51
|
Lee CH, Murrell CE, Chu A, Pan X. Circadian Regulation of Apolipoproteins in the Brain: Implications in Lipid Metabolism and Disease. Int J Mol Sci 2023; 24:17415. [PMID: 38139244 PMCID: PMC10743770 DOI: 10.3390/ijms242417415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/08/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023] Open
Abstract
The circadian rhythm is a 24 h internal clock within the body that regulates various factors, including sleep, body temperature, and hormone secretion. Circadian rhythm disruption is an important risk factor for many diseases including neurodegenerative illnesses. The central and peripheral oscillators' circadian clock network controls the circadian rhythm in mammals. The clock genes govern the central clock in the suprachiasmatic nucleus (SCN) of the brain. One function of the circadian clock is regulating lipid metabolism. However, investigations of the circadian regulation of lipid metabolism-associated apolipoprotein genes in the brain are lacking. This review summarizes the rhythmic expression of clock genes and lipid metabolism-associated apolipoprotein genes within the SCN in Mus musculus. Nine of the twenty apolipoprotein genes identified from searching the published database (SCNseq and CircaDB) are highly expressed in the SCN. Most apolipoprotein genes (ApoE, ApoC1, apoA1, ApoH, ApoM, and Cln) show rhythmic expression in the brain in mice and thus might be regulated by the master clock. Therefore, this review summarizes studies on lipid-associated apolipoprotein genes in the SCN and other brain locations, to understand how apolipoproteins associated with perturbed cerebral lipid metabolism cause multiple brain diseases and disorders. This review describes recent advancements in research, explores current questions, and identifies directions for future research.
Collapse
Affiliation(s)
- Chaeeun Hannah Lee
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Charlotte Ellzabeth Murrell
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Alexander Chu
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Xiaoyue Pan
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Langone Hospital-Long Island, Mineola, NY 11501, USA
| |
Collapse
|
52
|
González-Vila A, Luengo-Mateos M, Silveira-Loureiro M, Garrido-Gil P, Ohinska N, González-Domínguez M, Labandeira-García JL, García-Cáceres C, López M, Barca-Mayo O. Astrocytic insulin receptor controls circadian behavior via dopamine signaling in a sexually dimorphic manner. Nat Commun 2023; 14:8175. [PMID: 38071352 PMCID: PMC10710518 DOI: 10.1038/s41467-023-44039-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Mammalian circadian clocks respond to feeding and light cues, adjusting internal rhythms with day/night cycles. Astrocytes serve as circadian timekeepers, driving daily physiological rhythms; however, it's unknown how they ensure precise cycle-to-cycle rhythmicity. This is critical for understanding why mistimed or erratic feeding, as in shift work, disrupts circadian physiology- a condition linked to type 2 diabetes and obesity. Here, we show that astrocytic insulin signaling sets the free-running period of locomotor activity in female mice and food entrainment in male mice. Additionally, ablating the insulin receptor in hypothalamic astrocytes alters cyclic energy homeostasis differently in male and female mice. Remarkably, the mutants exhibit altered dopamine metabolism, and the pharmacological modulation of dopaminergic signaling partially restores distinct circadian traits in both male and female mutant mice. Our findings highlight the role of astrocytic insulin-dopaminergic signaling in conveying time-of-feeding or lighting cues to the astrocyte clock, thus governing circadian behavior in a sex-specific manner.
Collapse
Affiliation(s)
- Antía González-Vila
- Circadian and Glial Biology Lab, Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- NeurObesity Lab, Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - María Luengo-Mateos
- Circadian and Glial Biology Lab, Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - María Silveira-Loureiro
- Circadian and Glial Biology Lab, Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- NeurObesity Lab, Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Pablo Garrido-Gil
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Department of Morphological Science, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Nataliia Ohinska
- Circadian and Glial Biology Lab, Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Marco González-Domínguez
- Circadian and Glial Biology Lab, Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Jose Luis Labandeira-García
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Department of Morphological Science, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich & German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336, Munich, Germany
| | - Miguel López
- NeurObesity Lab, Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Olga Barca-Mayo
- Circadian and Glial Biology Lab, Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
53
|
Postnov D, Semyachkina-Glushkovskaya O, Litvinenko E, Kurths J, Penzel T. Mechanisms of Activation of Brain's Drainage during Sleep: The Nightlife of Astrocytes. Cells 2023; 12:2667. [PMID: 37998402 PMCID: PMC10670149 DOI: 10.3390/cells12222667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
The study of functions, mechanisms of generation, and pathways of movement of cerebral fluids has a long history, but the last decade has been especially productive. The proposed glymphatic hypothesis, which suggests a mechanism of the brain waste removal system (BWRS), caused an active discussion on both the criticism of some of the perspectives and our intensive study of new experimental facts. It was especially found that the intensity of the metabolite clearance changes significantly during the transition between sleep and wakefulness. Interestingly, at the cellular level, a number of aspects of this problem have been focused on, such as astrocytes-glial cells, which, over the past two decades, have been recognized as equal partners of neurons and perform many important functions. In particular, an important role was assigned to astrocytes within the framework of the glymphatic hypothesis. In this review, we return to the "astrocytocentric" view of the BWRS function and the explanation of its activation during sleep from the viewpoint of new findings over the last decade. Our main conclusion is that the BWRS's action may be analyzed both at the systemic (whole-brain) and at the local (cellular) level. The local level means here that the neuro-glial-vascular unit can also be regarded as the smallest functional unit of sleep, and therefore, the smallest functional unit of the BWRS.
Collapse
Affiliation(s)
- Dmitry Postnov
- Department of Optics and Biophotonics, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia;
| | - Oxana Semyachkina-Glushkovskaya
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia; (O.S.-G.); (J.K.)
- Physics Department, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany
| | - Elena Litvinenko
- Department of Optics and Biophotonics, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia;
| | - Jürgen Kurths
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia; (O.S.-G.); (J.K.)
- Physics Department, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany
- Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
| | - Thomas Penzel
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia; (O.S.-G.); (J.K.)
- Charité — Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
54
|
Kara N, Iweka CA, Blacher E. Chrono-Gerontology: Integrating Circadian Rhythms and Aging in Stroke Research. Adv Biol (Weinh) 2023; 7:e2300048. [PMID: 37409422 DOI: 10.1002/adbi.202300048] [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: 01/31/2023] [Revised: 05/14/2023] [Indexed: 07/07/2023]
Abstract
Stroke is a significant public health concern for elderly individuals. However, the majority of pre-clinical studies utilize young and healthy rodents, which may result in failure of candidate therapies in clinical trials. In this brief review/perspective, the complex link between circadian rhythms, aging, innate immunity, and the gut microbiome to ischemic injury onset, progression, and recovery is discussed. Short-chain fatty acids and nicotinamide adenine dinucleotide+ (NAD+ ) production by the gut microbiome are highlighted as key mechanisms with profound rhythmic behavior, and it is suggested to boost them as prophylactic/therapeutic approaches. Integrating aging, its associated comorbidities, and circadian regulation of physiological processes into stroke research may increase the translational value of pre-clinical studies and help to schedule the optimal time window for existing practices to improve stroke outcome and recovery.
Collapse
Affiliation(s)
- Nirit Kara
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus Givat-Ram, Jerusalem, 9190401, Israel
| | - Chinyere Agbaegbu Iweka
- Department of Neurology & Neurological Sciences, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Eran Blacher
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus Givat-Ram, Jerusalem, 9190401, Israel
| |
Collapse
|
55
|
Murgo E, Colangelo T, Bellet MM, Malatesta F, Mazzoccoli G. Role of the Circadian Gas-Responsive Hemeprotein NPAS2 in Physiology and Pathology. BIOLOGY 2023; 12:1354. [PMID: 37887064 PMCID: PMC10603908 DOI: 10.3390/biology12101354] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/14/2023] [Accepted: 10/20/2023] [Indexed: 10/28/2023]
Abstract
Neuronal PAS domain protein 2 (NPAS2) is a hemeprotein comprising a basic helix-loop-helix domain (bHLH) and two heme-binding sites, the PAS-A and PAS-B domains. This protein acts as a pyridine nucleotide-dependent and gas-responsive CO-dependent transcription factor and is encoded by a gene whose expression fluctuates with circadian rhythmicity. NPAS2 is a core cog of the molecular clockwork and plays a regulatory role on metabolic pathways, is important for the function of the central nervous system in mammals, and is involved in carcinogenesis as well as in normal biological functions and processes, such as cardiovascular function and wound healing. We reviewed the scientific literature addressing the various facets of NPAS2 and framing this gene/protein in several and very different research and clinical fields.
Collapse
Affiliation(s)
- Emanuele Murgo
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Laboratory, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy;
| | - Tommaso Colangelo
- Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71100 Foggia, Italy;
- Cancer Cell Signaling Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy
| | - Maria Marina Bellet
- Department of Medicine and Surgery, University of Perugia, P.le L. Severi 1, 06132 Perugia, Italy;
| | - Francesco Malatesta
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Gianluigi Mazzoccoli
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Laboratory, Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy;
| |
Collapse
|
56
|
Xu X, Wang J, Chen G. Circadian cycle and neuroinflammation. Open Life Sci 2023; 18:20220712. [PMID: 37872969 PMCID: PMC10590615 DOI: 10.1515/biol-2022-0712] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/05/2023] [Accepted: 08/06/2023] [Indexed: 10/25/2023] Open
Abstract
Circadian cycle is a fundamental characteristic of life formed in the long-term evolution of organisms and plays an important role in maintaining the proliferation, migration, and activation of immune cells. Studies have shown that circadian rhythm disorders affect the occurrence and development of neuroinflammation by inducing glial cell activation and peripheral immune responses. In this article, we briefly described the research progress of neuroinflammation and circadian rhythm in recent years and explored the effects and possible mechanism of circadian rhythmicity on microglia, astrocytes, and peripheral immune function.
Collapse
Affiliation(s)
- Xinzi Xu
- College of Clinical Chinese Medicine, Hubei University of Chinese Medicine, Wuhan430065, China
| | - Junli Wang
- Department of Neurology, Wuhan No. 1 Hospital, Wuhan430022, China
| | - Guohua Chen
- Department of Neurology, Wuhan No. 1 Hospital, Wuhan430022, China
| |
Collapse
|
57
|
Onodera K, Tsuno Y, Hiraoka Y, Tanaka K, Maejima T, Mieda M. In vivo recording of the circadian calcium rhythm in Prokineticin 2 neurons of the suprachiasmatic nucleus. Sci Rep 2023; 13:16974. [PMID: 37813987 PMCID: PMC10562406 DOI: 10.1038/s41598-023-44282-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/05/2023] [Indexed: 10/11/2023] Open
Abstract
Prokineticin 2 (Prok2) is a small protein expressed in a subpopulation of neurons in the suprachiasmatic nucleus (SCN), the primary circadian pacemaker in mammals. Prok2 has been implicated as a candidate output molecule from the SCN to control multiple circadian rhythms. Genetic manipulation specific to Prok2-producing neurons would be a powerful approach to understanding their function. Here, we report the generation of Prok2-tTA knock-in mice expressing the tetracycline transactivator (tTA) specifically in Prok2 neurons and an application of these mice to in vivo recording of Ca2+ rhythms in these neurons. First, the specific and efficient expression of tTA in Prok2 neurons was verified by crossing the mice with EGFP reporter mice. Prok2-tTA mice were then used to express a fluorescent Ca2+ sensor protein to record the circadian Ca2+ rhythm in SCN Prok2 neurons in vivo. Ca2+ in these cells showed clear circadian rhythms in both light-dark and constant dark conditions, with their peaks around midday. Notably, the hours of high Ca2+ nearly coincided with the rest period of the behavioral rhythm. These observations fit well with the predicted function of Prok2 neurons as a candidate output pathway of the SCN by suppressing locomotor activity during both daytime and subjective daytime.
Collapse
Affiliation(s)
- Kaito Onodera
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Yusuke Tsuno
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Yuichi Hiraoka
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kohichi Tanaka
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takashi Maejima
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan.
| |
Collapse
|
58
|
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
|
59
|
Brazhe A, Verisokin A, Verveyko D, Postnov D. Astrocytes: new evidence, new models, new roles. Biophys Rev 2023; 15:1303-1333. [PMID: 37975000 PMCID: PMC10643736 DOI: 10.1007/s12551-023-01145-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/08/2023] [Indexed: 11/19/2023] Open
Abstract
Astrocytes have been in the limelight of active research for about 3 decades now. Over this period, ideas about their function and role in the nervous system have evolved from simple assistance in energy supply and homeostasis maintenance to a complex informational and metabolic hub that integrates data on local neuronal activity, sensory and arousal context, and orchestrates many crucial processes in the brain. Rapid progress in experimental techniques and data analysis produces a growing body of data, which can be used as a foundation for formulation of new hypotheses, building new refined mathematical models, and ultimately should lead to a new level of understanding of the contribution of astrocytes to the cognitive tasks performed by the brain. Here, we highlight recent progress in astrocyte research, which we believe expands our understanding of how low-level signaling at a cellular level builds up to processes at the level of the whole brain and animal behavior. We start our review with revisiting data on the role of noradrenaline-mediated astrocytic signaling in locomotion, arousal, sensory integration, memory, and sleep. We then briefly review astrocyte contribution to the regulation of cerebral blood flow regulation, which is followed by a discussion of biophysical mechanisms underlying astrocyte effects on different brain processes. The experimental section is closed by an overview of recent experimental techniques available for modulation and visualization of astrocyte dynamics. We then evaluate how the new data can be potentially incorporated into the new mathematical models or where and how it already has been done. Finally, we discuss an interesting prospect that astrocytes may be key players in important processes such as the switching between sleep and wakefulness and the removal of toxic metabolites from the brain milieu.
Collapse
Affiliation(s)
- Alexey Brazhe
- Department of Biophysics, Biological Faculty, Lomonosov Moscow State University, Leninskie Gory, 1/24, Moscow, 119234 Russia
- Department of Molecular Neurobiology, Institute of Bioorganic Chemistry RAS, GSP-7, Miklukho-Maklay Str., 16/10, Moscow, 117997 Russia
| | - Andrey Verisokin
- Department of Theoretical Physics, Kursk State University, Radishcheva st., 33, Kursk, 305000 Russia
| | - Darya Verveyko
- Department of Theoretical Physics, Kursk State University, Radishcheva st., 33, Kursk, 305000 Russia
| | - Dmitry Postnov
- Department of Optics and Biophotonics, Saratov State University, Astrakhanskaya st., 83, Saratov, 410012 Russia
| |
Collapse
|
60
|
Yao ZM, Sun XR, Huang J, Chen L, Dong SY. Astrocyte-Neuronal Communication and Its Role in Stroke. Neurochem Res 2023; 48:2996-3006. [PMID: 37329448 DOI: 10.1007/s11064-023-03966-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/19/2023]
Abstract
Astrocytes are the most abundant glial cells in the central nervous system. These cells are an important hub for intercellular communication. They participate in various pathophysiological processes, including synaptogenesis, metabolic transformation, scar production, and blood-brain barrier repair. The mechanisms and functional consequences of astrocyte-neuron signaling are more complex than previously thought. Stroke is a disease associated with neurons in which astrocytes also play an important role. Astrocytes respond to the alterations in the brain microenvironment after stroke, providing required substances to neurons. However, they can also have harmful effects. In this review, we have summarized the function of astrocytes, their association with neurons, and two paradigms of the inflammatory response, which suggest that targeting astrocytes may be an effective strategy for treating stroke.
Collapse
Affiliation(s)
- Zi-Meng Yao
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Xiao-Rong Sun
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Jie Huang
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Lei Chen
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Shu-Ying Dong
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China.
- Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu, Anhui, China.
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, Anhui, China.
| |
Collapse
|
61
|
Lemcke R, Egebjerg C, Berendtsen NT, Egerod KL, Thomsen AR, Pers TH, Christensen JP, Kornum BR. Molecular consequences of peripheral Influenza A infection on cell populations in the murine hypothalamus. eLife 2023; 12:RP87515. [PMID: 37698546 PMCID: PMC10497288 DOI: 10.7554/elife.87515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023] Open
Abstract
Infection with Influenza A virus (IAV) causes the well-known symptoms of the flu, including fever, loss of appetite, and excessive sleepiness. These responses, mediated by the brain, will normally disappear once the virus is cleared from the system, but a severe respiratory virus infection may cause long-lasting neurological disturbances. These include encephalitis lethargica and narcolepsy. The mechanisms behind such long lasting changes are unknown. The hypothalamus is a central regulator of the homeostatic response during a viral challenge. To gain insight into the neuronal and non-neuronal molecular changes during an IAV infection, we intranasally infected mice with an H1N1 virus and extracted the brain at different time points. Using single-nucleus RNA sequencing (snRNA-seq) of the hypothalamus, we identify transcriptional effects in all identified cell populations. The snRNA-seq data showed the most pronounced transcriptional response at 3 days past infection, with a strong downregulation of genes across all cell types. General immune processes were mainly impacted in microglia, the brain resident immune cells, where we found increased numbers of cells expressing pro-inflammatory gene networks. In addition, we found that most neuronal cell populations downregulated genes contributing to the energy homeostasis in mitochondria and protein translation in the cytosol, indicating potential reduced cellular and neuronal activity. This might be a preventive mechanism in neuronal cells to avoid intracellular viral replication and attack by phagocytosing cells. The change of microglia gene activity suggest that this is complemented by a shift in microglia activity to provide increased surveillance of their surroundings.
Collapse
Affiliation(s)
- René Lemcke
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Christine Egebjerg
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Nicolai T Berendtsen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Kristoffer L Egerod
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Allan R Thomsen
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Jan P Christensen
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Birgitte R Kornum
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| |
Collapse
|
62
|
Muñoz EM, Martínez Cerdeño V. Editorial: Transcription regulation - Brain development and homeostasis - A finely tuned and orchestrated scenario in physiology and pathology, volume II. Front Mol Neurosci 2023; 16:1280573. [PMID: 37736114 PMCID: PMC10509287 DOI: 10.3389/fnmol.2023.1280573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 08/25/2023] [Indexed: 09/23/2023] Open
Affiliation(s)
- Estela M. Muñoz
- Institute of Histology and Embryology of Mendoza (IHEM), National University of Cuyo (UNCuyo), National Scientific and Technical Research Council (CONICET), Mendoza, Argentina
| | - Verónica Martínez Cerdeño
- Department of Pathology and Laboratory Medicine, Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children of Northern California, and MIND Institute at the UC Davis Medical Center, University of California Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
63
|
Thirouin ZS, Gizowski C, Murtaz A, Bourque CW. Sex-specific differences in the circadian pattern of action potential firing by rat suprachiasmatic nucleus vasopressin neurons. J Neuroendocrinol 2023; 35:e13273. [PMID: 37132408 DOI: 10.1111/jne.13273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 05/04/2023]
Abstract
The suprachiasmatic nucleus (SCN) of the hypothalamus serves as the master circadian clock in mammals. Most SCN neurons express the inhibitory neurotransmitter GABA (gamma amino butyric acid) along with a peptide cotransmitter. Notably, the neuropeptides vasopressin (VP) and vasoactive intestinal peptide (VIP) define two prominent clusters within the SCN: those located in the ventral core (VIP) and those forming the dorsomedial "shell" of the nucleus (VP). Axons emerging from VP neurons in the shell are thought to mediate much of the SCN's output to other brain regions as well as VP release into the cerebrospinal fluid (CSF). Previous work has shown that VP release by SCN neurons is activity dependent and SCN VP neurons fire action potentials at a higher rate during the light phase. Accordingly, CSF VP levels are higher during daytime. Interestingly, the amplitude of the CSF VP rhythm is greater in males than females, suggesting the existence of sex differences in the electrical activity of SCN VP neurons. Here we investigated this hypothesis by performing cell-attached recordings from 1070 SCN VP neurons across the entire circadian cycle in both sexes of transgenic rats that express green fluorescent protein (GFP) driven by the VP gene promoter. Using an immunocytochemical approach we confirmed that >60% of SCN VP neurons display visible GFP. Recordings in acute coronal slices revealed that VP neurons display a striking circadian pattern of action potential firing, but the characteristics of this activity cycle differ in males and females. Specifically, neurons in males reached a significantly higher peak firing frequency during subjective daytime compared to females and the acrophase occurred ~1 h earlier in females. Peak firing rates in females were not significantly different at various phases of the estrous cycle.
Collapse
Affiliation(s)
- Zahra S Thirouin
- Center for Research in Neuroscience, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Claire Gizowski
- Center for Research in Neuroscience, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Anzala Murtaz
- Center for Research in Neuroscience, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Charles W Bourque
- Center for Research in Neuroscience, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| |
Collapse
|
64
|
Csemer A, Kovács A, Maamrah B, Pocsai K, Korpás K, Klekner Á, Szücs P, Nánási PP, Pál B. Astrocyte- and NMDA receptor-dependent slow inward currents differently contribute to synaptic plasticity in an age-dependent manner in mouse and human neocortex. Aging Cell 2023; 22:e13939. [PMID: 37489544 PMCID: PMC10497838 DOI: 10.1111/acel.13939] [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] [Received: 12/06/2022] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/26/2023] Open
Abstract
Slow inward currents (SICs) are known as excitatory events of neurons elicited by astrocytic glutamate via activation of extrasynaptic NMDA receptors. By using slice electrophysiology, we tried to provide evidence that SICs can elicit synaptic plasticity. Age dependence of SICs and their impact on synaptic plasticity was also investigated in both on murine and human cortical slices. It was found that SICs can induce a moderate synaptic plasticity, with features similar to spike timing-dependent plasticity. Overall SIC activity showed a clear decline with aging in humans and completely disappeared above a cutoff age. In conclusion, while SICs contribute to a form of astrocyte-dependent synaptic plasticity both in mice and humans, this plasticity is differentially affected by aging. Thus, SICs are likely to play an important role in age-dependent physiological and pathological alterations of synaptic plasticity.
Collapse
Affiliation(s)
- Andrea Csemer
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- Doctoral School of Molecular MedicineUniversity of DebrecenDebrecenHungary
| | - Adrienn Kovács
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Baneen Maamrah
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- Doctoral School of Molecular MedicineUniversity of DebrecenDebrecenHungary
| | - Krisztina Pocsai
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Kristóf Korpás
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Álmos Klekner
- Department of Neurosurgery, Clinical CentreUniversity of DebrecenDebrecenHungary
| | - Péter Szücs
- Department of Anatomy, Histology and Embryology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Péter P. Nánási
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- Department of Dental Physiology and Pharmacology, Faculty of DentistryUniversity of DebrecenDebrecenHungary
| | - Balázs Pál
- Department of Physiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- Doctoral School of Molecular MedicineUniversity of DebrecenDebrecenHungary
| |
Collapse
|
65
|
Luengo-Mateos M, González-Vila A, Vicente Dragano NR, Ohinska N, Silveira-Loureiro M, González-Domínguez M, Estévez-Salguero Á, Novelle-Rodríguez P, López M, Barca-Mayo O. Hypothalamic astrocytic-BMAL1 regulates energy homeostasis in a sex-dependent manner. Cell Rep 2023; 42:112949. [PMID: 37542717 DOI: 10.1016/j.celrep.2023.112949] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 06/12/2023] [Accepted: 07/20/2023] [Indexed: 08/07/2023] Open
Abstract
Here, we demonstrate that hypothalamic astrocytic BMAL1 computes cyclic metabolic information to optimize energetic resources in a sexually dimorphic manner. Knockdown of BMAL1 in female astrocytes leads to negative energy balance and alters basal metabolic cycles without affecting circadian locomotor activity. Thus, astrocytic BMAL1 contributes to the control of energy balance through the modulation of the metabolic rate, hepatic and white adipose tissue lipogenesis, and the activity of brown adipose tissue. Importantly, most of these alterations are specific to hypothalamic astrocytic BMAL1. Moreover, female mice with BMAL1 knockdown in astrocytes exhibited a "male-like" metabolic obese phenotype when fed a high-fat diet. Overall, our results suggest a sexually dimorphic effect of astrocytic BMAL1 on the regulation of energy homeostasis, which may be of interest in the physiopathology of obesity and related comorbidities.
Collapse
Affiliation(s)
- María Luengo-Mateos
- Physiology Department, Molecular Medicine, and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Antía González-Vila
- Physiology Department, Molecular Medicine, and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Nathalia Romanelli Vicente Dragano
- Physiology Department, Molecular Medicine, and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Nataliia Ohinska
- Physiology Department, Molecular Medicine, and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - María Silveira-Loureiro
- Physiology Department, Molecular Medicine, and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Marco González-Domínguez
- Physiology Department, Molecular Medicine, and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ánxela Estévez-Salguero
- Physiology Department, Molecular Medicine, and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Paula Novelle-Rodríguez
- Physiology Department, Molecular Medicine, and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Miguel López
- Physiology Department, Molecular Medicine, and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain.
| | - Olga Barca-Mayo
- Physiology Department, Molecular Medicine, and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
66
|
Davidson AJ, Beckner D, Bonnefont X. A Journey in the Brain's Clock: In Vivo Veritas? BIOLOGY 2023; 12:1136. [PMID: 37627020 PMCID: PMC10452196 DOI: 10.3390/biology12081136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023]
Abstract
The suprachiasmatic nuclei (SCN) of the hypothalamus contain the circadian pacemaker that coordinates mammalian rhythms in tune with the day-night cycle. Understanding the determinants of the intrinsic rhythmicity of this biological clock, its outputs, and resetting by environmental cues, has been a longstanding goal of the field. Integrated techniques of neurophysiology, including lesion studies and in vivo multi-unit electrophysiology, have been key to characterizing the rhythmic nature and outputs of the SCN in animal models. In parallel, reduced ex vivo and in vitro approaches have permitted us to unravel molecular, cellular, and multicellular mechanisms underlying the pacemaker properties of the SCN. New questions have emerged in recent years that will require combining investigation at a cell resolution within the physiological context of the living animal: What is the role of specific cell subpopulations in the SCN neural network? How do they integrate various external and internal inputs? What are the circuits involved in controlling other body rhythms? Here, we review what we have already learned about the SCN from in vivo studies, and how the recent development of new genetically encoded tools and cutting-edge imaging technology in neuroscience offers chronobiologists the opportunity to meet these challenges.
Collapse
Affiliation(s)
- Alec J. Davidson
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Delaney Beckner
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Xavier Bonnefont
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier, France
| |
Collapse
|
67
|
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: 7] [Impact Index Per Article: 3.5] [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
|
68
|
Ballester Roig MN, Roy PG, Hannou L, Delignat-Lavaud B, Sully Guerrier TA, Bélanger-Nelson E, Dufort-Gervais J, Mongrain V. Transcriptional regulation of the mouse EphA4, Ephrin-B2 and Ephrin-A3 genes by the circadian clock machinery. Chronobiol Int 2023; 40:983-1003. [PMID: 37551686 DOI: 10.1080/07420528.2023.2237580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 08/09/2023]
Abstract
Circadian rhythms originate from molecular feedback loops. In mammals, the transcription factors CLOCK and BMAL1 act on regulatory elements (i.e. E-boxes) to shape biological functions in a rhythmic manner. The EPHA4 receptor and its ligands Ephrins (EFN) are cell adhesion molecules regulating neurotransmission and neuronal morphology. Previous studies showed the presence of E-boxes in the genes of EphA4 and specific Ephrins, and that EphA4 knockout mice have an altered circadian rhythm of locomotor activity. We thus hypothesized that the core clock machinery regulates the gene expression of EphA4, EfnB2 and EfnA3. CLOCK and BMAL1 (or NPAS2 and BMAL2) were found to have transcriptional activity on distal and proximal regions of EphA4, EfnB2 and EfnA3 putative promoters. A constitutively active form of glycogen synthase kinase 3β (GSK3β; a negative regulator of CLOCK and BMAL1) blocked the transcriptional induction. Mutating the E-boxes of EphA4 distal promoter sequence reduced transcriptional induction. EPHA4 and EFNB2 protein levels did not show circadian variations in the mouse suprachiasmatic nucleus or prefrontal cortex. The findings uncover that core circadian transcription factors can regulate the gene expression of elements of the Eph/Ephrin system, which might contribute to circadian rhythmicity in biological processes in the brain or peripheral tissues.
Collapse
Affiliation(s)
- Maria Neus Ballester Roig
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada
- Centre de Recherche du CHUM, Montreal, Quebec, Canada
- Recherche CIUSSS-NIM, Montreal, Quebec, Canada
| | - Pierre-Gabriel Roy
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada
- Recherche CIUSSS-NIM, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | | - Valérie Mongrain
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada
- Centre de Recherche du CHUM, Montreal, Quebec, Canada
- Recherche CIUSSS-NIM, Montreal, Quebec, Canada
| |
Collapse
|
69
|
Hastings MH, Brancaccio M, Gonzalez-Aponte MF, Herzog ED. Circadian Rhythms and Astrocytes: The Good, the Bad, and the Ugly. Annu Rev Neurosci 2023; 46:123-143. [PMID: 36854316 PMCID: PMC10381027 DOI: 10.1146/annurev-neuro-100322-112249] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
This review explores the interface between circadian timekeeping and the regulation of brain function by astrocytes. Although astrocytes regulate neuronal activity across many time domains, their cell-autonomous circadian clocks exert a particular role in controlling longer-term oscillations of brain function: the maintenance of sleep states and the circadian ordering of sleep and wakefulness. This is most evident in the central circadian pacemaker, the suprachiasmatic nucleus, where the molecular clock of astrocytes suffices to drive daily cycles of neuronal activity and behavior. In Alzheimer's disease, sleep impairments accompany cognitive decline. In mouse models of the disease, circadian disturbances accelerate astroglial activation and other brain pathologies, suggesting that daily functions in astrocytes protect neuronal homeostasis. In brain cancer, treatment in the morning has been associated with prolonged survival, and gliomas have daily rhythms in gene expression and drug sensitivity. Thus, circadian time is fast becoming critical to elucidating reciprocal astrocytic-neuronal interactions in health and disease.
Collapse
Affiliation(s)
- Michael H Hastings
- Division of Neurobiology, Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom;
| | - Marco Brancaccio
- UK Dementia Research Institute and Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Maria F Gonzalez-Aponte
- Department of Biology, Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri, USA;
| | - Erik D Herzog
- Department of Biology, Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri, USA;
| |
Collapse
|
70
|
Peng YF, Wang LL, Gu JH, Zeng YQ. Effects of astaxanthin on depressive and sleep symptoms: A narrative mini-review. Heliyon 2023; 9:e18288. [PMID: 37539097 PMCID: PMC10393630 DOI: 10.1016/j.heliyon.2023.e18288] [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: 11/04/2022] [Revised: 07/03/2023] [Accepted: 07/13/2023] [Indexed: 08/05/2023] Open
Abstract
Major depressive disorder (MDD) is a prevalent psychiatric condition that results in persistent feelings of sadness and loss of interest, imposing a significant economic burden on health systems and society. Impaired sleep is both a symptom and a risk factor for depression. Natural astaxanthin (AST), a carotenoid primarily derived from algae and aquatic animals, possesses multiple pharmacological properties such as anti-inflammatory, anti-apoptotic, and antioxidant stress effects. Prior research suggests that AST may have antidepressant properties. This mini-review highlights the potential mechanisms by which AST can prevent depression, providing novel insights into drug research for depression treatment. Specifically, this mechanism suggests that astaxanthin may improve sleep and thus potentially aid in the treatment of depression.
Collapse
Affiliation(s)
| | | | | | - Yue-Qin Zeng
- Corresponding author. Academy of Biomedical Engineering, Kunming Medical University, Kunming, China.
| |
Collapse
|
71
|
Francis TC, Porcu A. Emotionally clocked out: cell-type specific regulation of mood and anxiety by the circadian clock system in the brain. Front Mol Neurosci 2023; 16:1188184. [PMID: 37441675 PMCID: PMC10333695 DOI: 10.3389/fnmol.2023.1188184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/29/2023] [Indexed: 07/15/2023] Open
Abstract
Circadian rhythms are self-sustained oscillations of biological systems that allow an organism to anticipate periodic changes in the environment and optimally align feeding, sleep, wakefulness, and the physiological and biochemical processes that support them within the 24 h cycle. These rhythms are generated at a cellular level by a set of genes, known as clock genes, which code for proteins that inhibit their own transcription in a negative feedback loop and can be perturbed by stress, a risk factor for the development of mood and anxiety disorders. A role for circadian clocks in mood and anxiety has been suggested for decades on the basis of clinical observations, and the dysregulation of circadian rhythms is a prominent clinical feature of stress-related disorders. Despite our understanding of central clock structure and function, the effect of circadian dysregulation in different neuronal subtypes in the suprachiasmatic nucleus (SCN), the master pacemaker region, as well as other brain systems regulating mood, including mesolimbic and limbic circuits, is just beginning to be elucidated. In the brain, circadian clocks regulate neuronal physiological functions, including neuronal activity, synaptic plasticity, protein expression, and neurotransmitter release which in turn affect mood-related behaviors via cell-type specific mechanisms. Both animal and human studies have revealed an association between circadian misalignment and mood disorders and suggest that internal temporal desynchrony might be part of the etiology of psychiatric disorders. To date, little work has been conducted associating mood-related phenotypes to cell-specific effects of the circadian clock disruptions. In this review, we discuss existing literature on how clock-driven changes in specific neuronal cell types might disrupt phase relationships among cellular communication, leading to neuronal circuit dysfunction and changes in mood-related behavior. In addition, we examine cell-type specific circuitry underlying mood dysfunction and discuss how this circuitry could affect circadian clock. We provide a focus for future research in this area and a perspective on chronotherapies for mood and anxiety disorders.
Collapse
Affiliation(s)
- T. Chase Francis
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Alessandra Porcu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
72
|
Que M, Li Y, Wang X, Zhan G, Luo X, Zhou Z. Role of astrocytes in sleep deprivation: accomplices, resisters, or bystanders? Front Cell Neurosci 2023; 17:1188306. [PMID: 37435045 PMCID: PMC10330732 DOI: 10.3389/fncel.2023.1188306] [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: 03/17/2023] [Accepted: 06/05/2023] [Indexed: 07/13/2023] Open
Abstract
Sleep plays an essential role in all studied animals with a nervous system. However, sleep deprivation leads to various pathological changes and neurobehavioral problems. Astrocytes are the most abundant cells in the brain and are involved in various important functions, including neurotransmitter and ion homeostasis, synaptic and neuronal modulation, and blood-brain barrier maintenance; furthermore, they are associated with numerous neurodegenerative diseases, pain, and mood disorders. Moreover, astrocytes are increasingly being recognized as vital contributors to the regulation of sleep-wake cycles, both locally and in specific neural circuits. In this review, we begin by describing the role of astrocytes in regulating sleep and circadian rhythms, focusing on: (i) neuronal activity; (ii) metabolism; (iii) the glymphatic system; (iv) neuroinflammation; and (v) astrocyte-microglia cross-talk. Moreover, we review the role of astrocytes in sleep deprivation comorbidities and sleep deprivation-related brain disorders. Finally, we discuss potential interventions targeting astrocytes to prevent or treat sleep deprivation-related brain disorders. Pursuing these questions would pave the way for a deeper understanding of the cellular and neural mechanisms underlying sleep deprivation-comorbid brain disorders.
Collapse
Affiliation(s)
- Mengxin Que
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yujuan Li
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Wang
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Gaofeng Zhan
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiao Luo
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Zhou
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
73
|
Purushotham SS, Buskila Y. Astrocytic modulation of neuronal signalling. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1205544. [PMID: 37332623 PMCID: PMC10269688 DOI: 10.3389/fnetp.2023.1205544] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/18/2023] [Indexed: 06/20/2023]
Abstract
Neuronal signalling is a key element in neuronal communication and is essential for the proper functioning of the CNS. Astrocytes, the most prominent glia in the brain play a key role in modulating neuronal signalling at the molecular, synaptic, cellular, and network levels. Over the past few decades, our knowledge about astrocytes and their functioning has evolved from considering them as merely a brain glue that provides structural support to neurons, to key communication elements. Astrocytes can regulate the activity of neurons by controlling the concentrations of ions and neurotransmitters in the extracellular milieu, as well as releasing chemicals and gliotransmitters that modulate neuronal activity. The aim of this review is to summarise the main processes through which astrocytes are modulating brain function. We will systematically distinguish between direct and indirect pathways in which astrocytes affect neuronal signalling at all levels. Lastly, we will summarize pathological conditions that arise once these signalling pathways are impaired focusing on neurodegeneration.
Collapse
Affiliation(s)
| | - Yossi Buskila
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
- The MARCS Institute, Western Sydney University, Campbelltown, NSW, Australia
| |
Collapse
|
74
|
Patton AP, Morris EL, McManus D, Wang H, Li Y, Chin JW, Hastings MH. Astrocytic control of extracellular GABA drives circadian timekeeping in the suprachiasmatic nucleus. Proc Natl Acad Sci U S A 2023; 120:e2301330120. [PMID: 37186824 PMCID: PMC10214171 DOI: 10.1073/pnas.2301330120] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
The hypothalamic suprachiasmatic nucleus (SCN) is the master mammalian circadian clock. Its cell-autonomous timing mechanism, a transcriptional/translational feedback loop (TTFL), drives daily peaks of neuronal electrical activity, which in turn control circadian behavior. Intercellular signals, mediated by neuropeptides, synchronize and amplify TTFL and electrical rhythms across the circuit. SCN neurons are GABAergic, but the role of GABA in circuit-level timekeeping is unclear. How can a GABAergic circuit sustain circadian cycles of electrical activity, when such increased neuronal firing should become inhibitory to the network? To explore this paradox, we show that SCN slices expressing the GABA sensor iGABASnFR demonstrate a circadian oscillation of extracellular GABA ([GABA]e) that, counterintuitively, runs in antiphase to neuronal activity, with a prolonged peak in circadian night and a pronounced trough in circadian day. Resolving this unexpected relationship, we found that [GABA]e is regulated by GABA transporters (GATs), with uptake peaking during circadian day, hence the daytime trough and nighttime peak. This uptake is mediated by the astrocytically expressed transporter GAT3 (Slc6a11), expression of which is circadian-regulated, being elevated in daytime. Clearance of [GABA]e in circadian day facilitates neuronal firing and is necessary for circadian release of the neuropeptide vasoactive intestinal peptide, a critical regulator of TTFL and circuit-level rhythmicity. Finally, we show that genetic complementation of the astrocytic TTFL alone, in otherwise clockless SCN, is sufficient to drive [GABA]e rhythms and control network timekeeping. Thus, astrocytic clocks maintain the SCN circadian clockwork by temporally controlling GABAergic inhibition of SCN neurons.
Collapse
Affiliation(s)
- Andrew P. Patton
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Emma L. Morris
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - David McManus
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Huan Wang
- State Key Laboratory of Membrane Biology, Peking University, School of Life Sciences, 100871Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University, School of Life Sciences, 100871Beijing, China
| | - Jason W. Chin
- PNAC Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Michael H. Hastings
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| |
Collapse
|
75
|
Kim YS, Lee CJ, Kim JH, Kim YB, Colwell CS, Kim YI. Activation of mGluR1 negatively modulates glutamate-induced phase shifts of the circadian pacemaker in the mouse suprachiasmatic nucleus. Neurobiol Sleep Circadian Rhythms 2023; 14:100089. [PMID: 36874931 PMCID: PMC9982032 DOI: 10.1016/j.nbscr.2023.100089] [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: 11/17/2022] [Revised: 01/25/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
In mammals, photic information delivered to the suprachiasmatic nucleus (SCN) via the retinohypothalamic tract (RHT) plays a crucial role in synchronizing the master circadian clock located in the SCN to the solar cycle. It is well known that glutamate released from the RHT terminals initiates the synchronizing process by activating ionotropic glutamate receptors (iGluRs) on retinorecipient SCN neurons. The potential role of metabotropic glutamate receptors (mGluRs) in modulating this signaling pathway has received less attention. In this study, using extracellular single-unit recordings in mouse SCN slices, we investigated the possible roles of the Gq/11 protein-coupled mGluRs, mGluR1 and mGluR5, in photic resetting. We found that mGluR1 activation in the early night produced phase advances in neural activity rhythms in the SCN, while activation in the late night produced phase delays. In contrast, mGluR5 activation had no significant effect on the phase of these rhythms. Interestingly, mGluR1 activation antagonized phase shifts induced by glutamate through a mechanism that was dependent upon CaV1.3 L-type voltage-gated Ca2+ channels (VGCCs). While both mGluR1-evoked phase delays and advances were inhibited by knockout (KO) of CaV1.3 L-type VGCCs, different signaling pathways appeared to be involved in mediating these effects, with mGluR1 working via protein kinase G in the early night and via protein kinase A signaling in the late night. We conclude that, in the mouse SCN, mGluR1s function to negatively modulate glutamate-evoked phase shifts.
Collapse
Affiliation(s)
- Yoon Sik Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul, 136-705, Republic of Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Ji-Hyeon Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul, 136-705, Republic of Korea
| | - Young-Beom Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul, 136-705, Republic of Korea
| | - Christopher S Colwell
- Department of Psychiatry & Biobehavioral Sciences, University of California-Los Angeles, 760 Westwood Plaza, Los Angeles, CA, 90024, USA
| | - Yang In Kim
- Department of Physiology and Neuroscience Research Institute, Korea University College of Medicine, Seoul, 136-705, Republic of Korea
| |
Collapse
|
76
|
Wright CJ, Milosavljevic S, Pocivavsek A. The stress of losing sleep: Sex-specific neurobiological outcomes. Neurobiol Stress 2023; 24:100543. [PMID: 37252645 PMCID: PMC10209346 DOI: 10.1016/j.ynstr.2023.100543] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/20/2023] [Accepted: 05/06/2023] [Indexed: 05/31/2023] Open
Abstract
Sleep is a vital and evolutionarily conserved process, critical to daily functioning and homeostatic balance. Losing sleep is inherently stressful and leads to numerous detrimental physiological outcomes. Despite sleep disturbances affecting everyone, women and female rodents are often excluded or underrepresented in clinical and pre-clinical studies. Advancing our understanding of the role of biological sex in the responses to sleep loss stands to greatly improve our ability to understand and treat health consequences of insufficient sleep. As such, this review discusses sex differences in response to sleep deprivation, with a focus on the sympathetic nervous system stress response and activation of the hypothalamic-pituitary-adrenal (HPA) axis. We review sex differences in several stress-related consequences of sleep loss, including inflammation, learning and memory deficits, and mood related changes. Focusing on women's health, we discuss the effects of sleep deprivation during the peripartum period. In closing, we present neurobiological mechanisms, including the contribution of sex hormones, orexins, circadian timing systems, and astrocytic neuromodulation, that may underlie potential sex differences in sleep deprivation responses.
Collapse
Affiliation(s)
- Courtney J. Wright
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Snezana Milosavljevic
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
77
|
Delgado L, Navarrete M. Shining the Light on Astrocytic Ensembles. Cells 2023; 12:1253. [PMID: 37174653 PMCID: PMC10177371 DOI: 10.3390/cells12091253] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
While neurons have traditionally been considered the primary players in information processing, the role of astrocytes in this mechanism has largely been overlooked due to experimental constraints. In this review, we propose that astrocytic ensembles are active working groups that contribute significantly to animal conduct and suggest that studying the maps of these ensembles in conjunction with neurons is crucial for a more comprehensive understanding of behavior. We also discuss available methods for studying astrocytes and argue that these ensembles, complementarily with neurons, code and integrate complex behaviors, potentially specializing in concrete functions.
Collapse
Affiliation(s)
| | - Marta Navarrete
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain
| |
Collapse
|
78
|
Acharyya D, Cooper J, Prosser RA. Ex vivo comparative investigation of suprachiasmatic nucleus excitotoxic resiliency. F1000Res 2023; 11:1242. [PMID: 39931657 PMCID: PMC11809682 DOI: 10.12688/f1000research.125332.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/06/2023] [Indexed: 02/13/2025] Open
Abstract
Background: Glutamate signaling in the brain is regulated by release, reuptake, and receptor responsiveness. In diseased conditions, glutamate signaling can exceed normal regulatory processes, giving rise to a condition called excitotoxicity. Although regional differences in the excitotoxic effects of glutamate in the brain have been reported, the extent and characteristics of these potential differences are not clear. Here we compared the excitotoxic resiliency of the suprachiasmatic nucleus (SCN), anterior hypothalamus (AH) and cortex. Methods: We treated acute brain slices containing either the SCN and AH or the cortex from adult male mice at different times across the diurnal cycle with varying concentrations of N-methyl-D-aspartate (NMDA), NMDA+ α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) or control medium. The extent of cell damage was assessed using propidium iodide (PI), a cell death marker. Results: The results indicate that all three brain regions exhibited increasing cell damage/death when treated with increasing concentrations of NMDA. However, higher concentrations of NMDA were needed to significantly increase cell damage in the SCN compared to the cortex and AH. All three brain regions also exhibited greater cell death/damage when treated in the nighttime compared to the daytime, although the SCN exhibited increased cell death during a more restricted time interval compared to the AH and cortex. Conclusions: Together, these data confirm previous studies showing excitotoxic resiliency in the SCN, while extending them in two ways. First, we demonstrate a dose-dependency in excitotoxic susceptibility that differentiates the SCN from the surrounding AH and the cortex using a brain slice preparation. Second, we demonstrate a diurnal rhythm in excitotoxic susceptibility with a broadly similar phase across all three brain regions. These data increase our understanding of the extent and nature of the SCN excitotoxic resiliency, which will inform future studies on the cellular mechanisms underlying this phenomenon.
Collapse
Affiliation(s)
- Debalina Acharyya
- Biochemistry & Cellular and Molecular Biology, University of Tennessee Knoxville, Knoxville, TN, 37996, USA
| | - Joanna Cooper
- The Center for Vascular and Inflammatory Disease, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Rebecca A. Prosser
- Biochemistry & Cellular and Molecular Biology, University of Tennessee Knoxville, Knoxville, TN, 37996, USA
| |
Collapse
|
79
|
Lehmann M, Haury K, Oster H, Astiz M. Circadian glucocorticoids throughout development. Front Neurosci 2023; 17:1165230. [PMID: 37179561 PMCID: PMC10166844 DOI: 10.3389/fnins.2023.1165230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/06/2023] [Indexed: 05/15/2023] Open
Abstract
Glucocorticoids (GCs) are essential drivers of mammalian tissue growth and maturation during one of the most critical developmental windows, the perinatal period. The developing circadian clock is shaped by maternal GCs. GC deficits, excess, or exposure at the wrong time of day leads to persisting effects later in life. During adulthood, GCs are one of the main hormonal outputs of the circadian system, peaking at the beginning of the active phase (i.e., the morning in humans and the evening in nocturnal rodents) and contributing to the coordination of complex functions such as energy metabolism and behavior, across the day. Our article discusses the current knowledge on the development of the circadian system with a focus on the role of GC rhythm. We explore the bidirectional interaction between GCs and clocks at the molecular and systemic levels, discuss the evidence of GC influence on the master clock in the suprachiasmatic nuclei (SCN) of the hypothalamus during development and in the adult system.
Collapse
Affiliation(s)
- Marianne Lehmann
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | - Katharina Haury
- Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Leioa, Spain
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | - Mariana Astiz
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
- Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
80
|
Zhang Y, Pan J, Liu Y, Zhang X, Cheng K. Effects of Ficus pandurata Hance var. angustifolia Cheng Flavonoids on Intestinal Barrier and Cognitive Function by Regulating Intestinal Microbiota. Foods 2023; 12:foods12081682. [PMID: 37107477 PMCID: PMC10137925 DOI: 10.3390/foods12081682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/25/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
More and more evidence has supported the interaction between circadian rhythms and intestinal microbes, which provides new insights into how dietary nutrition can improve host health. Our research showed that Ficus pandurata Hance var. angustifolia Cheng flavonoids (FCF) ameliorated the pathological damage of colon and abnormal intestinal microflora structure in mice with circadian clock disorder and improved their exploration and memory behaviors. Mechanism studies have shown that FCF is involved in regulating metabolic pathways and related metabolites, regulating the expression of related tight junction proteins in the colon and the levels of Aβ and inflammatory factors in the hippocampus. Further analysis found that these metabolites showed a certain correlation with intestinal flora and played a certain role in alleviating intestinal physiological damage and cognitive decline.
Collapse
Affiliation(s)
- Yuting Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Junjie Pan
- Chemical Biology Center, Lishui Institute of Agriculture and Forestry Sciences, Lishui 323000, China
| | - Yanan Liu
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Kejun Cheng
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China
- Chemical Biology Center, Lishui Institute of Agriculture and Forestry Sciences, Lishui 323000, China
| |
Collapse
|
81
|
Ahern J, Chrobok Ł, Champneys AR, Piggins HD. A new phase model of the spatiotemporal relationships between three circadian oscillators in the brainstem. Sci Rep 2023; 13:5480. [PMID: 37016055 PMCID: PMC10073201 DOI: 10.1038/s41598-023-32315-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/25/2023] [Indexed: 04/06/2023] Open
Abstract
Analysis of ex vivo Per2 bioluminescent rhythm previously recorded in the mouse dorsal vagal complex reveals a characteristic phase relationship between three distinct circadian oscillators. These signals represent core clock gene expression in the area postrema (AP), the nucleus of the solitary tract (NTS) and the ependymal cells surrounding the 4th ventricle (4Vep). Initially, the data suggests a consistent phasing in which the AP peaks first, followed shortly by the NTS, with the 4Vep peaking 8-9 h later. Wavelet analysis reveals that this pattern is not consistently maintained throughout a recording, however, the phase dynamics strongly imply that oscillator interactions are present. A simple phase model of the three oscillators is developed and it suggests that realistic phase dynamics occur between three model oscillators with coupling close to a synchronisation transition. The coupling topology suggests that the AP bidirectionally communicates phase information to the NTS and the 4Vep to synchronise the three structures. A comparison of the model with previous experimental manipulations demonstrates its feasibility to explain DVC circadian phasing. Finally, we show that simulating steadily decaying coupling improves the model's ability to capture experimental phase dynamics.
Collapse
Affiliation(s)
- Jake Ahern
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
- Engineering Mathematics, University of Bristol, Bristol, BS8 1TW, UK
| | - Łukasz Chrobok
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Alan R Champneys
- Engineering Mathematics, University of Bristol, Bristol, BS8 1TW, UK
| | - Hugh D Piggins
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
82
|
Murakami A, Tsuji K, Isoda M, Matsuo M, Abe Y, Yasui M, Okamura H, Tominaga K. Prolonged Light Exposure Induces Circadian Impairment in Aquaporin-4-Knockout Mice. J Biol Rhythms 2023; 38:208-214. [PMID: 36694941 DOI: 10.1177/07487304221146242] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Astrocytes are densely present in the suprachiasmatic nucleus (SCN), which is the master circadian oscillator in mammals, and are presumed to play a key role in circadian oscillation. However, specific astrocytic molecules that regulate the circadian clock are not yet well understood. In our study, we found that the water channel aquaporin-4 (AQP4) was abundantly expressed in SCN astrocytes, and we further examined its circadian role using AQP4-knockout mice. There was no prominent difference in circadian behavioral rhythms between Aqp4-/- and Aqp4+/+ mice subjected to light-dark cycles and constant dark conditions. However, exposure to constant light induced a greater decrease in the Aqp4-/- mice rhythmicity. Although the damped rhythm in long-term constant light recovered after transfer to constant dark conditions in both genotypes, the period until the reappearance of original rhythmicity was severely prolonged in Aqp4-/- mice. In conclusion, AQP4 absence exacerbates the prolonged light-induced impairment of circadian oscillations and delays their recovery to normal rhythmicity.
Collapse
Affiliation(s)
- Atsumi Murakami
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Kouki Tsuji
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Japan
| | - Minako Isoda
- Graduate School of Science, Kyoto University, Sakyo-ku, Japan
| | - Masahiro Matsuo
- Department of Psychiatry, Shiga University Graduate School of Medicine, Otsu, Japan
| | - Yoichiro Abe
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
- Keio University Global Research Institute, Center for Water Biology and Medicine, Tokyo, Japan
| | - Masato Yasui
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
- Keio University Global Research Institute, Center for Water Biology and Medicine, Tokyo, Japan
| | - Hitoshi Okamura
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Japan
- Department of Neurobiology, Graduate School of Medicine, Kyoto University, Sakyō-ku, Japan
| | - Keiko Tominaga
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
83
|
Brécier A, Li VW, Smith CS, Halievski K, Ghasemlou N. Circadian rhythms and glial cells of the central nervous system. Biol Rev Camb Philos Soc 2023; 98:520-539. [PMID: 36352529 DOI: 10.1111/brv.12917] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/17/2022] [Accepted: 10/25/2022] [Indexed: 11/12/2022]
Abstract
Glial cells are the most abundant cells in the central nervous system and play crucial roles in neural development, homeostasis, immunity, and conductivity. Over the past few decades, glial cell activity in mammals has been linked to circadian rhythms, the 24-h chronobiological clocks that regulate many physiological processes. Indeed, glial cells rhythmically express clock genes that cell-autonomously regulate glial function. In addition, recent findings in rodents have revealed that disruption of the glial molecular clock could impact the entire organism. In this review, we discuss the impact of circadian rhythms on the function of the three major glial cell types - astrocytes, microglia, and oligodendrocytes - across different locations within the central nervous system. We also review recent evidence uncovering the impact of glial cells on the body's circadian rhythm. Together, this sheds new light on the involvement of glial clock machinery in various diseases.
Collapse
Affiliation(s)
- Aurélie Brécier
- Pain Chronobiology & Neuroimmunology Laboratory, Queen's University, Botterell Hall, room 754, Kingston, ON, K7L 3N6, Canada
- Department of Biomedical & Molecular Sciences, 18 Stuart Street, Kingston, ON, K7L 3N6, Canada
| | - Vina W Li
- Pain Chronobiology & Neuroimmunology Laboratory, Queen's University, Botterell Hall, room 754, Kingston, ON, K7L 3N6, Canada
- Department of Biomedical & Molecular Sciences, 18 Stuart Street, Kingston, ON, K7L 3N6, Canada
| | - Chloé S Smith
- Pain Chronobiology & Neuroimmunology Laboratory, Queen's University, Botterell Hall, room 754, Kingston, ON, K7L 3N6, Canada
- Department of Biomedical & Molecular Sciences, 18 Stuart Street, Kingston, ON, K7L 3N6, Canada
| | - Katherine Halievski
- Pain Chronobiology & Neuroimmunology Laboratory, Queen's University, Botterell Hall, room 754, Kingston, ON, K7L 3N6, Canada
| | - Nader Ghasemlou
- Pain Chronobiology & Neuroimmunology Laboratory, Queen's University, Botterell Hall, room 754, Kingston, ON, K7L 3N6, Canada
- Department of Biomedical & Molecular Sciences, 18 Stuart Street, Kingston, ON, K7L 3N6, Canada
- Department of Anesthesiology & Perioperative Medicine, 76 Stuart Street, Kingston, ON, K7L 2V7, Canada
- Centre for Neuroscience Studies, Queen's University, 18 Stuart Street, Kingston, ON, K7L 3N6, Canada
| |
Collapse
|
84
|
Giantomasi L, Ribeiro JF, Barca-Mayo O, Malerba M, Miele E, De Pietri Tonelli D, Berdondini L. Astrocytes actively support long-range molecular clock synchronization of segregated neuronal populations. Sci Rep 2023; 13:4815. [PMID: 36964220 PMCID: PMC10038999 DOI: 10.1038/s41598-023-31966-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 03/20/2023] [Indexed: 03/26/2023] Open
Abstract
In mammals, the suprachiasmatic nucleus of the hypothalamus is the master circadian pacemaker that synchronizes the clocks in the central nervous system and periphery, thus orchestrating rhythms throughout the body. However, little is known about how so many cellular clocks within and across brain circuits can be effectively synchronized. In this work, we investigated the implication of two possible pathways: (i) astrocytes-mediated synchronization and (ii) neuronal paracrine factors-mediated synchronization. By taking advantage of a lab-on-a-chip microfluidic device developed in our laboratory, here we report that both pathways are involved. We found the paracrine factors-mediated synchronization of molecular clocks is diffusion-limited and, in our device, effective only in case of a short distance between neuronal populations. Interestingly, interconnecting astrocytes define an active signaling channel that can synchronize molecular clocks of neuronal populations also at longer distances. At mechanism level, we found that astrocytes-mediated synchronization involves both GABA and glutamate, while neuronal paracrine factors-mediated synchronization occurs through GABA signaling. These findings identify a previously unknown role of astrocytes as active cells that might distribute long-range signals to synchronize the brain clocks, thus further strengthening the importance of reciprocal interactions between glial and neuronal cells in the context of circadian circuitry.
Collapse
Affiliation(s)
- Lidia Giantomasi
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano Di Tecnologia (IIT), 16163, Genova, Italy
| | - João F Ribeiro
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano Di Tecnologia (IIT), 16163, Genova, Italy
| | - Olga Barca-Mayo
- Neurobiology of miRNA, Fondazione Istituto Italiano Di Tecnologia (IIT), 16163, Genova, Italy
- Circadian and Glial Biology Lab, Physiology Department, Molecular Medicine, and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Mario Malerba
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano Di Tecnologia (IIT), 16163, Genova, Italy
- Centre de Nanosciences et de Nanotechnologies (C2N), CNRS UMR 9001, Université Paris-Saclay, 91120, Palaiseau, France
| | - Ermanno Miele
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano Di Tecnologia (IIT), 16163, Genova, Italy
- NanoPhotonics Centre, Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | | | - Luca Berdondini
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano Di Tecnologia (IIT), 16163, Genova, Italy.
| |
Collapse
|
85
|
Vaughen JP, Theisen E, Clandinin TR. From seconds to days: Neural plasticity viewed through a lipid lens. Curr Opin Neurobiol 2023; 80:102702. [PMID: 36965206 DOI: 10.1016/j.conb.2023.102702] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/31/2023] [Accepted: 02/16/2023] [Indexed: 03/27/2023]
Abstract
Many adult neurons are dynamically remodeled across timescales ranging from the rapid addition and removal of specific synaptic connections, to largescale structural plasticity events that reconfigure circuits over hours, days, and months. Membrane lipids, including brain-enriched sphingolipids, play crucial roles in these processes. In this review, we summarize progress at the intersection of neuronal activity, lipids, and structural remodeling. We highlight how brain activity modulates lipid metabolism to enable adaptive structural plasticity, and showcase glia as key players in membrane remodeling. These studies reveal that lipids act as critical signaling molecules that instruct the dynamic architecture of the brain.
Collapse
Affiliation(s)
- John P Vaughen
- Department of Neurobiology, Stanford University, Stanford, CA, 94305, United States; Department of Developmental Biology, Stanford University, Stanford, CA, 94305, United States. https://twitter.com/gliaful
| | - Emma Theisen
- Department of Neurobiology, Stanford University, Stanford, CA, 94305, United States. https://twitter.com/emmaktheisen
| | - Thomas R Clandinin
- Department of Neurobiology, Stanford University, Stanford, CA, 94305, United States.
| |
Collapse
|
86
|
De Luca SN, Chan SMH, Dobric A, Wang H, Seow HJ, Brassington K, Mou K, Alateeq R, Akhtar A, Bozinovski S, Vlahos R. Cigarette smoke-induced pulmonary impairment is associated with social recognition memory impairments and alterations in microglial profiles within the suprachiasmatic nucleus of the hypothalamus. Brain Behav Immun 2023; 109:292-307. [PMID: 36775074 DOI: 10.1016/j.bbi.2023.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/29/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major, incurable respiratory condition that is primarily caused by cigarette smoking (CS). Neurocognitive disorders including cognitive dysfunction, anxiety and depression are highly prevalent in people with COPD. It is understood that increased lung inflammation and oxidative stress from CS exposure may 'spill over' into the systemic circulation to promote the onset of these extra-pulmonary comorbidities, and thus impacts the quality of life of people with COPD. The precise role of the 'spill-over' of inflammation and oxidative stress in the onset of COPD-related neurocognitive disorders are unclear. The present study investigated the impact of chronic CS exposure on anxiety-like behaviors and social recognition memory, with a particular focus on the role of the 'spill-over' of inflammation and oxidative stress from the lungs. Adult male BALB/c mice were exposed to either room air (sham) or CS (9 cigarettes per day, 5 days a week) for 24 weeks and were either daily co-administered with the NOX2 inhibitor, apocynin (5 mg/kg, in 0.01 % DMSO diluted in saline, i.p.) or vehicle (0.01 % DMSO in saline) one hour before the initial CS exposure of the day. After 23 weeks, mice underwent behavioral testing and physiological diurnal rhythms were assessed by monitoring diurnal regulation profiles. Lungs were collected and assessed for hallmark features of COPD. Consistent with its anti-inflammatory and oxidative stress properties, apocynin treatment partially lessened lung inflammation and lung function decline in CS mice. CS-exposed mice displayed marked anxiety-like behavior and impairments in social recognition memory compared to sham mice, which was prevented by apocynin treatment. Apocynin was unable to restore the decreased Bmal1-positive cells, key in cells in diurnal regulation, in the suprachiasmatic nucleus of the hypothalamus to that of sham levels. CS-exposed mice treated with apocynin was associated with a restoration of microglial area per cell and basal serum corticosterone. This data suggests that we were able to model the CS-induced social recognition memory impairments seen in humans with COPD. The preventative effects of apocynin on memory impairments may be via a microglial dependent mechanism.
Collapse
Affiliation(s)
- Simone N De Luca
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Stanley M H Chan
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Aleksandar Dobric
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Hao Wang
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Huei Jiunn Seow
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Kurt Brassington
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Kevin Mou
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Rana Alateeq
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Alina Akhtar
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Steven Bozinovski
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Ross Vlahos
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia.
| |
Collapse
|
87
|
Chu Y, He H, Liu Q, Jia S, Fan W, Huang F. The Circadian Clocks, Oscillations of Pain-Related Mediators, and Pain. Cell Mol Neurobiol 2023; 43:511-523. [PMID: 35179680 PMCID: PMC11415172 DOI: 10.1007/s10571-022-01205-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/06/2022] [Indexed: 01/07/2023]
Abstract
The circadian clock is a biochemical oscillator that is synchronized with solar time. Normal circadian rhythms are necessary for many physiological functions. Circadian rhythms have also been linked with many physiological functions, several clinical symptoms, and diseases. Accumulating evidence suggests that the circadian clock appears to modulate the processing of nociceptive information. Many pain conditions display a circadian fluctuation pattern clinically. Thus, the aim of this review is to summarize the existing knowledge about the circadian clocks involved in diurnal rhythms of pain. Possible cellular and molecular mechanisms regarding the connection between the circadian clocks and pain are discussed.
Collapse
Affiliation(s)
- Yanhao Chu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen Uni-versity, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Hongwen He
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Qing Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen Uni-versity, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Shilin Jia
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen Uni-versity, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Wenguo Fan
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
| | - Fang Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen Uni-versity, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
| |
Collapse
|
88
|
Granados-Fuentes D, Cho K, Patti GJ, Costa R, Herzog ED, Montagnese S. Hyperammonaemia disrupts daily rhythms reversibly by elevating glutamate in the central circadian pacemaker. Liver Int 2023; 43:673-683. [PMID: 36367321 PMCID: PMC9974605 DOI: 10.1111/liv.15476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/21/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
Patients with cirrhosis exhibit features of circadian disruption. Hyperammonaemia has been suggested to impair both homeostatic and circadian sleep regulation. Here, we tested if hyperammonaemia directly disrupts circadian rhythm generation in the central pacemaker, the suprachiasmatic nuclei (SCN) of the hypothalamus. Wheel-running activity was recorded from mice fed with a hyperammonaemic or normal diet for ~35 days in a 12:12 light-dark (LD) cycle followed by ~15 days in constant darkness (DD). The expression of the clock protein PERIOD2 (PER2) was recorded from SCN explants before, during and after ammonia exposure, ±glutamate receptor antagonists. In LD, hyperammonaemic mice advanced their daily activity onset time by ~1 h (16.8 ± 0.3 vs. 18.1 ± 0.04 h, p = .009) and decreased their total activity, concentrating it during the first half of the night. In DD, hyperammonaemia reduced the amplitude of daily activity (551.5 ± 27.7 vs. 724.9 ± 59 counts, p = .007), with no changes in circadian period. Ammonia (≥0.01 mM) rapidly and significantly reduced PER2 amplitude, and slightly increased circadian period. The decrease in PER2 amplitude correlated with decreased synchrony among circadian cells in the SCN and increased extracellular glutamate, which was rescued by AMPA glutamate receptor antagonists. These data suggest that hyperammonaemia affects circadian regulation of rest-activity behaviour by increasing extracellular glutamate in the SCN.
Collapse
Affiliation(s)
| | - Kevin Cho
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, USA
| | - Gary J. Patti
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, USA
| | - Rodolfo Costa
- Department of Biology, University of Padova, Padova, Italy
- Institute of Neuroscience, National Research Council of Italy (CNR), Padova, Italy
- Chronobiology Section, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Erik D. Herzog
- Biology Department, Washington University in St. Louis, USA
| | - Sara Montagnese
- Chronobiology Section, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
- Department of Medicine, University of Padova, Italy
| |
Collapse
|
89
|
Brain Waste Removal System and Sleep: Photobiomodulation as an Innovative Strategy for Night Therapy of Brain Diseases. Int J Mol Sci 2023; 24:ijms24043221. [PMID: 36834631 PMCID: PMC9965491 DOI: 10.3390/ijms24043221] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Emerging evidence suggests that an important function of the sleeping brain is the removal of wastes and toxins from the central nervous system (CNS) due to the activation of the brain waste removal system (BWRS). The meningeal lymphatic vessels (MLVs) are an important part of the BWRS. A decrease in MLV function is associated with Alzheimer's and Parkinson's diseases, intracranial hemorrhages, brain tumors and trauma. Since the BWRS is activated during sleep, a new idea is now being actively discussed in the scientific community: night stimulation of the BWRS might be an innovative and promising strategy for neurorehabilitation medicine. This review highlights new trends in photobiomodulation of the BWRS/MLVs during deep sleep as a breakthrough technology for the effective removal of wastes and unnecessary compounds from the brain in order to increase the neuroprotection of the CNS as well as to prevent or delay various brain diseases.
Collapse
|
90
|
Shigetomi E, Koizumi S. The role of astrocytes in behaviors related to emotion and motivation. Neurosci Res 2023; 187:21-39. [PMID: 36181908 DOI: 10.1016/j.neures.2022.09.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 10/14/2022]
Abstract
Astrocytes are present throughout the brain and intimately interact with neurons and blood vessels. Three decades of research have shown that astrocytes reciprocally communicate with neurons and other non-neuronal cells in the brain and dynamically regulate cell function. Astrocytes express numerous receptors for neurotransmitters, neuromodulators, and cytokines and receive information from neurons, other astrocytes, and other non-neuronal cells. Among those receptors, the main focus has been G-protein coupled receptors. Activation of G-protein coupled receptors leads to dramatic changes in intracellular signaling (Ca2+ and cAMP), which is considered a form of astrocyte activity. Methodological improvements in measurement and manipulation of astrocytes have advanced our understanding of the role of astrocytes in circuits and have begun to reveal unexpected functions of astrocytes in behavior. Recent studies have suggested that astrocytic activity regulates behavior flexibility, such as coping strategies for stress exposure, and plays an important role in behaviors related to emotion and motivation. Preclinical evidence suggests that impairment of astrocytic function contributes to psychiatric diseases, especially major depression. Here, we review recent progress on the role of astrocytes in behaviors related to emotion and motivation.
Collapse
Affiliation(s)
- Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Japan; Yamanashi GLIA Center, Graduate School of Medical Science, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Japan.
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Japan; Yamanashi GLIA Center, Graduate School of Medical Science, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Japan.
| |
Collapse
|
91
|
Silva-Parra J, Sandu C, Felder-Schmittbuhl MP, Hernández-Kelly LC, Ortega A. Aryl Hydrocarbon Receptor in Glia Cells: A Plausible Glutamatergic Neurotransmission Orchestrator. Neurotox Res 2023; 41:103-117. [PMID: 36607593 DOI: 10.1007/s12640-022-00623-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/23/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023]
Abstract
Glutamate is the major excitatory amino acid in the vertebrate brain. Glutamatergic signaling is involved in most of the central nervous system functions. Its main components, namely receptors, ion channels, and transporters, are tightly regulated at the transcriptional, translational, and post-translational levels through a diverse array of extracellular signals, such as food, light, and neuroactive molecules. An exquisite and well-coordinated glial/neuronal bidirectional communication is required for proper excitatory amino acid signal transactions. Biochemical shuttles such as the glutamate/glutamine and the astrocyte-neuronal lactate represent the fundamental involvement of glial cells in glutamatergic transmission. In fact, the disruption of any of these coordinated biochemical intercellular cascades leads to an excitotoxic insult that underlies some aspects of most of the neurodegenerative diseases characterized thus far. In this contribution, we provide a comprehensive summary of the involvement of the Aryl hydrocarbon receptor, a ligand-dependent transcription factor in the gene expression regulation of glial glutamate transporters. These receptors might serve as potential targets for the development of novel strategies for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Janisse Silva-Parra
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México
| | - Cristina Sandu
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Marie-Paule Felder-Schmittbuhl
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Luisa C Hernández-Kelly
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México.
| |
Collapse
|
92
|
Clyburn C, Carson KE, Smith CR, Travagli RA, Browning KN. Brainstem astrocytes control homeostatic regulation of caloric intake. J Physiol 2023; 601:801-829. [PMID: 36696965 PMCID: PMC10026361 DOI: 10.1113/jp283566] [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: 07/06/2022] [Accepted: 12/08/2022] [Indexed: 01/27/2023] Open
Abstract
Prolonged high-fat diet (HFD) exposure is associated with hyperphagia, excess caloric intake and weight gain. After initial exposure to a HFD, a brief (24-48 h) period of hyperphagia is followed by the regulation of caloric intake and restoration of energy balance within an acute (3-5 day) period. Previous studies have demonstrated this occurs via a vagally mediated signalling cascade that increases glutamatergic transmission via activation of NMDA receptors located on gastric-projecting neurons of the dorsal motor nucleus of the vagus (DMV). The present study used electrophysiological recordings from thin brainstem slice preparations, in vivo recordings of gastric motility and tone, measurement of gastric emptying rates, and food intake studies to investigate the hypothesis that activation of brainstem astrocytes in response to acute HFD exposure is responsible for the increased glutamatergic drive to DMV neurons and the restoration of caloric balance. Pharmacological and chemogenetic inhibition of brainstem astrocytes reduced glutamatergic signalling and DMV excitability, dysregulated gastric tone and motility, attenuated the homeostatic delay in gastric emptying, and prevented the decrease in food intake that is observed during the period of energy regulation following initial exposure to HFD. Understanding the mechanisms involved in caloric regulation may provide critical insights into energy balance as well as into the hyperphagia that develops as these mechanisms are overcome. KEY POINTS: Initial exposure to a high fat diet is associated with a brief period of hyperphagia before caloric intake and energy balance is restored. This period of homeostatic regulation is associated with a vagally mediated signalling cascade that increases glutamatergic transmission to dorsal motor nucleus of the vagus (DMV) neurons via activation of synaptic NMDA receptors. The present study demonstrates that pharmacological and chemogenetic inhibition of brainstem astrocytes reduced glutamatergic signalling and DMV neuronal excitability, dysregulated gastric motility and tone and emptying, and prevented the regulation of food intake following high-fat diet exposure. Astrocyte regulation of glutamatergic transmission to DMV neurons appears to involve release of the gliotransmitters glutamate and ATP. Understanding the mechanisms involved in caloric regulation may provide critical insights into energy balance as well as into the hyperphagia that develops as these mechanisms are overcome.
Collapse
Affiliation(s)
- Courtney Clyburn
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
- Current position: Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, 97056
| | - Kaitlin E. Carson
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
| | - Caleb R. Smith
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
| | - R. Alberto Travagli
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
- Current position: Neurobiology Research, Newport, NC 28570
| | - Kirsteen N. Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
| |
Collapse
|
93
|
Guzmán-Ruiz MA, Guerrero-Vargas NN, Lagunes-Cruz A, González-González S, García-Aviles JE, Hurtado-Alvarado G, Mendez-Hernández R, Chavarría-Krauser A, Morin JP, Arriaga-Avila V, Buijs RM, Guevara-Guzmán R. Circadian modulation of microglial physiological processes and immune responses. Glia 2023; 71:155-167. [PMID: 35971989 PMCID: PMC10087862 DOI: 10.1002/glia.24261] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/11/2022] [Accepted: 08/01/2022] [Indexed: 11/09/2022]
Abstract
Microglia is considered the central nervous system (CNS) resident macrophages that establish an innate immune response against pathogens and toxins. However, the recent studies have shown that microglial gene and protein expression follows a circadian pattern; several immune activation markers and clock genes are expressed rhythmically without the need for an immune stimulus. Furthermore, microglia responds to an immune challenge with different magnitudes depending on the time of the day. This review examines the circadian control of microglia function and the possible physiological implications. For example, we discuss that synaptic prune is performed in the cortex at a certain moment of the day. We also consider the implications of daily microglial function for maintaining biological rhythms like general activity, body temperature, and food intake. We conclude that the developmental stage, brain region, and pathological state are not the only factors to consider for the evaluation of microglial functions; instead, emerging evidence indicates that circadian time as an essential aspect for a better understanding of the role of microglia in CNS physiology.
Collapse
Affiliation(s)
- Mara A Guzmán-Ruiz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Natalí N Guerrero-Vargas
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Alejandra Lagunes-Cruz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Shellye González-González
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Jesús Enrique García-Aviles
- Área de Neurociencias, Departamento de Biología de la Reproducción, Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, México City, Mexico
| | | | - Rebeca Mendez-Hernández
- Instituto Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, Mexico
| | - Anahí Chavarría-Krauser
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Jean-Pascal Morin
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Virginia Arriaga-Avila
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Ruud M Buijs
- Instituto Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, Mexico
| | - Rosalinda Guevara-Guzmán
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| |
Collapse
|
94
|
Vasciaveo V, Iadarola A, Casile A, Dante D, Morello G, Minotta L, Tamagno E, Cicolin A, Guglielmotto M. Sleep fragmentation affects glymphatic system through the different expression of AQP4 in wild type and 5xFAD mouse models. Acta Neuropathol Commun 2023; 11:16. [PMID: 36653878 PMCID: PMC9850555 DOI: 10.1186/s40478-022-01498-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/18/2022] [Indexed: 01/20/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by genetic and multifactorial risk factors. Many studies correlate AD to sleep disorders. In this study, we performed and validated a mouse model of AD and sleep fragmentation, which properly mimics a real condition of intermittent awakening. We noticed that sleep fragmentation induces a general acceleration of AD progression in 5xFAD mice, while in wild type mice it affects cognitive behaviors in particular learning and memory. Both these events may be correlated to aquaporin-4 (AQP4) modulation, a crucial player of the glymphatic system activity. In particular, sleep fragmentation differentially affects aquaporin-4 channel (AQP4) expression according to the stage of the disease, with an up-regulation in younger animals, while such change cannot be detected in older ones. Moreover, in wild type mice sleep fragmentation affects cognitive behaviors, in particular learning and memory, by compromising the glymphatic system through the decrease of AQP4. Nevertheless, an in-depth study is needed to better understand the mechanism by which AQP4 is modulated and whether it could be considered a risk factor for the disease development in wild type mice. If our hypotheses are going to be confirmed, AQP4 modulation may represent the convergence point between AD and sleep disorder pathogenic mechanisms.
Collapse
Affiliation(s)
- Valeria Vasciaveo
- grid.7605.40000 0001 2336 6580Department of Neuroscience Rita Levi Montalcini, University of Torino, Via Cherasco 15, 10126 Turin, Italy ,grid.7605.40000 0001 2336 6580Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Antonella Iadarola
- grid.432329.d0000 0004 1789 4477Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza, Corso Bramante 88, 10126 Turin, Italy
| | - Antonino Casile
- grid.5602.10000 0000 9745 6549School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 9, 62032 Camerino, MC Italy
| | - Davide Dante
- grid.7605.40000 0001 2336 6580Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Giulia Morello
- grid.7605.40000 0001 2336 6580Department of Neuroscience Rita Levi Montalcini, University of Torino, Via Cherasco 15, 10126 Turin, Italy ,grid.7605.40000 0001 2336 6580Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Lorenzo Minotta
- grid.7605.40000 0001 2336 6580Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Elena Tamagno
- grid.7605.40000 0001 2336 6580Department of Neuroscience Rita Levi Montalcini, University of Torino, Via Cherasco 15, 10126 Turin, Italy ,grid.7605.40000 0001 2336 6580Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Alessandro Cicolin
- grid.7605.40000 0001 2336 6580Department of Neuroscience Rita Levi Montalcini, University of Torino, Via Cherasco 15, 10126 Turin, Italy
| | - Michela Guglielmotto
- grid.7605.40000 0001 2336 6580Department of Neuroscience Rita Levi Montalcini, University of Torino, Via Cherasco 15, 10126 Turin, Italy ,grid.7605.40000 0001 2336 6580Neuroscience Institute of Cavalieri Ottolenghi Foundation (NICO), University of Torino, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| |
Collapse
|
95
|
Xue H, Wu M, Wang Y, Zhao Y, Zhang M, Zhang H. The circadian rhythms regulated by Cx43-signaling in the pathogenesis of Neuromyelitis Optica. Front Immunol 2023; 13:1021703. [PMID: 36726988 PMCID: PMC9885795 DOI: 10.3389/fimmu.2022.1021703] [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: 08/17/2022] [Accepted: 12/29/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction Neuromyelitis Optica (NMO) is an inflammatory demyelinating disease of the central nervous system (CNS). NMO manifests as selective and severe attacks on axons and myelin of the optic nerve and spinal cord, resulting in necrotic cavities. The circadian rhythms are well demonstrated to profoundly impact cellular function, behavior, and disease. This study is aimed to explore the role and molecular basis of circadian rhythms in NMO. Methods We used an Aquaporin 4(AQP4) IgG-induced NMO cell model in isolated astrocytes. The expression of Cx43 and Bmal1 were detected by real-time PCR and Western Blot. TAT-Gap19 and DQP-1105 were used to inhibit Cx43 and glutamate receptor respectively. The knockdown of Bmal1 were performed with the shRNA containing adenovirus. The levels of glutamate, anterior visual pathway (AVP), and vasoactive intestinal peptide (VIP) were quantified by ELISA kits. Results We found that Bmal1 and Clock, two essential components of the circadian clock, were significantly decreased in NMO astrocytes, which were reversed by Cx43 activation (linoleic acid) or glutamate. Moreover, the expression levels of Bmal1 and Clock were also decreased by Cx43 blockade (TAT-Gap19) or glutamate receptor inhibition (DQP-1105). Furthermore, adenovirus-mediated Bmal1 knockdown by shRNA (Ad-sh-Bmal1) dramatically decreased the levels of glutamate, AVP, and VIP from neurons, and significantly down-regulated the protein level of Cx43 in NMO astrocytes with Cx43 activation (linoleic acid) or glutamate treatment. However, Bmal1 knockdown did not alter these levels in normal astrocytes with Cx43 blockade (TAT-Gap19) or glutamate receptor inhibition (DQP-1105). Discussion Collectively, these results suggest that Cx43-glutamate signaling would be a critical upstream regulator that contributes to the NMO-induced rhythmic damage in SCN astrocytes.
Collapse
Affiliation(s)
- Huiru Xue
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China,First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Minghui Wu
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongle Wang
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yunfei Zhao
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Meini Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China,*Correspondence: Meini Zhang, ; Hui Zhang,
| | - Hui Zhang
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China,Department of Medical Imaging, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China,*Correspondence: Meini Zhang, ; Hui Zhang,
| |
Collapse
|
96
|
Schurhoff N, Toborek M. Circadian rhythms in the blood-brain barrier: impact on neurological disorders and stress responses. Mol Brain 2023; 16:5. [PMID: 36635730 PMCID: PMC9835375 DOI: 10.1186/s13041-023-00997-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023] Open
Abstract
Circadian disruption has become more prevalent in society due to the increase in shift work, sleep disruption, blue light exposure, and travel via different time zones. The circadian rhythm is a timed transcription-translation feedback loop with positive regulators, BMAL1 and CLOCK, that interact with negative regulators, CRY and PER, to regulate both the central and peripheral clocks. This review highlights the functions of the circadian rhythm, specifically in the blood-brain barrier (BBB), during both healthy and pathological states. The BBB is a highly selective dynamic interface composed of CNS endothelial cells, astrocytes, pericytes, neurons, and microglia that form the neurovascular unit (NVU). Circadian rhythms modulate BBB integrity through regulating oscillations of tight junction proteins, assisting in functions of the NVU, and modulating transporter functions. Circadian disruptions within the BBB have been observed in stress responses and several neurological disorders, including brain metastasis, epilepsy, Alzheimer's disease, and Parkinson's disease. Further understanding of these interactions may facilitate the development of improved treatment options and preventative measures.
Collapse
Affiliation(s)
- Nicolette Schurhoff
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Suite 528, 1011 NW 15th Street, Miami, FL, 33155, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Suite 528, 1011 NW 15th Street, Miami, FL, 33155, USA.
- Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, 40-065, Katowice, Poland.
| |
Collapse
|
97
|
Zheng Y, Pan L, Wang F, Yan J, Wang T, Xia Y, Yao L, Deng K, Zheng Y, Xia X, Su Z, Chen H, Lin J, Ding Z, Zhang K, Zhang M, Chen Y. Neural function of Bmal1: an overview. Cell Biosci 2023; 13:1. [PMID: 36593479 PMCID: PMC9806909 DOI: 10.1186/s13578-022-00947-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023] Open
Abstract
Bmal1 (Brain and muscle arnt-like, or Arntl) is a bHLH/PAS domain transcription factor central to the transcription/translation feedback loop of the biologic clock. Although Bmal1 is well-established as a major regulator of circadian rhythm, a growing number of studies in recent years have shown that dysfunction of Bmal1 underlies a variety of psychiatric, neurodegenerative-like, and endocrine metabolism-related disorders, as well as potential oncogenic roles. In this review, we systematically summarized Bmal1 expression in different brain regions, its neurological functions related or not to circadian rhythm and biological clock, and pathological phenotypes arising from Bmal1 knockout. This review also discusses oscillation and rhythmicity, especially in the suprachiasmatic nucleus, and provides perspective on future progress in Bmal1 research.
Collapse
Affiliation(s)
- Yuanjia Zheng
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China ,grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingyun Pan
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Feixue Wang
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinglan Yan
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Taiyi Wang
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yucen Xia
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Yao
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kelin Deng
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuqi Zheng
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoye Xia
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhikai Su
- grid.411866.c0000 0000 8848 7685The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong China
| | - Hongjie Chen
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jie Lin
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhenwei Ding
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kaitong Zhang
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meng Zhang
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yongjun Chen
- grid.464402.00000 0000 9459 9325Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China ,grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China ,Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, China
| |
Collapse
|
98
|
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
|
99
|
Gudkov SV, Burmistrov DE, Kondakova EV, Sarimov RM, Yarkov RS, Franceschi C, Vedunova MV. An emerging role of astrocytes in aging/neuroinflammation and gut-brain axis with consequences on sleep and sleep disorders. Ageing Res Rev 2023; 83:101775. [PMID: 36334910 DOI: 10.1016/j.arr.2022.101775] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/05/2022] [Accepted: 10/30/2022] [Indexed: 11/18/2022]
Abstract
Understanding the role of astrocytes in the central nervous system has changed dramatically over the last decade. The accumulating findings indicate that glial cells are involved not only in the maintenance of metabolic and ionic homeostasis and in the implementation of trophic functions but also in cognitive functions and information processing in the brain. Currently, there are some controversies regarding the role of astrocytes in complex processes such as aging of the nervous system and the pathogenesis of age-related neurodegenerative diseases. Many findings confirm the important functional role of astrocytes in age-related brain changes, including sleep disturbance and the development of neurodegenerative diseases and particularly Alzheimer's disease. Until recent years, neurobiological research has focused mainly on neuron-glial interactions, in which individual astrocytes locally modulate neuronal activity and communication between neurons. The review considers the role of astrocytes in the physiology of sleep and as an important "player" in the development of neurodegenerative diseases. In addition, the features of the astrocytic network reorganization during aging are discussed.
Collapse
Affiliation(s)
- Sergey V Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia; Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Dmitriy E Burmistrov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| | - Elena V Kondakova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Ruslan M Sarimov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| | - Roman S Yarkov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Claudio Franceschi
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Maria V Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| |
Collapse
|
100
|
Kamat PK, Khan MB, Smith C, Siddiqui S, Baban B, Dhandapani K, Hess DC. The time dimension to stroke: Circadian effects on stroke outcomes and mechanisms. Neurochem Int 2023; 162:105457. [PMID: 36442686 PMCID: PMC9839555 DOI: 10.1016/j.neuint.2022.105457] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/14/2022] [Accepted: 11/19/2022] [Indexed: 11/26/2022]
Abstract
The circadian system is widely involved in the various pathological outcomes affected by time dimension changes. In the brain, the master circadian clock, also known as the "pacemaker," is present in the hypothalamus's suprachiasmatic nucleus (SCN). The SCN consists of molecular circadian clocks that operate in each neuron and other brain cells. These circadian mechanisms are controlled by the transcription and translation of specific genes such as the clock circadian regulator (Clock) and brain and muscle ARNT-Like 1 (Bmal1). Period (Per1-3) and cryptochrome (Cry1 and 2) negatively feedback and regulate the clock genes. Variations in the circadian cycle and these clock genes can affect stroke outcomes. Studies suggest that the peak stroke occurs in the morning after patients awaken from sleep, while stroke severity and poor outcomes worsen at midnight. The main risk factor associated with stroke is high blood pressure (hypertension). Blood pressure usually dips by 15-20% during sleep, but many hypertensives do not display this normal dipping pattern and are non-dippers. A sleep blood pressure is the primary determinant of stroke risk. This article discusses the possible mechanism associated with circadian rhythm and stroke outcomes.
Collapse
Affiliation(s)
- Pradip K Kamat
- Departments of Neurology, Medical College of Georgia, Augusta University, USA.
| | | | - Cameron Smith
- Departments of Neurology, Medical College of Georgia, Augusta University, USA
| | - Shahneela Siddiqui
- Departments of Neurology, Medical College of Georgia, Augusta University, USA
| | - Babak Baban
- Departments of Oral Biology, Dental College of Georgia, Augusta University, USA
| | - Krishnan Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, USA
| | - David C Hess
- Departments of Neurology, Medical College of Georgia, Augusta University, USA
| |
Collapse
|