1
|
Ma S, Ge J, Chen X, Chen Z, Han Y, Xie Z, Chen J, Dai H, Xiao Q, Xu L, Jia Y. Dietary calcium intake at breakfast is associated with a lower risk of cognitive impairment than at dinner in Chinese adults: the CHNS cohort study. Asia Pac J Clin Nutr 2024; 33:405-412. [PMID: 38965728 PMCID: PMC11397565 DOI: 10.6133/apjcn.202409_33(3).0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/14/2024] [Accepted: 04/09/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND AND OBJECTIVES If the proportion of calcium intake over a whole day is related to the risk of cognitive impairment in adults is still largely unknown. This research aimed to examine the relation of dietary calcium intake at dinner versus breakfast with the risk of cognitive impairment by using data from the China Health and Nutrition Survey (CHNS). METHODS AND STUDY DESIGN A total of 2,099 participants (including 668 cognitive impairment) in the CHNS (1997-2006) were included. The participants were categorized into 5 groups in accordance with the ratio of dietary calcium intake at dinner and breakfast (Δ = dinner/breakfast). After adjustment was conducted for a series of confounding factors, Cox hazard regression modelling was performed to discuss the relation of Δ with cognitive impairment. Dietary substitution models were used to explore the changes in cognitive impairment risk when a 5% dietary calcium intake at dinner was replaced with dietary calcium intake at breakfast. RESULTS Participants in the highest distribution of Δ showed a greater susceptibility to cognitive impairment than those in the lowest quintile, with an adjusted hazard ratio of cognitive impairment of 1.38 (95% CI: 1.08-1.76). When maintaining total calcium intake, substituting 5% of dietary calcium intake at dinner with calcium intake at breakfast was related to an 8% decrease in the risk of cognitive impairment. CONCLUSIONS Higher dietary calcium intake at dinner was associated with an increased risk of cognitive impairment, emphasizing the importance of appropriately distributing dietary calcium intake between breakfast and dinner.
Collapse
Affiliation(s)
- Shuli Ma
- School of Public Health, Qiqihar Medical University, Qiqihar, China
| | - Jie Ge
- School of Public Health, Qiqihar Medical University, Qiqihar, China
| | - Xiaoting Chen
- Sanitary Analysis Center, Scientific Research Office, Qiqihar Medical University, Qiqihar, China
| | - Zhe Chen
- School of Public Health, Qiqihar Medical University, Qiqihar, China
| | - Yunfeng Han
- School of Public Health, Qiqihar Medical University, Qiqihar, China
| | - Zhiping Xie
- School of Public Health, Qiqihar Medical University, Qiqihar, China
| | - Jiaxin Chen
- School of Public Health, Qiqihar Medical University, Qiqihar, China
| | - Hang Dai
- Foreign Language Department, Qiqihar Medical University, Qiqihar, China
| | - Qiong Xiao
- School of Nursing, Qiqihar Medical University, Qiqihar, China
| | - Liang Xu
- State-owned Assets Administration Office, Qiqihar Medical University, Qiqihar, China
| | - Yuehui Jia
- School of Public Health, Qiqihar Medical University, Qiqihar, China.
| |
Collapse
|
2
|
Curtis L, Piggins HD. Diverse genetic alteration dysregulates neuropeptide and intracellular signalling in the suprachiasmatic nuclei. Eur J Neurosci 2024; 60:3921-3945. [PMID: 38924215 DOI: 10.1111/ejn.16443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/12/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024]
Abstract
In mammals, intrinsic 24 h or circadian rhythms are primarily generated by the suprachiasmatic nuclei (SCN). Rhythmic daily changes in the transcriptome and proteome of SCN cells are controlled by interlocking transcription-translation feedback loops (TTFLs) of core clock genes and their proteins. SCN cells function as autonomous circadian oscillators, which synchronize through intercellular neuropeptide signalling. Physiological and behavioural rhythms can be severely disrupted by genetic modification of a diverse range of genes and proteins in the SCN. With the advent of next generation sequencing, there is unprecedented information on the molecular profile of the SCN and how it is affected by genetically targeted alteration. However, whether the expression of some genes is more readily affected by genetic alteration of the SCN is unclear. Here, using publicly available datasets from recent RNA-seq assessments of the SCN from genetically altered and control mice, we evaluated whether there are commonalities in transcriptome dysregulation. This was completed for four different phases across the 24 h cycle and was augmented by Gene Ontology Molecular Function (GO:MF) and promoter analysis. Common differentially expressed genes (DEGs) and/or enriched GO:MF terms included signalling molecules, their receptors, and core clock components. Finally, examination of the JASPAR database indicated that E-box and CRE elements in the promoter regions of several commonly dysregulated genes. From this analysis, we identify differential expression of genes coding for molecules involved in SCN intra- and intercellular signalling as a potential cause of abnormal circadian rhythms.
Collapse
Affiliation(s)
- Lucy Curtis
- School of Biological Sciences, University of Bristol, Bristol, UK
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, UK
| | - Hugh D Piggins
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
3
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [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
|
4
|
Abstract
The blood-brain barrier (BBB) is a critical interface separating the central nervous system from the peripheral circulation, ensuring brain homeostasis and function. Recent research has unveiled a profound connection between the BBB and circadian rhythms, the endogenous oscillations synchronizing biological processes with the 24-hour light-dark cycle. This review explores the significance of circadian rhythms in the context of BBB functions, with an emphasis on substrate passage through the BBB. Our discussion includes efflux transporters and the molecular timing mechanisms that regulate their activities. A significant focus of this review is the potential implications of chronotherapy, leveraging our knowledge of circadian rhythms for improving drug delivery to the brain. Understanding the temporal changes in BBB can lead to optimized timing of drug administration, to enhance therapeutic efficacy for neurological disorders while reducing side effects. By elucidating the interplay between circadian rhythms and drug transport across the BBB, this review offers insights into innovative therapeutic interventions.
Collapse
Affiliation(s)
- Mari Kim
- Cell Biology Department, Emory University, Atlanta, GA, USA (M.K., S.L.Z.)
| | - Richard F Keep
- Neurosurgery, University of Michigan, Ann Arbor, MI, USA (R.F.K.)
| | - Shirley L Zhang
- Cell Biology Department, Emory University, Atlanta, GA, USA (M.K., S.L.Z.)
| |
Collapse
|
5
|
Li W, Tiedt S, Lawrence JH, Harrington ME, Musiek ES, Lo EH. Circadian Biology and the Neurovascular Unit. Circ Res 2024; 134:748-769. [PMID: 38484026 DOI: 10.1161/circresaha.124.323514] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/14/2024] [Indexed: 03/19/2024]
Abstract
Mammalian physiology and cellular function are subject to significant oscillations over the course of every 24-hour day. It is likely that these daily rhythms will affect function as well as mechanisms of disease in the central nervous system. In this review, we attempt to survey and synthesize emerging studies that investigate how circadian biology may influence the neurovascular unit. We examine how circadian clocks may operate in neural, glial, and vascular compartments, review how circadian mechanisms regulate cell-cell signaling, assess interactions with aging and vascular comorbidities, and finally ask whether and how circadian effects and disruptions in rhythms may influence the risk and progression of pathophysiology in cerebrovascular disease. Overcoming identified challenges and leveraging opportunities for future research might support the development of novel circadian-based treatments for stroke.
Collapse
Affiliation(s)
- Wenlu Li
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (W.L., E.H.L.)
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
| | - Steffen Tiedt
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany (S.T.)
| | - Jennifer H Lawrence
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
- Department of Neurology, Washington University School of Medicine, St. Louis, MO (J.H.L., E.S.M.)
| | - Mary E Harrington
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
- Neuroscience Program, Smith College, Northampton, MA (M.E.H.)
| | - Erik S Musiek
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
- Department of Neurology, Washington University School of Medicine, St. Louis, MO (J.H.L., E.S.M.)
| | - Eng H Lo
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (W.L., E.H.L.)
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
| |
Collapse
|
6
|
Wang Y, Dong Y, Zhai Q, Zhang W, Xu Y, Yang L. A critical signal for phenotype transition driven by negative feedback loops. iScience 2024; 27:108716. [PMID: 38226166 PMCID: PMC10788427 DOI: 10.1016/j.isci.2023.108716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/13/2023] [Accepted: 12/11/2023] [Indexed: 01/17/2024] Open
Abstract
The biological rhythms governed by negative feedback loops have undergone extensive investigation. However, developing reliable and versatile warning signals to predict periodic fluctuations in physiological processes and behaviors associated with these rhythms remains a challenge. Here, we monitored the heart rate and tracked ovulation dates of 91 fertile women. The finding strongly links the velocity (derivative) of heart rate with ovulation in menstrual cycles, providing a predictive warning signal. Similarly, an analysis of calcium signaling in the suprachiasmatic nucleus (SCN) of mice reveals that the maximum velocity of rising calcium signal aligns with locomotor activity offsets. To demonstrate the generality of derivative-transitions link, numerical simulations using a negative feedback loop model were conducted. Statistical analysis indicated that over 90% of the oscillations exhibited a correlation between maximum velocity and transition points. Consequently, the maximum velocity derived from oscillatory curves holds significant potential as an early warning signal for critical transitions.
Collapse
Affiliation(s)
- Yao Wang
- School of Mathematical Science, Soochow University, Suzhou 215006, China
- Center for Systems Biology, Soochow University, Suzhou 215006, China
| | - Yingying Dong
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Suzhou medical college of Soochow University, Suzhou 215123, China
| | - Qiaocheng Zhai
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Suzhou medical college of Soochow University, Suzhou 215123, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China
| | - Ying Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Suzhou medical college of Soochow University, Suzhou 215123, China
| | - Ling Yang
- School of Mathematical Science, Soochow University, Suzhou 215006, China
- Center for Systems Biology, Soochow University, Suzhou 215006, China
| |
Collapse
|
7
|
Hiro S, Kobayashi K, Nemoto T, Enoki R. In-phasic cytosolic-nuclear Ca 2+ rhythms in suprachiasmatic nucleus neurons. Front Neurosci 2023; 17:1323565. [PMID: 38178840 PMCID: PMC10765503 DOI: 10.3389/fnins.2023.1323565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus is the master circadian clock in mammals. SCN neurons exhibit circadian Ca2+ rhythms in the cytosol, which is thought to act as a messenger linking the transcriptional/translational feedback loop (TTFL) and physiological activities. Transcriptional regulation occurs in the nucleus in the TTFL model, and Ca2+-dependent kinase regulates the clock gene transcription. However, the Ca2+ regulatory mechanisms between cytosol and nucleus as well as the ionic origin of Ca2+ rhythms remain unclear. In the present study, we monitored circadian-timescale Ca2+ dynamics in the nucleus and cytosol of SCN neurons at the single-cell and network levels. We observed robust nuclear Ca2+ rhythm in the same phase as the cytosolic rhythm in single SCN neurons and entire regions. Neuronal firing inhibition reduced the amplitude of both nuclear and cytosolic Ca2+ rhythms, whereas blocking of Ca2+ release from the endoplasmic reticulum (ER) via ryanodine and inositol 1,4,5-trisphosphate (IP3) receptors had a minor effect on either Ca2+ rhythms. We conclude that the in-phasic circadian Ca2+ rhythms in the cytosol and nucleus are mainly driven by Ca2+ influx from the extracellular space, likely through the nuclear pore. It also raises the possibility that nuclear Ca2+ rhythms directly regulate transcription in situ.
Collapse
Affiliation(s)
- Sota Hiro
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Division of Biophotonics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi, Japan
| | - Kenta Kobayashi
- School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi, Japan
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Tomomi Nemoto
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Division of Biophotonics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi, Japan
| | - Ryosuke Enoki
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Division of Biophotonics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi, Japan
| |
Collapse
|
8
|
Chen R, Routh BN, Gaudet AD, Fonken LK. Circadian Regulation of the Neuroimmune Environment Across the Lifespan: From Brain Development to Aging. J Biol Rhythms 2023; 38:419-446. [PMID: 37357738 PMCID: PMC10475217 DOI: 10.1177/07487304231178950] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Circadian clocks confer 24-h periodicity to biological systems, to ultimately maximize energy efficiency and promote survival in a world with regular environmental light cycles. In mammals, circadian rhythms regulate myriad physiological functions, including the immune, endocrine, and central nervous systems. Within the central nervous system, specialized glial cells such as astrocytes and microglia survey and maintain the neuroimmune environment. The contributions of these neuroimmune cells to both homeostatic and pathogenic demands vary greatly across the day. Moreover, the function of these cells changes across the lifespan. In this review, we discuss circadian regulation of the neuroimmune environment across the lifespan, with a focus on microglia and astrocytes. Circadian rhythms emerge in early life concurrent with neuroimmune sculpting of brain circuits and wane late in life alongside increasing immunosenescence and neurodegeneration. Importantly, circadian dysregulation can alter immune function, which may contribute to susceptibility to neurodevelopmental and neurodegenerative diseases. In this review, we highlight circadian neuroimmune interactions across the lifespan and share evidence that circadian dysregulation within the neuroimmune system may be a critical component in human neurodevelopmental and neurodegenerative diseases.
Collapse
Affiliation(s)
- Ruizhuo Chen
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Brandy N. Routh
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| | - Andrew D. Gaudet
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
- Department of Psychology, The University of Texas at Austin, Austin, Texas
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Laura K. Fonken
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
9
|
Tsuno Y, Peng Y, Horike SI, Wang M, Matsui A, Yamagata K, Sugiyama M, Nakamura TJ, Daikoku T, Maejima T, Mieda M. In vivo recording of suprachiasmatic nucleus dynamics reveals a dominant role of arginine vasopressin neurons in circadian pacesetting. PLoS Biol 2023; 21:e3002281. [PMID: 37643163 PMCID: PMC10465001 DOI: 10.1371/journal.pbio.3002281] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 07/28/2023] [Indexed: 08/31/2023] Open
Abstract
The central circadian clock of the suprachiasmatic nucleus (SCN) is a network consisting of various types of neurons and glial cells. Individual cells have the autonomous molecular machinery of a cellular clock, but their intrinsic periods vary considerably. Here, we show that arginine vasopressin (AVP) neurons set the ensemble period of the SCN network in vivo to control the circadian behavior rhythm. Artificial lengthening of cellular periods by deleting casein kinase 1 delta (CK1δ) in the whole SCN lengthened the free-running period of behavior rhythm to an extent similar to CK1δ deletion specific to AVP neurons. However, in SCN slices, PER2::LUC reporter rhythms of these mice only partially and transiently recapitulated the period lengthening, showing a dissociation between the SCN shell and core with a period instability in the shell. In contrast, in vivo calcium rhythms of both AVP and vasoactive intestinal peptide (VIP) neurons in the SCN of freely moving mice demonstrated stably lengthened periods similar to the behavioral rhythm upon AVP neuron-specific CK1δ deletion, without changing the phase relationships between each other. Furthermore, optogenetic activation of AVP neurons acutely induced calcium increase in VIP neurons in vivo. These results indicate that AVP neurons regulate other SCN neurons, such as VIP neurons, in vivo and thus act as a primary determinant of the SCN ensemble period.
Collapse
Affiliation(s)
- Yusuke Tsuno
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yubo Peng
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Shin-ichi Horike
- Division of Integrated Omics Research, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Mohan Wang
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Ayako Matsui
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kanato Yamagata
- Child Brain Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Mizuki Sugiyama
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Takahiro J. Nakamura
- Laboratory of Animal Physiology, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Takiko Daikoku
- Division of Animal Disease Model, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Takashi Maejima
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
10
|
Makrygianni EA, Chrousos GP. Neural Progenitor Cells and the Hypothalamus. Cells 2023; 12:1822. [PMID: 37508487 PMCID: PMC10378393 DOI: 10.3390/cells12141822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 07/30/2023] Open
Abstract
Neural progenitor cells (NPCs) are multipotent neural stem cells (NSCs) capable of self-renewing and differentiating into neurons, astrocytes and oligodendrocytes. In the postnatal/adult brain, NPCs are primarily located in the subventricular zone (SVZ) of the lateral ventricles (LVs) and subgranular zone (SGZ) of the hippocampal dentate gyrus (DG). There is evidence that NPCs are also present in the postnatal/adult hypothalamus, a highly conserved brain region involved in the regulation of core homeostatic processes, such as feeding, metabolism, reproduction, neuroendocrine integration and autonomic output. In the rodent postnatal/adult hypothalamus, NPCs mainly comprise different subtypes of tanycytes lining the wall of the 3rd ventricle. In the postnatal/adult human hypothalamus, the neurogenic niche is constituted by tanycytes at the floor of the 3rd ventricle, ependymal cells and ribbon cells (showing a gap-and-ribbon organization similar to that in the SVZ), as well as suprachiasmatic cells. We speculate that in the postnatal/adult human hypothalamus, neurogenesis occurs in a highly complex, exquisitely sophisticated neurogenic niche consisting of at least four subniches; this structure has a key role in the regulation of extrahypothalamic neurogenesis, and hypothalamic and extrahypothalamic neural circuits, partly through the release of neurotransmitters, neuropeptides, extracellular vesicles (EVs) and non-coding RNAs (ncRNAs).
Collapse
Affiliation(s)
- Evanthia A Makrygianni
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
11
|
Zhang N, Yu H, Liu T, Zhou Z, Feng B, Wang Y, Qian Z, Hou X, Zou J. Bmal1 downregulation leads to diabetic cardiomyopathy by promoting Bcl2/IP3R-mediated mitochondrial Ca 2+ overload. Redox Biol 2023; 64:102788. [PMID: 37356134 DOI: 10.1016/j.redox.2023.102788] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/11/2023] [Accepted: 06/15/2023] [Indexed: 06/27/2023] Open
Abstract
Brain and muscle arnt-like protein 1 (Bmal1) is a crucial transcription factor, regulating circadian rhythm and involved in multiple heart diseases. However, it is unknown whether Bmal1 promotes diabetic cardiomyopathy (DCM) pathogenesis. The objective of this investigation was to ascertain the vital role of Bmal1 in the progression of DCM. Mice with T2D and H9c2 cardiomyoblasts exposed to high glucose and palmitic acid (HGHP) were used. Cardiomyocyte-specific knockout mouse of Bmal1 (CKB) was also generated, and cardiac Bmal1 was overexpressed in type 2 diabetes (T2D) mice using an adeno-associated virus. Bmal1 gene recombinant adenovirus was used to either knockdown or overexpress in H9c2 cardiomyoblasts. Bmal1 expression was significantly altered in diabetic mice hearts. Bmal1 downregulation in CKB and T2D mice heart accelerated cardiac hypertrophy and diastolic dysfunction, while Bmal1 overexpression ameliorated these pathological changes in DCM mice. Furthermore, DCM mice had significant mitochondrial ultrastructural defects, reactive oxygen species accumulation, and apoptosis, which could be alleviated by overexpressing Bmal1. In H9c2 cardiomyoblasts, genetic downregulation of Bmal1 or HGHP markedly decreased the binding of Bcl2 to IP3R, thus increasing Ca2+ release to mitochondria through mitochondria-associated endoplasmic reticulum membranes. Importantly, chromatin immunoprecipitation revealed Bmal1 could bind directly to the Bcl2 gene promoter region. Bmal1 overexpression augmented the Bmal1/Bcl2 binding, enhancing the inhibition of Bcl2 on IP3R activity, thus alleviating mitochondrial Ca2+ overload and subsequent cell apoptosis. These results show that Bmal1 is involved in the DCM development through Bcl2/IP3R-mediated mitochondria Ca2+ overload. Therapy targeting the circadian clock (Bmal1) can treat DCM.
Collapse
Affiliation(s)
- Nannan Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Hao Yu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Tianzi Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zihao Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bin Feng
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yao Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhiyong Qian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaofeng Hou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiangang Zou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
12
|
Ragozzino FJ, Peterson B, Karatsoreos IN, Peters JH. Circadian regulation of glutamate release pathways shapes synaptic throughput in the brainstem nucleus of the solitary tract (NTS). J Physiol 2023; 601:1881-1896. [PMID: 36975145 PMCID: PMC10192157 DOI: 10.1113/jp284370] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Abstract
Circadian regulation of autonomic reflex pathways pairs physiological function with the daily light cycle. The brainstem nucleus of the solitary tract (NTS) is a key candidate for rhythmic control of the autonomic nervous system. Here we investigated circadian regulation of NTS neurotransmission and synaptic throughput using patch-clamp electrophysiology in brainstem slices from mice. We found that spontaneous quantal glutamate release onto NTS neurons showed strong circadian rhythmicity, with the highest rate of release during the light phase and the lowest in the dark, that were sufficient to drive day/night differences in constitutive postsynaptic action potential firing. In contrast, afferent evoked action potential throughput was enhanced during the dark and diminished in the light. Afferent-driven synchronous release pathways showed a similar decrease in release probability that did not explain the enhanced synaptic throughput during the night. However, analysis of postsynaptic membrane properties revealed diurnal changes in conductance, which, when coupled with the circadian changes in glutamate release pathways, tuned synaptic throughput between the light and dark phases. These coordinated pre-/postsynaptic changes encode nuanced control over synaptic performance and pair NTS action potential firing and vagal throughput with time of day. KEY POINTS: Vagal afferent neurons relay information from peripheral organs to the brainstem nucleus of the solitary tract (NTS) to initiate autonomic reflex pathways as well as providing important controls of food intake, digestive function and energy balance. Vagally mediated reflexes and behaviours are under strong circadian regulation. Diurnal fluctuations in presynaptic vesicle release pathways and postsynaptic membrane conductances provide nuanced control over NTS action potential firing and vagal synaptic throughput. Coordinated pre-/postsynaptic changes represent a fundamental mechanism mediating daily changes in vagal afferent signalling and autonomic function.
Collapse
Affiliation(s)
- Forrest J. Ragozzino
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - BreeAnne Peterson
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Ilia N. Karatsoreos
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - James H. Peters
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
13
|
Lodovichi C, Ratto GM. Control of circadian rhythm on cortical excitability and synaptic plasticity. Front Neural Circuits 2023; 17:1099598. [PMID: 37063387 PMCID: PMC10098176 DOI: 10.3389/fncir.2023.1099598] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/09/2023] [Indexed: 04/18/2023] Open
Abstract
Living organisms navigate through a cyclic world: activity, feeding, social interactions are all organized along the periodic succession of night and day. At the cellular level, periodic activity is controlled by the molecular machinery driving the circadian regulation of cellular homeostasis. This mechanism adapts cell function to the external environment and its crucial importance is underlined by its robustness and redundancy. The cell autonomous clock regulates cell function by the circadian modulation of mTOR, a master controller of protein synthesis. Importantly, mTOR integrates the circadian modulation with synaptic activity and extracellular signals through a complex signaling network that includes the RAS-ERK pathway. The relationship between mTOR and the circadian clock is bidirectional, since mTOR can feedback on the cellular clock to shift the cycle to maintain the alignment with the environmental conditions. The mTOR and ERK pathways are crucial determinants of synaptic plasticity and function and thus it is not surprising that alterations of the circadian clock cause defective responses to environmental challenges, as witnessed by the bi-directional relationship between brain disorders and impaired circadian regulation. In physiological conditions, the feedback between the intrinsic clock and the mTOR pathway suggests that also synaptic plasticity should undergo circadian regulation.
Collapse
Affiliation(s)
- Claudia Lodovichi
- Institute of Neuroscience, Consiglio Nazionale delle Ricerche (CNR), Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
- Padova Neuroscience Center, Universitá degli Studi di Padova, Padova, Italy
| | - Gian Michele Ratto
- Institute of Neuroscience, Consiglio Nazionale delle Ricerche (CNR), Padova, Italy
- Padova Neuroscience Center, Universitá degli Studi di Padova, Padova, Italy
- National Enterprise for NanoScience and NanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore, Pisa, Italy
| |
Collapse
|
14
|
Stangherlin A. Ion dynamics and the regulation of circadian cellular physiology. Am J Physiol Cell Physiol 2023; 324:C632-C643. [PMID: 36689675 DOI: 10.1152/ajpcell.00378.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Circadian rhythms in physiology and behavior allow organisms to anticipate the daily environmental changes imposed by the rotation of our planet around its axis. Although these rhythms eventually manifest at the organismal level, a cellular basis for circadian rhythms has been demonstrated. Significant contributors to these cell-autonomous rhythms are daily cycles in gene expression and protein translation. However, recent data revealed cellular rhythms in other biological processes, including ionic currents, ion transport, and cytosolic ion abundance. Circadian rhythms in ion currents sustain circadian variation in action potential firing rate, which coordinates neuronal behavior and activity. Circadian regulation of metal ions abundance and dynamics is implicated in distinct cellular processes, from protein translation to membrane activity and osmotic homeostasis. In turn, studies showed that manipulating ion abundance affects the expression of core clock genes and proteins, suggestive of a close interplay. However, the relationship between gene expression cycles, ion dynamics, and cellular function is still poorly characterized. In this review, I will discuss the mechanisms that generate ion rhythms, the cellular functions they govern, and how they feed back to regulate the core clock machinery.
Collapse
Affiliation(s)
- Alessandra Stangherlin
- Faculty of Medicine and University Hospital Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Institute for Mitochondrial Diseases and Ageing, University of Cologne, Cologne, Germany
| |
Collapse
|
15
|
Ono D, Wang H, Hung CJ, Wang HT, Kon N, Yamanaka A, Li Y, Sugiyama T. Network-driven intracellular cAMP coordinates circadian rhythm in the suprachiasmatic nucleus. SCIENCE ADVANCES 2023; 9:eabq7032. [PMID: 36598978 PMCID: PMC11318661 DOI: 10.1126/sciadv.abq7032] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 11/23/2022] [Indexed: 06/17/2023]
Abstract
The mammalian central circadian clock, located in the suprachiasmatic nucleus (SCN), coordinates the timing of physiology and behavior to local time cues. In the SCN, second messengers, such as cAMP and Ca2+, are suggested to be involved in the input and/or output of the molecular circadian clock. However, the functional roles of second messengers and their dynamics in the SCN remain largely unclear. In the present study, we visualized the spatiotemporal patterns of circadian rhythms of second messengers and neurotransmitter release in the SCN. Here, we show that neuronal activity regulates the rhythmic release of vasoactive intestinal peptides from the SCN, which drives the circadian rhythms of intracellular cAMP in the SCN. Furthermore, optical manipulation of intracellular cAMP levels in the SCN shifts molecular and behavioral circadian rhythms. Together, our study demonstrates that intracellular cAMP is a key molecule in the organization of the SCN circadian neuronal network.
Collapse
Affiliation(s)
- Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Huan Wang
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Chi Jung Hung
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hsin-tzu Wang
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
- Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
- Department of Biological Sciences, School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Naohiro Kon
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
- Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Akihiro Yamanaka
- Chinese Institute for Brain Research (CIBR), Beijing, 102206, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Takashi Sugiyama
- Advanced Optics and Biological Engineering, Evident Corporation, Tokyo, Japan
| |
Collapse
|
16
|
Abstract
Our physiology and behavior follow precise daily programs that adapt us to the alternating opportunities and challenges of day and night. Under experimental isolation, these rhythms persist with a period of approximately one day (circadian), demonstrating their control by an internal autonomous clock. Circadian time is created at the cellular level by a transcriptional/translational feedback loop (TTFL) in which the protein products of the Period and Cryptochrome genes inhibit their own transcription. Because the accumulation of protein is slow and delayed, the system oscillates spontaneously with a period of ∼24 hours. This cell-autonomous TTFL controls cycles of gene expression in all major tissues and these cycles underpin our daily metabolic programs. In turn, our innumerable cellular clocks are coordinated by a central pacemaker, the suprachiasmatic nucleus (SCN) of the hypothalamus. When isolated in slice culture, the SCN TTFL and its dependent cycles of neural activity persist indefinitely, operating as "a clock in a dish". In vivo, SCN time is synchronized to solar time by direct innervation from specialized retinal photoreceptors. In turn, the precise circadian cycle of action potential firing signals SCN-generated time to hypothalamic and brain stem targets, which co-ordinate downstream autonomic, endocrine, and behavioral (feeding) cues to synchronize and sustain the distributed cellular clock network. Circadian time therefore pervades every level of biological organization, from molecules to society. Understanding its mechanisms offers important opportunities to mitigate the consequences of circadian disruption, so prevalent in modern societies, that arise from shiftwork, aging, and neurodegenerative diseases, not least Huntington's disease.
Collapse
Affiliation(s)
- Andrew P. Patton
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | | |
Collapse
|
17
|
El Cheikh Hussein L, Fontanaud P, Mollard P, Bonnefont X. Nested calcium dynamics support daily cell unity and diversity in the suprachiasmatic nuclei of free-behaving mice. PNAS NEXUS 2022; 1:pgac112. [PMID: 36741435 PMCID: PMC9896879 DOI: 10.1093/pnasnexus/pgac112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023]
Abstract
The suprachiasmatic nuclei (SCN) of the anterior hypothalamus host the circadian pacemaker that synchronizes mammalian rhythms with the day-night cycle. SCN neurons are intrinsically rhythmic, thanks to a conserved cell-autonomous clock mechanism. In addition, circuit-level emergent properties confer a unique degree of precision and robustness to SCN neuronal rhythmicity. However, the multicellular functional organization of the SCN is not yet fully understood. Indeed, although SCN neurons are well-coordinated, experimental evidences indicate that some neurons oscillate out of phase in SCN explants, and possibly to a larger extent in vivo. Here, to tackle this issue we used microendoscopic Ca2+ i imaging and investigated SCN rhythmicity at a single cell resolution in free-behaving mice. We found that SCN neurons in vivo exhibited fast Ca2+ i spikes superimposed upon slow changes in baseline Ca2+ i levels. Both spikes and baseline followed a time-of-day modulation in many neurons, but independently from each other. Daily rhythms in basal Ca2+ i were highly coordinated, while spike activity from the same neurons peaked at multiple times of the light cycle, and unveiled clock-independent coactivity in neuron subsets. Hence, fast Ca2+ i spikes and slow changes in baseline Ca2+ i levels highlighted how multiple individual activity patterns could articulate within the temporal unity of the SCN cell network in vivo, and provided support for a multiplex neuronal code in the circadian pacemaker.
Collapse
Affiliation(s)
- Lama El Cheikh Hussein
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, IGF, 141 Rue de la Cardonille, F-34094 Montpellier, Cedex 5, France,BioCampus Montpellier, Université de Montpellier, CNRS, INSERM, 141 Rue de la Cardonille, F-34094 Montpellier, Cedex 5, France
| | - Pierre Fontanaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, IGF, 141 Rue de la Cardonille, F-34094 Montpellier, Cedex 5, France,BioCampus Montpellier, Université de Montpellier, CNRS, INSERM, 141 Rue de la Cardonille, F-34094 Montpellier, Cedex 5, France
| | - Patrice Mollard
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, IGF, 141 Rue de la Cardonille, F-34094 Montpellier, Cedex 5, France,BioCampus Montpellier, Université de Montpellier, CNRS, INSERM, 141 Rue de la Cardonille, F-34094 Montpellier, Cedex 5, France
| | | |
Collapse
|
18
|
Morioka E, Kasuga Y, Kanda Y, Moritama S, Koizumi H, Yoshikawa T, Miura N, Ikeda M, Higashida H, Holmes TC, Ikeda M. Mitochondrial LETM1 drives ionic and molecular clock rhythms in circadian pacemaker neurons. Cell Rep 2022; 39:110787. [PMID: 35545046 DOI: 10.1016/j.celrep.2022.110787] [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] [Received: 01/13/2021] [Revised: 03/14/2022] [Accepted: 04/14/2022] [Indexed: 11/18/2022] Open
Abstract
The mechanisms that generate robust ionic oscillation in circadian pacemaker neurons are under investigation. Here, we demonstrate critical functions of the mitochondrial cation antiporter leucine zipper-EF-hand-containing transmembrane protein 1 (LETM1), which exchanges K+/H+ in Drosophila and Ca2+/H+ in mammals, in circadian pacemaker neurons. Letm1 knockdown in Drosophila pacemaker neurons reduced circadian cytosolic H+ rhythms and prolonged nuclear PERIOD/TIMELESS expression rhythms and locomotor activity rhythms. In rat pacemaker neurons in the hypothalamic suprachiasmatic nucleus (SCN), circadian rhythms in cytosolic Ca2+ and Bmal1 transcription were dampened by Letm1 knockdown. Mitochondrial Ca2+ uptake peaks late during the day were also observed in rat SCN neurons following photolytic elevation of cytosolic Ca2+. Since cation transport by LETM1 is coupled to mitochondrial energy synthesis, we propose that LETM1 integrates metabolic, ionic, and molecular clock rhythms in the central clock system in both invertebrates and vertebrates.
Collapse
Affiliation(s)
- Eri Morioka
- Graduate School of Science and Engineering, University of Toyama, Gofuku, Toyama 930-8555, Japan
| | - Yusuke Kasuga
- Graduate School of Science and Engineering, University of Toyama, Gofuku, Toyama 930-8555, Japan
| | - Yuzuki Kanda
- Graduate School of Science and Engineering, University of Toyama, Gofuku, Toyama 930-8555, Japan
| | - Saki Moritama
- Graduate School of Science and Engineering, University of Toyama, Gofuku, Toyama 930-8555, Japan
| | - Hayato Koizumi
- Graduate School of Innovative Life Science, University of Toyama, Gofuku, Toyama 930-8555, Japan
| | - Tomoko Yoshikawa
- Organization for International Education and Exchange, University of Toyama, Toyama 930-8555, Japan
| | - Nobuhiko Miura
- Department of Health Medicine, Yokohama University of Pharmacy, Yokohama, Kanagawa 245-0061, Japan
| | - Masaaki Ikeda
- Department of Physiology, Faculty of Medicine, Saitama Medical University, Saitama 350-0495, Japan
| | - Haruhiro Higashida
- Research Center for Child Mental Development, Kanazawa University, Ishikawa 920-8640, Japan
| | - Todd C Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Masayuki Ikeda
- Graduate School of Innovative Life Science, University of Toyama, Gofuku, Toyama 930-8555, Japan; Organization for International Education and Exchange, University of Toyama, Toyama 930-8555, Japan.
| |
Collapse
|
19
|
Liang X, Holy TE, Taghert PH. Circadian pacemaker neurons display cophasic rhythms in basal calcium level and in fast calcium fluctuations. Proc Natl Acad Sci U S A 2022; 119:e2109969119. [PMID: 35446620 PMCID: PMC9173584 DOI: 10.1073/pnas.2109969119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 03/17/2022] [Indexed: 12/02/2022] Open
Abstract
Circadian pacemaker neurons in the Drosophila brain display daily rhythms in the levels of intracellular calcium. These calcium rhythms are driven by molecular clocks and are required for normal circadian behavior. To study their biological basis, we employed genetic manipulations in conjunction with improved methods of in vivo light-sheet microscopy to measure calcium dynamics in individual pacemaker neurons over complete 24-h durations at sampling frequencies as high as 5 Hz. This technological advance unexpectedly revealed cophasic daily rhythms in basal calcium levels and in high-frequency calcium fluctuations. Further, we found that the rhythms of basal calcium levels and of fast calcium fluctuations reflect the activities of two proteins that mediate distinct forms of calcium fluxes. One is the inositol trisphosphate receptor (ITPR), a channel that mediates calcium fluxes from internal endoplasmic reticulum calcium stores, and the other is a T-type voltage-gated calcium channel, which mediates extracellular calcium influx. These results suggest that Drosophila molecular clocks regulate ITPR and T-type channels to generate two distinct but coupled rhythms in basal calcium and in fast calcium fluctuations. We propose that both internal and external calcium fluxes are essential for circadian pacemaker neurons to provide rhythmic outputs and thereby, regulate the activities of downstream brain centers.
Collapse
Affiliation(s)
- Xitong Liang
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO 63110
| | - Timothy E. Holy
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO 63110
| | - Paul H. Taghert
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO 63110
| |
Collapse
|
20
|
Morris EL, Patton AP, Chesham JE, Crisp A, Adamson A, Hastings MH. Single-cell transcriptomics of suprachiasmatic nuclei reveal a Prokineticin-driven circadian network. EMBO J 2021; 40:e108614. [PMID: 34487375 PMCID: PMC8521297 DOI: 10.15252/embj.2021108614] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 11/22/2022] Open
Abstract
Circadian rhythms in mammals are governed by the hypothalamic suprachiasmatic nucleus (SCN), in which 20,000 clock cells are connected together into a powerful time‐keeping network. In the absence of network‐level cellular interactions, the SCN fails as a clock. The topology and specific roles of its distinct cell populations (nodes) that direct network functions are, however, not understood. To characterise its component cells and network structure, we conducted single‐cell sequencing of SCN organotypic slices and identified eleven distinct neuronal sub‐populations across circadian day and night. We defined neuropeptidergic signalling axes between these nodes, and built neuropeptide‐specific network topologies. This revealed their temporal plasticity, being up‐regulated in circadian day. Through intersectional genetics and real‐time imaging, we interrogated the contribution of the Prok2‐ProkR2 neuropeptidergic axis to network‐wide time‐keeping. We showed that Prok2‐ProkR2 signalling acts as a key regulator of SCN period and rhythmicity and contributes to defining the network‐level properties that underpin robust circadian co‐ordination. These results highlight the diverse and distinct contributions of neuropeptide‐modulated communication of temporal information across the SCN.
Collapse
Affiliation(s)
- Emma L Morris
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Andrew P Patton
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Johanna E Chesham
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Alastair Crisp
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Antony Adamson
- The Genome Editing Unit, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Michael H Hastings
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK
| |
Collapse
|
21
|
Plante AE, Rao VP, Rizzo MA, Meredith AL. Comparative Ca 2+ channel contributions to intracellular Ca 2+ levels in the circadian clock. BIOPHYSICAL REPORTS 2021; 1:100005. [PMID: 35330949 PMCID: PMC8942421 DOI: 10.1016/j.bpr.2021.100005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/08/2021] [Indexed: 11/22/2022]
Abstract
Circadian rhythms in mammals are coordinated by the central clock in the brain, located in the suprachiasmatic nucleus (SCN). Multiple molecular and cellular signals display a circadian variation within SCN neurons, including intracellular Ca2+, but the mechanisms are not definitively established. SCN cytosolic Ca2+ levels exhibit a peak during the day, when both action potential firing and Ca2+ channel activity are increased, and are decreased at night, correlating with a reduction in firing rate. In this study, we employ a single-color fluorescence anisotropy reporter (FLARE), Venus FLARE-Cameleon, and polarization inverted selective-plane illumination microscopy to measure rhythmic changes in cytosolic Ca2+ in SCN neurons. Using this technique, the Ca2+ channel subtypes contributing to intracellular Ca2+ at the peak and trough of the circadian cycle were assessed using a pharmacological approach with Ca2+ channel inhibitors. Peak (218 ± 16 nM) and trough (172 ± 13 nM) Ca2+ levels were quantified, indicating a 1.3-fold circadian variance in Ca2+ concentration. Inhibition of ryanodine-receptor-mediated Ca2+ release produced a larger relative decrease in cytosolic Ca2+ at both time points compared to voltage-gated Ca2+channels. These results support the hypothesis that circadian Ca2+ rhythms in SCN neurons are predominantly driven by intracellular Ca2+ channels, although not exclusively so. The study provides a foundation for future experiments to probe Ca2+ signaling in a dynamic biological context using FLAREs.
Collapse
Affiliation(s)
- Amber E. Plante
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Vishnu P. Rao
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Megan A. Rizzo
- 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
|
22
|
|
23
|
Abdel-Rahman EA, Hosseiny S, Aaliya A, Adel M, Yasseen B, Al-Okda A, Radwan Y, Saber SH, Elkholy N, Elhanafy E, Walker EE, Zuniga-Hertz JP, Patel HH, Griffiths HR, Ali SS. Sleep/wake calcium dynamics, respiratory function, and ROS production in cardiac mitochondria. J Adv Res 2021; 31:35-47. [PMID: 34194831 PMCID: PMC8240107 DOI: 10.1016/j.jare.2021.01.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/24/2020] [Accepted: 01/07/2021] [Indexed: 12/22/2022] Open
Abstract
Introduction Incidents of myocardial infarction and sudden cardiac arrest vary with time of the day, but the mechanism for this effect is not clear. We hypothesized that diurnal changes in the ability of cardiac mitochondria to control calcium homeostasis dictate vulnerability to cardiovascular events. Objectives Here we investigate mitochondrial calcium dynamics, respiratory function, and reactive oxygen species (ROS) production in mouse heart during different phases of wake versus sleep periods. Methods We assessed time-of-the-day dependence of calcium retention capacity of isolated heart mitochondria from young male C57BL6 mice. Rhythmicity of mitochondrial-dependent oxygen consumption, ROS production and transmembrane potential in homogenates were explored using the Oroboros O2k Station equipped with a fluorescence detection module. Changes in expression of essential clock and calcium dynamics genes/proteins were also determined at sleep versus wake time points. Results Our results demonstrate that cardiac mitochondria exhibit higher calcium retention capacity and higher rates of calcium uptake during sleep period. This was associated with higher expression of clock gene Bmal1, lower expression of per2, greater expression of MICU1 gene (mitochondrial calcium uptake 1), and lower expression of the mitochondrial transition pore regulator gene cyclophilin D. Protein levels of mitochondrial calcium uniporter (MCU), MICU2, and sodium/calcium exchanger (NCLX) were also higher at sleep onset relative to wake period. While complex I and II-dependent oxygen utilization and transmembrane potential of cardiac mitochondria were lower during sleep, ROS production was increased presumably due to mitochondrial calcium sequestration. Conclusions Taken together, our results indicate that retaining mitochondrial calcium in the heart during sleep dissipates membrane potential, slows respiratory activities, and increases ROS levels, which may contribute to increased vulnerability to cardiac stress during sleep-wake transition. This pronounced daily oscillations in mitochondrial functions pertaining to stress vulnerability may at least in part explain diurnal prevalence of cardiac pathologies.
Collapse
Affiliation(s)
- Engy A. Abdel-Rahman
- Center for Aging and Associated Diseases, Zewail City of Science and Technology, Giza, Egypt
- 57357 Children's Cancer Hospital, Basic Research Department, Cairo, Egypt
- Department of Pharmacology, Faculty of Medicine, Assuit University, Assuit, Egypt
| | - Salma Hosseiny
- Center for Aging and Associated Diseases, Zewail City of Science and Technology, Giza, Egypt
| | - Abdullah Aaliya
- Center for Aging and Associated Diseases, Zewail City of Science and Technology, Giza, Egypt
| | - Mohamed Adel
- Center for Aging and Associated Diseases, Zewail City of Science and Technology, Giza, Egypt
| | - Basma Yasseen
- Center for Aging and Associated Diseases, Zewail City of Science and Technology, Giza, Egypt
- 57357 Children's Cancer Hospital, Basic Research Department, Cairo, Egypt
| | - Abdelrahman Al-Okda
- Center for Aging and Associated Diseases, Zewail City of Science and Technology, Giza, Egypt
| | - Yasmine Radwan
- Center for Aging and Associated Diseases, Zewail City of Science and Technology, Giza, Egypt
| | - Saber H. Saber
- Center for Aging and Associated Diseases, Zewail City of Science and Technology, Giza, Egypt
| | - Nada Elkholy
- Center for Aging and Associated Diseases, Zewail City of Science and Technology, Giza, Egypt
| | - Eslam Elhanafy
- Center for Aging and Associated Diseases, Zewail City of Science and Technology, Giza, Egypt
| | - Emily E. Walker
- Veterans Affairs San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA 92161, USA
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Juan P. Zuniga-Hertz
- Veterans Affairs San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA 92161, USA
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hemal H. Patel
- Veterans Affairs San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA 92161, USA
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - Sameh S. Ali
- Center for Aging and Associated Diseases, Zewail City of Science and Technology, Giza, Egypt
- 57357 Children's Cancer Hospital, Basic Research Department, Cairo, Egypt
| |
Collapse
|
24
|
Cavieres-Lepe J, Ewer J. Reciprocal Relationship Between Calcium Signaling and Circadian Clocks: Implications for Calcium Homeostasis, Clock Function, and Therapeutics. Front Mol Neurosci 2021; 14:666673. [PMID: 34045944 PMCID: PMC8144308 DOI: 10.3389/fnmol.2021.666673] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/09/2021] [Indexed: 12/03/2022] Open
Abstract
In animals, circadian clocks impose a daily rhythmicity to many behaviors and physiological processes. At the molecular level, circadian rhythms are driven by intracellular transcriptional/translational feedback loops (TTFL). Interestingly, emerging evidence indicates that they can also be modulated by multiple signaling pathways. Among these, Ca2+ signaling plays a key role in regulating the molecular rhythms of clock genes and of the resulting circadian behavior. In addition, the application of in vivo imaging approaches has revealed that Ca2+ is fundamental to the synchronization of the neuronal networks that make up circadian pacemakers. Conversely, the activity of circadian clocks may influence Ca2+ signaling. For instance, several genes that encode Ca2+ channels and Ca2+-binding proteins display a rhythmic expression, and a disruption of this cycling affects circadian function, underscoring their reciprocal relationship. Here, we review recent advances in our understanding of how Ca2+ signaling both modulates and is modulated by circadian clocks, focusing on the regulatory mechanisms described in Drosophila and mice. In particular, we examine findings related to the oscillations in intracellular Ca2+ levels in circadian pacemakers and how they are regulated by canonical clock genes, neuropeptides, and light stimuli. In addition, we discuss how Ca2+ rhythms and their associated signaling pathways modulate clock gene expression at the transcriptional and post-translational levels. We also review evidence based on transcriptomic analyzes that suggests that mammalian Ca2+ channels and transporters (e.g., ryanodine receptor, ip3r, serca, L- and T-type Ca2+ channels) as well as Ca2+-binding proteins (e.g., camk, cask, and calcineurin) show rhythmic expression in the central brain clock and in peripheral tissues such as the heart and skeletal muscles. Finally, we discuss how the discovery that Ca2+ signaling is regulated by the circadian clock could influence the efficacy of pharmacotherapy and the outcomes of clinical interventions.
Collapse
Affiliation(s)
- Javier Cavieres-Lepe
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - John Ewer
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
25
|
Buijink MR, Michel S. A multi-level assessment of the bidirectional relationship between aging and the circadian clock. J Neurochem 2021; 157:73-94. [PMID: 33370457 PMCID: PMC8048448 DOI: 10.1111/jnc.15286] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022]
Abstract
The daily temporal order of physiological processes and behavior contribute to the wellbeing of many organisms including humans. The central circadian clock, which coordinates the timing within our body, is located in the suprachiasmatic nucleus (SCN) of the hypothalamus. Like in other parts of the brain, aging impairs the SCN function, which in turn promotes the development and progression of aging-related diseases. We here review the impact of aging on the different levels of the circadian clock machinery-from molecules to organs-with a focus on the role of the SCN. We find that the molecular clock is less effected by aging compared to other cellular components of the clock. Proper rhythmic regulation of intracellular signaling, ion channels and neuronal excitability of SCN neurons are greatly disturbed in aging. This suggests a disconnection between the molecular clock and the electrophysiology of these cells. The neuronal network of the SCN is able to compensate for some of these cellular deficits. However, it still results in a clear reduction in the amplitude of the SCN electrical rhythm, suggesting a weakening of the output timing signal. Consequently, other brain areas and organs not only show aging-related deficits in their own local clocks, but also receive a weaker systemic timing signal. The negative spiral completes with the weakening of positive feedback from the periphery to the SCN. Consequently, chronotherapeutic interventions should aim at strengthening overall synchrony in the circadian system using life-style and/or pharmacological approaches.
Collapse
Affiliation(s)
- M. Renate Buijink
- Department of Cellular and Chemical BiologyLaboratory for NeurophysiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Stephan Michel
- Department of Cellular and Chemical BiologyLaboratory for NeurophysiologyLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
26
|
Kon N, Wang HT, Kato YS, Uemoto K, Kawamoto N, Kawasaki K, Enoki R, Kurosawa G, Nakane T, Sugiyama Y, Tagashira H, Endo M, Iwasaki H, Iwamoto T, Kume K, Fukada Y. Na +/Ca 2+ exchanger mediates cold Ca 2+ signaling conserved for temperature-compensated circadian rhythms. SCIENCE ADVANCES 2021; 7:7/18/eabe8132. [PMID: 33931447 PMCID: PMC8087402 DOI: 10.1126/sciadv.abe8132] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 03/11/2021] [Indexed: 05/25/2023]
Abstract
Circadian rhythms are based on biochemical oscillations generated by clock genes/proteins, which independently evolved in animals, fungi, plants, and cyanobacteria. Temperature compensation of the oscillation speed is a common feature of the circadian clocks, but the evolutionary-conserved mechanism has been unclear. Here, we show that Na+/Ca2+ exchanger (NCX) mediates cold-responsive Ca2+ signaling important for the temperature-compensated oscillation in mammalian cells. In response to temperature decrease, NCX elevates intracellular Ca2+, which activates Ca2+/calmodulin-dependent protein kinase II and accelerates transcriptional oscillations of clock genes. The cold-responsive Ca2+ signaling is conserved among mice, Drosophila, and Arabidopsis The mammalian cellular rhythms and Drosophila behavioral rhythms were severely attenuated by NCX inhibition, indicating essential roles of NCX in both temperature compensation and autonomous oscillation. NCX also contributes to the temperature-compensated transcriptional rhythms in cyanobacterial clock. Our results suggest that NCX-mediated Ca2+ signaling is a common mechanism underlying temperature-compensated circadian rhythms both in eukaryotes and prokaryotes.
Collapse
Affiliation(s)
- Naohiro Kon
- Department of Biological Sciences, School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hsin-Tzu Wang
- Department of Biological Sciences, School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yoshiaki S Kato
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Kyouhei Uemoto
- Graduate School of Biological Science, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Naohiro Kawamoto
- Department of Electrical Engineering and Bioscience, Waseda University, Tokyo 162-8480, Japan
| | - Koji Kawasaki
- Department of Electrical Engineering and Bioscience, Waseda University, Tokyo 162-8480, Japan
| | - Ryosuke Enoki
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki, Aichi 444-8787, Japan
- Division of Biophotonics, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki, Aichi 444-8787, Japan
| | | | - Tatsuto Nakane
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, Kagawa 761-0795, Japan
| | - Yasunori Sugiyama
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, Kagawa 761-0795, Japan
| | - Hideaki Tagashira
- Department of Pharmacology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | - Motomu Endo
- Graduate School of Biological Science, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
| | - Hideo Iwasaki
- Department of Electrical Engineering and Bioscience, Waseda University, Tokyo 162-8480, Japan
| | - Takahiro Iwamoto
- Department of Pharmacology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan.
| | - Kazuhiko Kume
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Yoshitaka Fukada
- Department of Biological Sciences, School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
27
|
Spencer C, Tripp E, Fu F, Pauls S. Evolutionary Constraints on Connectivity Patterns in the Mammalian Suprachiasmatic Nucleus. FRONTIERS IN NETWORK PHYSIOLOGY 2021; 1:716883. [PMID: 36925572 PMCID: PMC10013059 DOI: 10.3389/fnetp.2021.716883] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/26/2021] [Indexed: 12/22/2022]
Abstract
The mammalian suprachiasmatic nucleus (SCN) comprises about 20,000 interconnected oscillatory neurons that create and maintain a robust circadian signal which matches to external light cues. Here, we use an evolutionary game theoretic framework to explore how evolutionary constraints can influence the synchronization of the system under various assumptions on the connection topology, contributing to the understanding of the structure of interneuron connectivity. Our basic model represents the SCN as a network of agents each with two properties-a phase and a flag that determines if it communicates with its neighbors or not. Communication comes at a cost to the agent, but synchronization of phases with its neighbors bears a benefit. Earlier work shows that when we have "all-to-all" connectivity, where every agent potentially communicates with every other agent, there is often a simple trade-off that leads to complete communication and synchronization of the system: the benefit must be greater than twice the cost. This trade-off for all-to-all connectivity gives us a baseline to compare to when looking at other topologies. Using simulations, we compare three plausible topologies to the all-to-all case, finding that convergence to synchronous dynamics occurs in all considered topologies under similar benefit and cost trade-offs. Consequently, sparser, less biologically costly topologies are reasonable evolutionary outcomes for organisms that develop a synchronizable oscillatory network. Our simulations also shed light on constraints imposed by the time scale on which we observe the SCN to arise in mammals. We find two conditions that allow for a synchronizable system to arise in relatively few generations. First, the benefits of connectivity must outweigh the cost of facilitating the connectivity in the network. Second, the game at the core of the model needs to be more cooperative than antagonistic games such as the Prisoner's Dilemma. These results again imply that evolutionary pressure may have driven the system towards sparser topologies, as they are less costly to create and maintain. Last, our simulations indicate that models based on the mutualism game fare the best in uptake of communication and synchronization compared to more antagonistic games such as the Prisoner's Dilemma.
Collapse
Affiliation(s)
- Connor Spencer
- Department of Mathematics, Dartmouth College, Hanover, NH, United States
| | - Elizabeth Tripp
- Department of Mathematics, Sacred Heart University, Fairfield, CT, United States
| | - Feng Fu
- Department of Mathematics, Dartmouth College, Hanover, NH, United States.,Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
| | - Scott Pauls
- Department of Mathematics, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
28
|
Aten S, Kalidindi A, Yoon H, Rumbaugh G, Hoyt KR, Obrietan K. SynGAP is expressed in the murine suprachiasmatic nucleus and regulates circadian-gated locomotor activity and light-entrainment capacity. Eur J Neurosci 2020; 53:732-749. [PMID: 33174316 DOI: 10.1111/ejn.15043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/15/2022]
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus functions as the master circadian clock. The phasing of the SCN oscillator is locked to the daily solar cycle, and an intracellular signaling cassette from the small GTPase Ras to the p44/42 mitogen-activated protein kinase (ERK/MAPK) pathway is central to this entrainment process. Here, we analyzed the expression and function of SynGAP-a GTPase-activating protein that serves as a negative regulator of Ras signaling-within the murine SCN. Using a combination of immunohistochemical and Western blotting approaches, we show that SynGAP is broadly expressed throughout the SCN. In addition, temporal profiling assays revealed that SynGAP expression is regulated over the circadian cycle, with peak expression occurring during the circadian night. Further, time-of-day-gated expression of SynGAP was not observed in clock arrhythmic BMAL1 null mice, indicating that the daily oscillation in SynGAP is driven by the inherent circadian timing mechanism. We also show that SynGAP phosphorylation at serine 1138-an event that has been found to modulate its functional efficacy-is regulated by clock time and is responsive to photic input. Finally, circadian phenotypic analysis of Syngap1 heterozygous mice revealed enhanced locomotor activity, increased sensitivity to light-evoked clock entrainment, and elevated levels of light-evoked MAPK activity, which is consistent with the role of SynGAP as a negative regulator of MAPK signaling. These findings reveal that SynGAP functions as a modulator of SCN clock entrainment, an effect that may contribute to sleep and circadian abnormalities observed in patients with SYNGAP1 gene mutations.
Collapse
Affiliation(s)
- Sydney Aten
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Anisha Kalidindi
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Hyojung Yoon
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Gavin Rumbaugh
- Scripps Research, Department of Neuroscience, Jupiter, FL, USA.,Scripps Research, Department of Molecular Medicine, Jupiter, FL, USA
| | - Kari R Hoyt
- Division of Pharmaceutics and Pharmacology, Ohio State University, Columbus, OH, USA
| | - Karl Obrietan
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| |
Collapse
|
29
|
Said R, Lobanova L, Papagerakis S, Papagerakis P. Calcium Sets the Clock in Ameloblasts. Front Physiol 2020; 11:920. [PMID: 32848861 PMCID: PMC7411184 DOI: 10.3389/fphys.2020.00920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/09/2020] [Indexed: 01/22/2023] Open
Abstract
Background Stromal interaction molecule 1 (STIM1) is one of the main components of the store operated Ca2+ entry (SOCE) signaling pathway. Individuals with mutated STIM1 present severely hypomineralized enamel characterized as amelogenesis imperfecta (AI) but the downstream molecular mechanisms involved remain unclear. Circadian clock signaling plays a key role in regulating the enamel thickness and mineralization, but the effects of STIM1-mediated AI on circadian clock are unknown. Objectives The aim of this study is to examine the potential links between SOCE and the circadian clock during amelogenesis. Methods We have generated mice with ameloblast-specific deletion of Stim1 (Stim1fl/fl/Amelx-iCre+/+, Stim1 cKO) and analyzed circadian gene expression profile in Stim1 cKO compared to control (Stim1fl/fl/Amelx-iCre–/–) using ameloblast micro-dissection and RNA micro-array of 84 circadian genes. Expression level changes were validated by qRT-PCR and immunohistochemistry. Results Stim1 deletion has resulted in significant upregulation of the core circadian activator gene Brain and Muscle Aryl Hydrocarbon Receptor Nuclear Translocation 1 (Bmal1) and downregulation of the circadian inhibitor Period 2 (Per2). Our analyses also revealed that SOCE disruption results in dysregulation of two additional circadian regulators; p38α mitogen-activated protein kinase (MAPK14) and transforming growth factor-beta1 (TGF-β1). Both MAPK14 and TGF-β1 pathways are known to play major roles in enamel secretion and their dysregulation has been previously implicated in the development of AI phenotype. Conclusion These data indicate that disruption of SOCE significantly affects the ameloblasts molecular circadian clock, suggesting that alteration of the circadian clock may be partly involved in the development of STIM1-mediated AI.
Collapse
Affiliation(s)
- Raed Said
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada.,College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Liubov Lobanova
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Silvana Papagerakis
- Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Petros Papagerakis
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada.,College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
30
|
Scrima R, Cela O, Agriesti F, Piccoli C, Tataranni T, Pacelli C, Mazzoccoli G, Capitanio N. Mitochondrial calcium drives clock gene-dependent activation of pyruvate dehydrogenase and of oxidative phosphorylation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118815. [PMID: 32763264 DOI: 10.1016/j.bbamcr.2020.118815] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/27/2020] [Accepted: 08/03/2020] [Indexed: 12/17/2022]
Abstract
Regulation of metabolism is emerging as a major output of circadian clock circuitry in mammals. Accordingly, mitochondrial oxidative metabolism undergoes both in vivo and in vitro daily oscillatory activities. In a previous study we showed that both glycolysis and mitochondrial oxygen consumption display a similar time-resolved rhythmic activity in synchronized HepG2 cell cultures, which translates in overall bioenergetic changes as here documented by measurement of the ATP level. Treatment of synchronized cells with specific metabolic inhibitors unveiled pyruvate as a major source of reducing equivalents to the respiratory chain with its oxidation driven by the rhythmic (de)phosphorylation of pyruvate dehydrogenase. Further investigation enabled to causally link the autonomous cadenced mitochondrial respiration to a synchronous increase of the mitochondrial Ca2+. The rhythmic change of the mitochondrial respiration was dampened by inhibitors of the mitochondrial Ca2+ uniporter as well as of the ryanodine receptor Ca2+ channel or the ADPR cyclase, indicating that the mitochondrial Ca2+ influx originated from the ER store, likely at contact sites with the mitochondrial compartment. Notably, blockage of the mitochondrial Ca2+ influx resulted in deregulation of the expression of canonical clock genes such as BMALl1, CLOCK, NR1D1. All together our findings unveil a hitherto unexplored function of Ca2+-mediated signaling in time keeping the mitochondrial metabolism and in its feed-back modulation of the circadian clockwork.
Collapse
Affiliation(s)
- Rosella Scrima
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| | - Olga Cela
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| | - Francesca Agriesti
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy.
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| | - Tiziana Tataranni
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy.
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| | - Gianluigi Mazzoccoli
- Department of Medical Sciences and Chronobiology Laboratory, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, (FG), Italy.
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| |
Collapse
|
31
|
Hartsock MJ, Spencer RL. Memory and the circadian system: Identifying candidate mechanisms by which local clocks in the brain may regulate synaptic plasticity. Neurosci Biobehav Rev 2020; 118:134-162. [PMID: 32712278 DOI: 10.1016/j.neubiorev.2020.07.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 07/14/2020] [Accepted: 07/18/2020] [Indexed: 12/11/2022]
Abstract
The circadian system is an endogenous biological network responsible for coordinating near-24-h cycles in behavior and physiology with daily timing cues from the external environment. In this review, we explore how the circadian system regulates memory formation, retention, and recall. Circadian rhythms in these memory processes may arise through several endogenous pathways, and recent work highlights the importance of genetic timekeepers found locally within tissues, called local clocks. We evaluate the circadian memory literature for evidence of local clock involvement in memory, identifying potential nodes for direct interactions between local clock components and mechanisms of synaptic plasticity. Our discussion illustrates how local clocks may pervasively modulate neuronal plastic capacity, a phenomenon that we designate here as circadian metaplasticity. We suggest that this function of local clocks supports the temporal optimization of memory processes, illuminating the potential for circadian therapeutic strategies in the prevention and treatment of memory impairment.
Collapse
Affiliation(s)
- Matthew J Hartsock
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado 80309, United States.
| | - Robert L Spencer
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado 80309, United States.
| |
Collapse
|
32
|
Patton AP, Edwards MD, Smyllie NJ, Hamnett R, Chesham JE, Brancaccio M, Maywood ES, Hastings MH. The VIP-VPAC2 neuropeptidergic axis is a cellular pacemaking hub of the suprachiasmatic nucleus circadian circuit. Nat Commun 2020; 11:3394. [PMID: 32636383 PMCID: PMC7341843 DOI: 10.1038/s41467-020-17110-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 06/05/2020] [Indexed: 12/01/2022] Open
Abstract
The hypothalamic suprachiasmatic nuclei (SCN) are the principal mammalian circadian timekeeper, co-ordinating organism-wide daily and seasonal rhythms. To achieve this, cell-autonomous circadian timing by the ~20,000 SCN cells is welded into a tight circuit-wide ensemble oscillation. This creates essential, network-level emergent properties of precise, high-amplitude oscillation with tightly defined ensemble period and phase. Although synchronised, regional cell groups exhibit differentially phased activity, creating stereotypical spatiotemporal circadian waves of cellular activation across the circuit. The cellular circuit pacemaking components that generate these critical emergent properties are unknown. Using intersectional genetics and real-time imaging, we show that SCN cells expressing vasoactive intestinal polypeptide (VIP) or its cognate receptor, VPAC2, are neurochemically and electrophysiologically distinct, but together they control de novo rhythmicity, setting ensemble period and phase with circuit-level spatiotemporal complexity. The VIP/VPAC2 cellular axis is therefore a neurochemically and topologically specific pacemaker hub that determines the emergent properties of the SCN timekeeper. Circadian activity modulation in the suprachiasmatic nucleus (SCN) is a network-level emergent property that requires neuropeptide VIP signaling, yet the precise cellular mechanisms are unknown. Patton et al. show that cells expressing VIP or its receptor VPAC2 together determine these emergent properties of the SCN.
Collapse
Affiliation(s)
- Andrew P Patton
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Mathew D Edwards
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK.,UCL Sainsbury Wellcome Centre for Neural Circuits and Behaviour, London, UK
| | - Nicola J Smyllie
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Ryan Hamnett
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK.,Department of Neurosurgery, Stanford University, Stanford, USA
| | - Johanna E Chesham
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Marco Brancaccio
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK.,Department of Brain Sciences, UK Dementia Research Institute, Imperial College London, London, UK
| | - Elizabeth S Maywood
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Michael H Hastings
- MRC Laboratory of Molecular Biology, Francis Crick Ave., Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK.
| |
Collapse
|
33
|
Coupling Neuropeptide Levels to Structural Plasticity in Drosophila Clock Neurons. Curr Biol 2020; 30:3154-3166.e4. [PMID: 32619484 DOI: 10.1016/j.cub.2020.06.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/05/2020] [Accepted: 06/02/2020] [Indexed: 12/24/2022]
Abstract
We have previously reported that pigment dispersing factor (PDF) neurons, which are essential in the control of rest-activity cycles in Drosophila, undergo circadian remodeling of their axonal projections, a phenomenon called circadian structural plasticity. Axonal arborizations display higher complexity during the day and become simpler at night, and this remodeling involves changes in the degree of connectivity. This phenomenon depends on the clock present within the ventrolateral neurons (LNvs) as well as in glia. In this work, we characterize in detail the contribution of the PDF neuropeptide to structural plasticity at different times across the day. Using diverse genetic strategies to temporally restrict its downregulation, we demonstrate that even subtle alterations to PDF cycling at the dorsal protocerebrum correlate with impaired remodeling, underscoring its relevance for the characteristic morning spread; PDF released from the small LNvs (sLNvs) and the large LNvs (lLNvs) contribute to the process. Moreover, forced depolarization recruits activity-dependent mechanisms to mediate growth only at night, overcoming the restriction imposed by the clock on membrane excitability. Interestingly, the active process of terminal remodeling requires PDF receptor (PDFR) signaling acting locally through the cyclic-nucleotide-gated channel ion channel subunit A (CNGA). Thus, clock-dependent PDF signaling shapes the connectivity of these essential clock neurons on daily basis.
Collapse
|
34
|
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]
|
35
|
Popović N, Morales-Delgado N, Vidal Mena D, Alonso A, Pascual Martínez M, Caballero Bleda M, Popović M. Verapamil and Alzheimer's Disease: Past, Present, and Future. Front Pharmacol 2020; 11:562. [PMID: 32431612 PMCID: PMC7214748 DOI: 10.3389/fphar.2020.00562] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/14/2020] [Indexed: 12/18/2022] Open
Abstract
Verapamil is a phenylalkylamine class calcium channel blocker that for half a century has been used for the treatment of cardiovascular diseases. Nowadays, verapamil is also considered as a drug option for the treatment of several neurological and psychiatric disorders, such as cluster headache, bipolar disorders, epilepsy, and neurodegenerative diseases. Here, we review insights into the potential preventive and therapeutic role of verapamil on Alzheimer’s disease (AD) based on limited experimental and clinical data. Pharmacological studies have shown that verapamil has a wide therapeutic spectrum, including antihypertensive, anti-inflammatory, and antioxidative effects, regulation of the blood-brain barrier function, due to its effect on P-glycoprotein, as well as adjustment of cellular calcium homeostasis, which may result in the delay of AD onset or ameliorate the symptoms of patients. However, the majority of the AD individuals are on polypharmacotherapy, and the interactions between verapamil and other drugs need to be considered. Therefore, for an appropriate and successful AD treatment, a personalized approach is more than necessary. A well-known narrow pharmacological window of verapamil efficacy may hinder this approach. It is therefore important to note that the verapamil efficacy may be conditioned by different factors. The onset, grade, and brain distribution of AD pathological hallmarks, the time-sequential appearances of AD-related cognitive and behavioral dysfunction, the chronobiologic and gender impact on calcium homeostasis and AD pathogenesis may somehow be influencing that success. In the future, such insights will be crucial for testing the validity of verapamil treatment on animal models of AD and clinical approaches.
Collapse
Affiliation(s)
- Natalija Popović
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia (IMIB), Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Nicanor Morales-Delgado
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia (IMIB), Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain.,Department of Histology and Anatomy, Faculty of Medicine, University of Miguel Hernández, Sant Joan Alacant, Spain
| | - David Vidal Mena
- Neurological Unit, University Hospital "Santa Lucia", Cartagena, Spain
| | - Antonia Alonso
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia (IMIB), Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | | | - María Caballero Bleda
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia (IMIB), Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Miroljub Popović
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia (IMIB), Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| |
Collapse
|
36
|
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: 56] [Impact Index Per Article: 11.2] [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
|
37
|
Buijink MR, Olde Engberink AHO, Wit CB, Almog A, Meijer JH, Rohling JHT, Michel S. Aging Affects the Capacity of Photoperiodic Adaptation Downstream from the Central Molecular Clock. J Biol Rhythms 2020; 35:167-179. [PMID: 31983261 PMCID: PMC7134598 DOI: 10.1177/0748730419900867] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Aging impairs circadian clock function, leading to disrupted sleep-wake patterns and a reduced capability to adapt to changes in environmental light conditions. This makes shift work or the changing of time zones challenging for the elderly and, importantly, is associated with the development of age-related diseases. However, it is unclear what levels of the clock machinery are affected by aging, which is relevant for the development of targeted interventions. We found that naturally aged mice of >24 months had a reduced rhythm amplitude in behavior compared with young controls (3-6 months). Moreover, the old animals had a strongly reduced ability to adapt to short photoperiods. Recording PER2::LUC protein expression in the suprachiasmatic nucleus revealed no impairment of the rhythms in PER2 protein under the 3 different photoperiods tested (LD: 8:16, 12:12, and 16:8). Thus, we observed a discrepancy between the behavioral phenotype and the molecular clock, and we conclude that the aging-related deficits emerge downstream of the core molecular clock. Since it is known that aging affects several intracellular and membrane components of the central clock cells, it is likely that an impairment of the interaction between the molecular clock and these components is contributing to the deficits in photoperiod adaptation.
Collapse
Affiliation(s)
- M Renate Buijink
- Department of Cellular and Chemical Biology, Laboratory for Neurophysiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anneke H O Olde Engberink
- Department of Cellular and Chemical Biology, Laboratory for Neurophysiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Charlotte B Wit
- Department of Cellular and Chemical Biology, Laboratory for Neurophysiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Assaf Almog
- Lorentz Institute for Theoretical Physics, Leiden University, Leiden, the Netherlands
| | - Johanna H Meijer
- Department of Cellular and Chemical Biology, Laboratory for Neurophysiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jos H T Rohling
- 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
| |
Collapse
|
38
|
Affiliation(s)
- Nadine Ehmann
- Department of Animal Physiology, Institute of Biology, Leipzig University, Leipzig, Germany
| | - Dennis Pauls
- Department of Animal Physiology, Institute of Biology, Leipzig University, Leipzig, Germany
| |
Collapse
|
39
|
Manoogian ENC, Kumar A, Obed D, Bergan J, Bittman EL. Suprachiasmatic function in a circadian period mutant: Duper alters light-induced activation of vasoactive intestinal peptide cells and PERIOD1 immunostaining. Eur J Neurosci 2019; 48:3319-3334. [PMID: 30346078 DOI: 10.1111/ejn.14214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 09/07/2018] [Accepted: 09/18/2018] [Indexed: 11/28/2022]
Abstract
Mammalian circadian rhythms are entrained by photic stimuli that are relayed by retinal projections to the core of the suprachiasmatic nucleus (SCN). Neuronal activation, as demonstrated by expression of the immediate early gene c-fos, leads to transcription of the core clock gene per1. The duper mutation in hamsters shortens circadian period and amplifies light-induced phase shifts. We performed two experiments to compare the number of c-FOS immunoreactive (ir) and PER1-ir cells, and the intensity of staining, in the SCN of wild-type (WT) and duper hamsters at various intervals after presentation of a 15-min light pulse in the early subjective night. Light-induced c-FOS-ir within 1 hr in the dorsocaudal SCN of duper, but not WT hamsters. In cells that express vasoactive intestinal peptide (VIP), which plays a critical role in synchronization of SCN cellular oscillators, light-induced c-FOS-ir was greater in duper than WT hamsters. After the light pulse, PER1-ir cells were found in more medial portions of the SCN than FOS-ir, and appeared with a longer latency and over a longer time course, in VIP cells of duper than wild-type hamsters. Our results indicate that the duper allele alters SCN function in ways that may contribute to changes in free running period and phase resetting.
Collapse
Affiliation(s)
- Emily N C Manoogian
- Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts
| | - Ajay Kumar
- Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts
| | - Doha Obed
- Department of Biology, University of Massachusetts, Amherst, Massachusetts
| | - Joseph Bergan
- Psychological and Brain Sciences and Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts
| | - Eric L Bittman
- Department of Biology, Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts
| |
Collapse
|
40
|
Circadian rhythms in Per1, PER2 and Ca 2+ of a solitary SCN neuron cultured on a microisland. Sci Rep 2019; 9:18271. [PMID: 31797953 PMCID: PMC6892917 DOI: 10.1038/s41598-019-54654-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/01/2019] [Indexed: 01/08/2023] Open
Abstract
Circadian rhythms in Per1, PER2 expression and intracellular Ca2+ were measured from a solitary SCN neuron or glial cell which was physically isolated from other cells. Dispersed cells were cultured on a platform of microisland (100–200 μm in diameter) in a culture dish. Significant circadian rhythms were detected in 57.1% for Per1 and 70.0% for PER2 expression. When two neurons were located on the same island, the circadian rhythms showed desynchronization, indicating a lack of oscillatory coupling. Circadian rhythms were also detected in intracellular Ca2+ of solitary SCN neurons. The ratio of circadian positive neurons was significantly larger without co-habitant of glial cells (84.4%) than with it (25.0%). A relatively large fraction of SCN neurons generates the intrinsic circadian oscillation without neural or humoral networks. In addition, glial cells seem to interrupt the expression of the circadian rhythmicity of intracellular Ca2+ under these conditions.
Collapse
|
41
|
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus is remarkable. Despite numbering only about 10,000 neurons on each side of the third ventricle, the SCN is our principal circadian clock, directing the daily cycles of behaviour and physiology that set the tempo of our lives. When this nucleus is isolated in organotypic culture, its autonomous timing mechanism can persist indefinitely, with precision and robustness. The discovery of the cell-autonomous transcriptional and post-translational feedback loops that drive circadian activity in the SCN provided a powerful exemplar of the genetic specification of complex mammalian behaviours. However, the analysis of circadian time-keeping is moving beyond single cells. Technical and conceptual advances, including intersectional genetics, multidimensional imaging and network theory, are beginning to uncover the circuit-level mechanisms and emergent properties that make the SCN a uniquely precise and robust clock. However, much remains unknown about the SCN, not least the intrinsic properties of SCN neurons, its circuit topology and the neuronal computations that these circuits support. Moreover, the convention that the SCN is a neuronal clock has been overturned by the discovery that astrocytes are an integral part of the timepiece. As a test bed for examining the relationships between genes, cells and circuits in sculpting complex behaviours, the SCN continues to offer powerful lessons and opportunities for contemporary neuroscience.
Collapse
|
42
|
The NRON complex controls circadian clock function through regulated PER and CRY nuclear translocation. Sci Rep 2019; 9:11883. [PMID: 31417156 PMCID: PMC6695496 DOI: 10.1038/s41598-019-48341-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 07/29/2019] [Indexed: 12/22/2022] Open
Abstract
Post-translational regulation plays a central role in the circadian clock mechanism. However, nucleocytoplasmic translocation of core clock proteins, a key step in circadian timekeeping, is not fully understood. Earlier we found that the NRON scaffolding complex regulates nuclear translocation of NFAT and its signaling. Here, we show that components of the NRON complex also regulate the circadian clock. In peripheral cell clock models, genetic perturbation of the NRON complex affects PER and CRY protein nuclear translocation, dampens amplitude, and alters period length. Further, we show small molecules targeting the NFAT pathway alter nuclear translocation of PER and CRY proteins and impact circadian rhythms in peripheral cells and tissue explants of the master clock in the suprachiasmatic nucleus. Taken together, these studies highlight a key role for the NRON complex in regulating PER/CRY subcellular localization and circadian timekeeping.
Collapse
|
43
|
Milićević N, Mazzaro N, de Bruin I, Wils E, Ten Brink J, Asbroek AT, Mendoza J, Bergen A, Felder-Schmittbuhl MP. Rev-Erbα and Photoreceptor Outer Segments modulate the Circadian Clock in Retinal Pigment Epithelial Cells. Sci Rep 2019; 9:11790. [PMID: 31409842 PMCID: PMC6692399 DOI: 10.1038/s41598-019-48203-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/30/2019] [Indexed: 12/24/2022] Open
Abstract
Retinal photoreceptor outer segments (POS) are renewed daily through phagocytosis by the adjacent retinal pigment epithelial (RPE) monolayer. Phagocytosis is mainly driven by the RPE circadian clock but the underlying molecular mechanisms remain elusive. Using ARPE-19 (human RPE cell-line) dispersed and monolayer cell cultures, we investigated the influence of cellular organization on the RPE clock and phagocytosis genes. PCR analysis revealed rhythmic expression of clock and phagocytosis genes in all ARPE-19 cultures. Monolayers had a tendency for higher amplitudes of clock gene oscillations. In all conditions ARNTL, CRY1, PER1-2, REV-ERBα, ITGB5, LAMP1 and PROS1 were rhythmically expressed with REV-ERBα being among the clock genes whose expression showed most robust rhythms in ARPE-19 cells. Using RPE-choroid explant preparations of the mPer2Luc knock-in mice we found that Rev-Erbα deficiency induced significantly longer periods and earlier phases of PER2-bioluminescence oscillations. Furthermore, early phagocytosis factors β5-Integrin and FAK and the lysosomal marker LAMP1 protein levels are rhythmic. Finally, POS incubation affects clock and clock-controlled phagocytosis gene expression in RPE monolayers in a time-dependent manner suggesting that POS can reset the RPE clock. These results shed some light on the complex interplay between POS, the RPE clock and clock-controlled phagocytosis machinery which is modulated by Rev-Erbα.
Collapse
Affiliation(s)
- Nemanja Milićević
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives (UPR 3212), 67000, Strasbourg, France.,Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Nadia Mazzaro
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives (UPR 3212), 67000, Strasbourg, France
| | - Ivanka de Bruin
- Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Esmée Wils
- Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jacoline Ten Brink
- Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Anneloor Ten Asbroek
- Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jorge Mendoza
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives (UPR 3212), 67000, Strasbourg, France
| | - Arthur Bergen
- Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Netherlands Institute for Neuroscience (NIN-KNAW), Amsterdam, The Netherlands
| | - Marie-Paule Felder-Schmittbuhl
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives (UPR 3212), 67000, Strasbourg, France.
| |
Collapse
|
44
|
Petersen AS, Barloese MCJ, Snoer A, Soerensen AMS, Jensen RH. Verapamil and Cluster Headache: Still a Mystery. A Narrative Review of Efficacy, Mechanisms and Perspectives. Headache 2019; 59:1198-1211. [DOI: 10.1111/head.13603] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 01/04/2023]
Affiliation(s)
- Anja S. Petersen
- Department of Neurology, Danish Headache Center Rigshospitalet‐Glostrup Glostrup Denmark
| | - Mads C. J. Barloese
- Department of Neurology, Danish Headache Center Rigshospitalet‐Glostrup Glostrup Denmark
- Department of Clinical Physiology and Nuclear Medicine, Center for Functional and Diagnostic Imaging Hvidovre Hospital Hvidovre Denmark
| | - Agneta Snoer
- Department of Neurology, Danish Headache Center Rigshospitalet‐Glostrup Glostrup Denmark
| | - Anne Mette S. Soerensen
- Department of Clinical Pharmacology Bispebjerg and Frederiksberg Hospital Copenhagen Denmark
| | - Rigmor H. Jensen
- Department of Neurology, Danish Headache Center Rigshospitalet‐Glostrup Glostrup Denmark
| |
Collapse
|
45
|
Ono D, Honma KI, Yanagawa Y, Yamanaka A, Honma S. GABA in the suprachiasmatic nucleus refines circadian output rhythms in mice. Commun Biol 2019; 2:232. [PMID: 31263776 PMCID: PMC6588595 DOI: 10.1038/s42003-019-0483-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 05/29/2019] [Indexed: 01/10/2023] Open
Abstract
In mammals, the circadian rhythms are regulated by the central clock located in the hypothalamic suprachiasmatic nucleus (SCN), which is composed of heterogeneous neurons with various neurotransmitters. Among them an inhibitory neurotransmitter, γ-Amino-Butyric-Acid (GABA), is expressed in almost all SCN neurons, however, its role in the circadian physiology is still unclear. Here, we show that the SCN of fetal mice lacking vesicular GABA transporter (VGAT-/-) or GABA synthesizing enzyme, glutamate decarboxylase (GAD65-/-/67-/-), shows burst firings associated with large Ca2+ spikes throughout 24 hours, which spread over the entire SCN slice in synchrony. By contrast, circadian PER2 rhythms in VGAT-/- and GAD65-/-/67-/- SCN remain intact. SCN-specific VGAT deletion in adult mice dampens circadian behavior rhythm. These findings indicate that GABA in the fetal SCN is necessary for refinement of the circadian firing rhythm and, possibly, for stabilizing the output signals, but not for circadian integration of multiple cellular oscillations.
Collapse
Affiliation(s)
- Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601 Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Ken-ichi Honma
- Research and Education Center for Brain Science, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638 Japan
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511 Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601 Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Sato Honma
- Research and Education Center for Brain Science, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638 Japan
| |
Collapse
|
46
|
Hastings MH, Maywood ES, Brancaccio M. The Mammalian Circadian Timing System and the Suprachiasmatic Nucleus as Its Pacemaker. BIOLOGY 2019; 8:E13. [PMID: 30862123 PMCID: PMC6466121 DOI: 10.3390/biology8010013] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/05/2019] [Accepted: 03/08/2019] [Indexed: 11/16/2022]
Abstract
The past twenty years have witnessed the most remarkable breakthroughs in our understanding of the molecular and cellular mechanisms that underpin circadian (approximately one day) time-keeping. Across model organisms in diverse taxa: cyanobacteria (Synechococcus), fungi (Neurospora), higher plants (Arabidopsis), insects (Drosophila) and mammals (mouse and humans), a common mechanistic motif of delayed negative feedback has emerged as the Deus ex machina for the cellular definition of ca. 24 h cycles. This review will consider, briefly, comparative circadian clock biology and will then focus on the mammalian circadian system, considering its molecular genetic basis, the properties of the suprachiasmatic nucleus (SCN) as the principal circadian clock in mammals and its role in synchronising a distributed peripheral circadian clock network. Finally, it will consider new directions in analysing the cell-autonomous and circuit-level SCN clockwork and will highlight the surprising discovery of a central role for SCN astrocytes as well as SCN neurons in controlling circadian behaviour.
Collapse
Affiliation(s)
- Michael H Hastings
- MRC Laboratory of Molecular Biology, Division of Neurobiology, CB2 0QH Cambridge, UK.
| | - Elizabeth S Maywood
- MRC Laboratory of Molecular Biology, Division of Neurobiology, CB2 0QH Cambridge, UK.
| | - Marco Brancaccio
- UK Dementia Research Institute at Imperial College London, Division of Brain Sciences, Department of Medicine, W12 0NN London, UK.
| |
Collapse
|
47
|
Asgari-Targhi A, Klerman EB. Mathematical modeling of circadian rhythms. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2019; 11:e1439. [PMID: 30328684 PMCID: PMC6375788 DOI: 10.1002/wsbm.1439] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 09/05/2018] [Accepted: 09/12/2018] [Indexed: 12/22/2022]
Abstract
Circadian rhythms are endogenous ~24-hr oscillations usually entrained to daily environmental cycles of light/dark. Many biological processes and physiological functions including mammalian body temperature, the cell cycle, sleep/wake cycles, neurobehavioral performance, and a wide range of diseases including metabolic, cardiovascular, and psychiatric disorders are impacted by these rhythms. Circadian clocks are present within individual cells and at tissue and organismal levels as emergent properties from the interaction of cellular oscillators. Mathematical models of circadian rhythms have been proposed to provide a better understanding of and to predict aspects of this complex physiological system. These models can be used to: (a) manipulate the system in silico with specificity that cannot be easily achieved using in vivo and in vitro experimental methods and at lower cost, (b) resolve apparently contradictory empirical results, (c) generate hypotheses, (d) design new experiments, and (e) to design interventions for altering circadian rhythms. Mathematical models differ in structure, the underlying assumptions, the number of parameters and variables, and constraints on variables. Models representing circadian rhythms at different physiologic scales and in different species are reviewed to promote understanding of these models and facilitate their use. This article is categorized under: Physiology > Mammalian Physiology in Health and Disease Models of Systems Properties and Processes > Organ, Tissue, and Physiological Models.
Collapse
|
48
|
Krylov VV, Kantserova NP, Lysenko LA, Osipova EA. A simulated geomagnetic storm unsynchronizes with diurnal geomagnetic variation affecting calpain activity in roach and great pond snail. INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2019; 63:241-246. [PMID: 30680619 DOI: 10.1007/s00484-018-01657-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 10/02/2018] [Accepted: 10/30/2018] [Indexed: 06/09/2023]
Abstract
It has been suggested that geomagnetic storms could be perceived by organisms via disruption of naturally occurring diurnal geomagnetic variation. This variation, in turn, is viewed by way of a zeitgeber for biological circadian rhythms. The biological effects of a geomagnetic storm, therefore, could depend on the local time of day when its main phase occurs. We have assessed calpain activity in tissues of roach (Rutilus rutilus) and great pond snail (Limnaea stagnalis) after exposure to a simulated geomagnetic storm, reproduced at different times of day, in order to evaluate this hypothesis. Significant decrease in calpain activity was observed in organisms exposed to the simulated geomagnetic storm whose main phase, and initial period of a recovery phase, did not coincide with the expected peak of diurnal geomagnetic variation. The results obtained are considered an experimental confirmation of the aforementioned hypothesis. Improvement of a correlative approach for the assessment of biological effects of geomagnetic activity can be achieved by considering information on the synchronization of geomagnetic storm's main phase with diurnal geomagnetic variation.
Collapse
Affiliation(s)
- Viacheslav V Krylov
- I.D. Papanin Institute for Biology of Inland Waters of Russian Academy of Sciences, Borok 109, Nekouz, Yaroslavl oblast, Russian Federation, 152742.
| | - N P Kantserova
- The Institute of Biology, Karelian Research Centre of Russian Academy of Sciences, Pushkinskaya, 11, Petrozavodsk, Russian Federation, 185910
| | - L A Lysenko
- The Institute of Biology, Karelian Research Centre of Russian Academy of Sciences, Pushkinskaya, 11, Petrozavodsk, Russian Federation, 185910
| | - E A Osipova
- I.D. Papanin Institute for Biology of Inland Waters of Russian Academy of Sciences, Borok 109, Nekouz, Yaroslavl oblast, Russian Federation, 152742
| |
Collapse
|
49
|
Collins B, Brown SA. Beyond the molecular clock. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2018.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
50
|
Freyberg Z, Logan RW. The Intertwined Roles of Circadian Rhythms and Neuronal Metabolism Fueling Drug Reward and Addiction. CURRENT OPINION IN PHYSIOLOGY 2018; 5:80-89. [PMID: 30631826 DOI: 10.1016/j.cophys.2018.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Drug addiction is a highly prevalent and devastating disorder with few effective treatments, resulting in enormous burdens on family and society. The cellular and behavioral effects of drugs of abuse are related to their abilities to elevate synaptic dopamine levels. Midbrain dopaminergic neurons projecting from the ventral tegmental area to the nucleus accumbens play crucial roles in substance-induced neural and behavioral plasticity. Significantly, increasing work suggests that interplay between the brain circadian system and the cellular bioenergetic machinery in these dopamine neurons plays a critical role in mediating the actions of drugs of abuse. Here, we describe recent progress in elucidating the interconnections between circadian and metabolic systems at the molecular and cellular levels and their relationships to modulation of drug reward and addiction.
Collapse
Affiliation(s)
- Zachary Freyberg
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, pittsburgh, PA, USA 15219.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA USA 15213
| | - Ryan W Logan
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, pittsburgh, PA, USA 15219.,Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, ME, 04609
| |
Collapse
|