonal affective disorder (SAD), and most find that their symptoms spontaneously improve in the spring when the days become longer again. Many also benefit from bright light therapy during the winter months, but not everyone responds fully to this treatment, so additional options are needed. The winter blues occur when the brain adjusts to changes in day length with the onset of winter. The brain region responsible for making this adjustment is the suprachiasmatic nucleus (SCN). The SCN is the master clock of the brain that coordinates the body’s circadian rhythms – the daily fluctuations in things like appetite, body temperature, sleep and wakefulness. But as well as being the brain’s clock, the SCN is also the brain’s calendar. In winter, when the days are short, SCN neurons coordinate their activity and fire in synchrony. But in summer, when the days are long, SCN neurons divide into two clusters, which fire at different times. By transitioning between these two states, the SCN helps the body adjust to seasonal changes in day length. Rohr, Pancholi et al. now provide new insight into the mechanism behind this process by showing that light alters the neurochemistry of the SCN. Exposing mice to long days causes a brain chemical called GABA to switch from inhibiting neurons in the SCN to activating them. Blocking this switch from inhibition to activation locks the SCN into its 'summer state'. Rohr, Pancholi et al. propose that this failure to transition to the winter state may be an interesting way to prevent the winter blues. While much remains to be learned about this process, these findings pave the way for better understanding the neurobiology of winter depression and how best to treat it.
Collapse
Affiliation(s)
- Kayla E Rohr
- Department of Biomedical Sciences, Marquette University, Milwaukee, United States
| | - Harshida Pancholi
- Department of Biomedical Sciences, Marquette University, Milwaukee, United States
| | - Shabi Haider
- Department of Biomedical Sciences, Marquette University, Milwaukee, United States
| | - Christopher Karow
- Department of Biomedical Sciences, Marquette University, Milwaukee, United States
| | - David Modert
- Department of Biomedical Sciences, Marquette University, Milwaukee, United States
| | - Nicholas J Raddatz
- Department of Biomedical Sciences, Marquette University, Milwaukee, United States
| | - Jennifer Evans
- Department of Biomedical Sciences, Marquette University, Milwaukee, United States
| |
Collapse
|
48
|
Chowdhury D, Wang C, Lu AP, Zhu HL. Understanding Quantitative Circadian Regulations Are Crucial Towards Advancing Chronotherapy. Cells 2019; 8:cells8080883. [PMID: 31412622 PMCID: PMC6721722 DOI: 10.3390/cells8080883] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/01/2019] [Accepted: 08/09/2019] [Indexed: 12/19/2022] Open
Abstract
Circadian rhythms have a deep impact on most aspects of physiology. In most organisms, especially mammals, the biological rhythms are maintained by the indigenous circadian clockwork around geophysical time (~24-h). These rhythms originate inside cells. Several core components are interconnected through transcriptional/translational feedback loops to generate molecular oscillations. They are tightly controlled over time. Also, they exert temporal controls over many fundamental physiological activities. This helps in coordinating the body’s internal time with the external environments. The mammalian circadian clockwork is composed of a hierarchy of oscillators, which play roles at molecular, cellular, and higher levels. The master oscillation has been found to be developed at the hypothalamic suprachiasmatic nucleus in the brain. It acts as the core pacemaker and drives the transmission of the oscillation signals. These signals are distributed across different peripheral tissues through humoral and neural connections. The synchronization among the master oscillator and tissue-specific oscillators offer overall temporal stability to mammals. Recent technological advancements help us to study the circadian rhythms at dynamic scale and systems level. Here, we outline the current understanding of circadian clockwork in terms of molecular mechanisms and interdisciplinary concepts. We have also focused on the importance of the integrative approach to decode several crucial intricacies. This review indicates the emergence of such a comprehensive approach. It will essentially accelerate the circadian research with more innovative strategies, such as developing evidence-based chronotherapeutics to restore de-synchronized circadian rhythms.
Collapse
Affiliation(s)
- Debajyoti Chowdhury
- HKBU Institute for Research and Continuing Education, Shenzhen 518057, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
| | - Chao Wang
- HKBU Institute for Research and Continuing Education, Shenzhen 518057, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
| | - Ai-Ping Lu
- HKBU Institute for Research and Continuing Education, Shenzhen 518057, China.
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Hai-Long Zhu
- HKBU Institute for Research and Continuing Education, Shenzhen 518057, China.
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| |
Collapse
|
49
|
Gaspar LS, Álvaro AR, Carmo‐Silva S, Mendes AF, Relógio A, Cavadas C. The importance of determining circadian parameters in pharmacological studies. Br J Pharmacol 2019; 176:2827-2847. [PMID: 31099023 PMCID: PMC6637036 DOI: 10.1111/bph.14712] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/26/2019] [Accepted: 04/01/2019] [Indexed: 12/25/2022] Open
Abstract
In mammals, most molecular and cellular processes show circadian changes, leading to daily variations in physiology and ultimately in behaviour. Such daily variations induce a temporal coordination of processes that is essential to ensure homeostasis and health. Thus, it is of no surprise that pharmacokinetics (PK) and pharmacodynamics (PD) of many drugs are also subject to circadian variations, profoundly affecting their efficacy and tolerability. Understanding how circadian rhythms influence drug PK, PD, and toxicity might significantly improve treatment efficacy and decrease related side effects. Therefore, it is essential to take circadian variations into account and to determine circadian parameters in pharmacological studies, especially when drugs have a short half-life or target rhythmic processes. This review provides an overview of the current knowledge on circadian rhythms and their relevance to the field of pharmacology. Methodologies to evaluate circadian rhythms in vitro, in rodent models and in humans, from experimental to computational approaches, are described and discussed. Lastly, we aim at alerting the scientific, medical, and regulatory communities to the relevance of the physiological time, as a key parameter to be considered when designing pharmacological studies. This will eventually lead to more successful preclinical and clinical trials and pave the way to a more personalized treatment to the benefit of the patients.
Collapse
Affiliation(s)
- Laetitia S. Gaspar
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB)University of CoimbraCoimbraPortugal
- Institute for Interdisciplinary Research (IIIUC)University of CoimbraCoimbraPortugal
| | - Ana Rita Álvaro
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB)University of CoimbraCoimbraPortugal
| | - Sara Carmo‐Silva
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB)University of CoimbraCoimbraPortugal
| | - Alexandrina Ferreira Mendes
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB)University of CoimbraCoimbraPortugal
- Faculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - Angela Relógio
- Institute for Theoretical BiologyCharité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt—Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research CenterCharité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt—Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Cláudia Cavadas
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Center for Innovation in Biomedicine and Biotechnology (CIBB)University of CoimbraCoimbraPortugal
- Faculty of PharmacyUniversity of CoimbraCoimbraPortugal
| |
Collapse
|
50
|
Lugena AB, Zhang Y, Menet JS, Merlin C. Genome-wide discovery of the daily transcriptome, DNA regulatory elements and transcription factor occupancy in the monarch butterfly brain. PLoS Genet 2019; 15:e1008265. [PMID: 31335862 PMCID: PMC6677324 DOI: 10.1371/journal.pgen.1008265] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 08/02/2019] [Accepted: 06/21/2019] [Indexed: 12/20/2022] Open
Abstract
The Eastern North American monarch butterfly, Danaus plexippus, is famous for its spectacular seasonal long-distance migration. In recent years, it has also emerged as a novel system to study how animal circadian clocks keep track of time and regulate ecologically relevant daily rhythmic activities and seasonal behavioral outputs. However, unlike in Drosophila and the mouse, little work has been undertaken in the monarch to identify rhythmic genes at the genome-wide level and elucidate the regulation of their diurnal expression. Here, we used RNA-sequencing and Assay for Transposase-Accessible Chromatin (ATAC)-sequencing to profile the diurnal transcriptome, open chromatin regions, and transcription factor (TF) footprints in the brain of wild-type monarchs and of monarchs with impaired clock function, including Cryptochrome 2 (Cry2), Clock (Clk), and Cycle-like loss-of-function mutants. We identified 217 rhythmically expressed genes in the monarch brain; many of them were involved in the regulation of biological processes key to brain function, such as glucose metabolism and neurotransmission. Surprisingly, we found no significant time-of-day and genotype-dependent changes in chromatin accessibility in the brain. Instead, we found the existence of a temporal regulation of TF occupancy within open chromatin regions in the vicinity of rhythmic genes in the brains of wild-type monarchs, which is disrupted in clock deficient mutants. Together, this work identifies for the first time the rhythmic genes and modes of regulation by which diurnal transcription rhythms are regulated in the monarch brain. It also illustrates the power of ATAC-sequencing to profile genome-wide regulatory elements and TF binding in a non-model organism for which TF-specific antibodies are not yet available. With a rich biology that includes a clock-regulated migratory behavior and a circadian clock possessing mammalian clock orthologues, the monarch butterfly is an unconventional system with broad appeal to study circadian and seasonal rhythms. While clockwork mechanisms and rhythmic behavioral outputs have been studied in this species, the rhythmic genes that regulate rhythmic daily and seasonal activities remain largely unknown. Likewise, the mechanisms regulating rhythmic gene expression have not been explored in the monarch. Here, we applied genome-wide sequencing approaches to identify genes with rhythmic diurnal expression in the monarch brain, revealing the coordination of key pathways for brain function. We also identified the monarch brain open chromatin regions and provide evidence that regulation of rhythmic gene expression does not occur through temporal regulation of chromatin opening but rather by the time-of-day dependent binding of transcription factors in cis-regulatory elements. Together, our data extend our knowledge of the molecular rhythmic pathways, which may prove important in understanding the mechanisms underlying the daily and seasonal biology of the migratory monarch butterflies.
Collapse
Affiliation(s)
- Aldrin B. Lugena
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, Texas, United States of America
| | - Ying Zhang
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, Texas, United States of America
| | - Jerome S. Menet
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, Texas, United States of America
| | - Christine Merlin
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, Texas, United States of America
- * E-mail:
| |
Collapse
|