1
|
McPartland M, Ashcroft F, Wagner M. Plastic chemicals disrupt molecular circadian rhythms via adenosine 1 receptor in vitro. ENVIRONMENT INTERNATIONAL 2025; 198:109422. [PMID: 40179621 DOI: 10.1016/j.envint.2025.109422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/28/2025] [Accepted: 03/27/2025] [Indexed: 04/05/2025]
Abstract
The adenosine 1 receptor (A1R) is a G protein-coupled receptor that transduces signals to regulate sleep-wake cycles and circadian rhythms. Plastic products contain thousands of chemicals, known to disrupt physiological function. Recent research has demonstrated that some of these chemicals are also A1R agonists, however, the extent to which such activation propagates downstream and results in cellular alterations remains unknown. Thus, we investigate whether chemicals extracted from polyurethane (PUR) and polyvinyl chloride (PVC) plastics disrupt circadian rhythms via agonism of A1R. We confirm that plastic chemicals in both plastics activate A1R and inhibit intracellular cAMP in U2OS cells. Notably, this inhibition is comparable to that induced by the highly specific A1R agonist 2'-MeCCPA. To assess circadian disruption, we quantify temporal expression patterns of the clock genes PER2 and CRY2 at 4-h intervals over 48 h. Here, exposure to plastic chemicals shifts the phase in the oscillatory expression cycles of both clock genes by 9-17 min. Importantly, these effects are dose-dependent and reversible when A1R is inhibited by a pharmacological antagonist. This demonstrates that plastic chemicals can disrupt circadian processes by interfering with A1R signaling and suggests a novel mechanism by which these and other chemicals may contribute to non-communicable diseases.
Collapse
Affiliation(s)
- Molly McPartland
- Department of Biology, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway.
| | - Felicity Ashcroft
- Department of Biology, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Martin Wagner
- Department of Biology, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway.
| |
Collapse
|
2
|
Oh JH, Han YY, Kim EB, Jo HN, Lee JS, Kim BM, Kim JM, Lee YS, Lee DY, Kim KW, Lee I, Lee YW, Park CS, Kim DO. Improvement of sleep disorders through the adenosine A receptor agonist effect of Phlomoides umbrosa Turczaninow root extract in pentobarbital-induced ICR mice. Food Sci Biotechnol 2025; 34:1149-1159. [PMID: 40093550 PMCID: PMC11904018 DOI: 10.1007/s10068-024-01663-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/02/2024] [Accepted: 07/11/2024] [Indexed: 03/19/2025] Open
Abstract
Sleep deprivation is a serious problem that deteriorates the health, quality of life, and social productivity of affected individuals. Research on safe and effective plant-based materials should be performed to help alleviate sleep disorders. Phlomoides umbrosa Turczaninow is a domestic plant in South Korea that has been used traditionally as a medicine to treat sleep disorders. The effects and mechanism of action of a P. umbrosa Turczaninow root extract (PUTRE) prepared using hot water extraction were investigated. ICR mice were injected with pentobarbital to induce sleep. The PUTRE-treated groups had significantly (p < 0.05) decreased sleep latency and increased sleep time compared with the controls. When PUTRE was co-administered with adenosine antagonists (caffeine and 8-cyclopentyl-1,3-dipropylxanthine), it acted specifically on the adenosine A1 receptor, because its sleep-enhancing effect was counteracted by the adenosine antagonist. This is the first study to demonstrate that PUTRE can induce sleep through adenosine A1 receptor-specific agonist activity. Therefore, PUTRE is suggested as a valuable sleep-enhancing substance. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-024-01663-4.
Collapse
Affiliation(s)
- Joo-Hyun Oh
- Herbal Hormone Research Institute, Naturalendo Tech Co., Ltd., Seongnam, 13486 South Korea
| | - Yoon-Young Han
- Herbal Hormone Research Institute, Naturalendo Tech Co., Ltd., Seongnam, 13486 South Korea
| | - Eun-Bi Kim
- Herbal Hormone Research Institute, Naturalendo Tech Co., Ltd., Seongnam, 13486 South Korea
| | - Ha-Neul Jo
- Herbal Hormone Research Institute, Naturalendo Tech Co., Ltd., Seongnam, 13486 South Korea
| | - Jae-Sun Lee
- Herbal Hormone Research Institute, Naturalendo Tech Co., Ltd., Seongnam, 13486 South Korea
| | - Bo-Mi Kim
- Herbal Hormone Research Institute, Naturalendo Tech Co., Ltd., Seongnam, 13486 South Korea
| | - Ji-Min Kim
- Herbal Hormone Research Institute, Naturalendo Tech Co., Ltd., Seongnam, 13486 South Korea
| | - Young-Seob Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration, Eumseong, 27709 South Korea
| | - Dae Young Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu, 41566 South Korea
| | - Kwan-Woo Kim
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration, Eumseong, 27709 South Korea
| | - Inil Lee
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin, 17104 South Korea
| | - Yong-Wook Lee
- Herbal Hormone Research Institute, Naturalendo Tech Co., Ltd., Seongnam, 13486 South Korea
| | - Chan-Sung Park
- Herbal Hormone Research Institute, Naturalendo Tech Co., Ltd., Seongnam, 13486 South Korea
| | - Dae-Ok Kim
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin, 17104 South Korea
| |
Collapse
|
3
|
Zhao L, Witter MP, Palomero-Gallagher N. Cyto-, gene, and multireceptor architecture of the early postnatal mouse hippocampal complex. Prog Neurobiol 2025; 245:102704. [PMID: 39709019 DOI: 10.1016/j.pneurobio.2024.102704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/27/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Neurotransmitter receptors are key molecules in signal transmission in the adult brain, and their precise spatial and temporal balance expressions also play a critical role in normal brain development. However, the specific balance expression of multiple receptors during hippocampal development is not well characterized. In this study, we used quantitative in vivo receptor autoradiography to measure the distributions and densities of 18 neurotransmitter receptor types in the mouse hippocampal complex at postnatal day 7, and compared them with the expressions of their corresponding encoding genes. We provide a novel and comprehensive characterization of the cyto-, gene, and multireceptor architecture of the developing mouse hippocampal and subicular regions during the developmental period, which typically differs from that in the adult brain. High-density receptor expressions with distinct regional and laminar distributions were observed for AMPA, Kainate, mGluR2/3, GABAA, GABAA/BZ, α2, and A1 receptors during this specific period, whereas NMDA, GABAB, α1, M1, M2, M3, nicotinic α4β2, 5-HT1A, 5-HT2, D1 and D2/D3 receptors exhibited relatively low and homogeneous expressions. This specific balance of multiple receptors aligns with regional cytoarchitecture, neurotransmitter distributions, and gene expressions. Moreover, contrasting with previous findings, we detected a high α2 receptor density, with distinct regional and laminar distribution patterns. A non-covariation differentiation phenomenon between α2 receptor distributions and corresponding gene expressions is also demonstrated in this early developmental period. The multimodal data provides new insights into understanding the hippocampal development from the perspective of cell, gene, and multireceptor levels, and contributes important resources for further interdisciplinary analyses.
Collapse
Affiliation(s)
- Ling Zhao
- Department of Psychology, School of Public Policy and Management, Nanchang University, Nanchang 330000, China; Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich 52425, Germany.
| | - Menno P Witter
- Kavli Institute for Systems Neuroscience, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Nicola Palomero-Gallagher
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich 52425, Germany; C. & O. Vogt Institute for Brain Research, Heinrich-Heine-University, Dusseldorf 40225, Germany
| |
Collapse
|
4
|
Hung CJ, Tsai CT, Rahaman SM, Yamanaka A, Seo W, Yokoyama T, Sakamoto M, Ono D. Neuropeptidergic Input from the Lateral Hypothalamus to the Suprachiasmatic Nucleus Alters the Circadian Period in Mice. J Neurosci 2025; 45:e0351242024. [PMID: 39622648 PMCID: PMC11756623 DOI: 10.1523/jneurosci.0351-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 10/09/2024] [Accepted: 11/19/2024] [Indexed: 01/24/2025] Open
Abstract
In mammals, the central circadian clock is located in the suprachiasmatic nucleus (SCN) of the hypothalamus, which transmits circadian information to other brain regions and regulates the timing of sleep and wakefulness. Neurons in the lateral hypothalamus (LH), particularly those producing melanin-concentrating hormone (MCH) and orexin, are key regulators of sleep and wakefulness. Although the SCN receives nonphotic input from other brain regions, the mechanisms of functional input from the LH to the SCN remain poorly understood. Here, we show that orexin and MCH peptides influence the circadian period within the SCN of both sexes. When these neurons are ablated, the circadian behavioral rhythms are lengthened under constant darkness. Using anterograde and retrograde tracing, we found that orexin and MCH neurons project to the SCN. Furthermore, the application of these peptides to cultured SCN slices shortened circadian rhythms and reduced intracellular cAMP levels. Additionally, pharmacological reduction of intracellular cAMP levels similarly shortened the circadian period in SCN slices. These findings suggest that orexin and MCH peptides from the LH contribute to the modulation of the circadian period in the SCN.
Collapse
Affiliation(s)
- Chi Jung Hung
- Stress Recognition and Response, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| | - Chang-Ting Tsai
- Stress Recognition and Response, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| | - Sheikh Mizanur Rahaman
- Stress Recognition and Response, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| | - Akihiro Yamanaka
- Chinese Institute for Brain Research (CIBR), Beijing 102206, China
| | - Wooseok Seo
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Tatsushi Yokoyama
- Department of Brain Development and Regeneration, Graduate School of Biostudies, Kyoto University, Kyoto 606-8507, Japan
| | - Masayuki Sakamoto
- Department of Brain Development and Regeneration, Graduate School of Biostudies, Kyoto University, Kyoto 606-8507, Japan
| | - Daisuke Ono
- Stress Recognition and Response, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
5
|
Blume C, Münch M. Effects of light on biological functions and human sleep. HANDBOOK OF CLINICAL NEUROLOGY 2025; 206:3-16. [PMID: 39864930 DOI: 10.1016/b978-0-323-90918-1.00008-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The nonvisual effects of light in humans are mainly conveyed by a subset of retinal ganglion cells that contain the pigment melanopsin which renders them intrinsically photosensitive (= intrinsically photosensitive retinal ganglion cells, ipRGCs). They have direct connections to the main circadian clock in the suprachiasmatic nuclei (SCN) of the hypothalamus and modulate a variety of physiological processes, pineal melatonin secretion, autonomic functions, cognitive processes such as attention, and behavior, including sleep and wakefulness. This is because efferent projections from the SCN reach other hypothalamic nuclei, the pineal gland, thalamus, basal forebrain, and the brainstem. The ipRGCs also directly impact the prefrontal cortex and the perihabenular nucleus (mood). In particular, light suppresses the secretion of melatonin in a dose-dependent manner, mainly depending on irradiance and spectral composition of light. There is evidence that exposure to light-emitting devices from luminaires and screens before bedtime can impact on sleep onset latency, sleep duration, and sleep quality. Likewise, light exposure during daytime modulates sleep architecture, duration, and sleep quality during the subsequent night. Therefore, the integration of acute, circadian, and long-term effects of light together influence sleep-wake quality and behavior in healthy individuals, as well as in patients with psychiatric or medical disorders.
Collapse
Affiliation(s)
- Christine Blume
- Centre for Chronobiology, Psychiatric Hospital of the University of Basel, Basel, Switzerland; Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Mirjam Münch
- Centre for Chronobiology, Psychiatric Hospital of the University of Basel, Basel, Switzerland; Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
6
|
Wu L, Zhao M, Chen X, Wang H. A miR-219-5p-bmal1b negative feedback loop contributes to circadian regulation in zebrafish. Commun Biol 2024; 7:1671. [PMID: 39702498 DOI: 10.1038/s42003-024-07309-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024] Open
Abstract
MicroRNAs post-transcriptionally regulate gene expression and contribute to numerous life processes, including circadian rhythms. However, whether miRNAs contribute to zebrafish circadian regulation has not yet been investigated. Here, we showed that mature miR-219-5p, and its three pre-miRNAs, mir-219-1, mir-219-2, and mir-219-3, are rhythmically expressed primarily in Tectum opticum (TeO), Corpus cerebelli (CCe), and Crista cerellaris (CC) of the zebrafish brain. While mir-219-1 and mir-219-2 are regulated by the circadian clock through the E-like box, mir-219-3 is regulated by light via the D-box. Deleting mir-219-1, mir-219-2, or mir-219-3 individually or knocking down miR-219-5p all results in a shortened period of locomotor rhythms and up-regulation of bmal1b. RIP assays with Ago2 and miRNA pull-down assays show that miR-219-5p binds to bmal1b in the RISC. Cell transfection and in Vivo assays show that miR219-5p inhibits bmal1b through binding to its 3'UTR. Further, transcriptome analysis of miR-219-5p knockdown zebrafish adult brain reveals possible roles of miR-219-5p in phototransduction and neuroactive ligand-receptor interaction. Together, our findings demonstrate that mir-219-1, mir-219-2, and mir-219-3 are controlled directly by the circadian clock; and in turn, miR-219-5p contributes to circadian regulation by targeting bmal1b, highlighting a miR-219-5p-bmal1b negative feedback loop in the zebrafish circadian circuit.
Collapse
Affiliation(s)
- Lianxin Wu
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Meng Zhao
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Xifeng Chen
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China.
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
7
|
Marciante AB, Tadjalli A, Nikodemova M, Burrowes KA, Oberto J, Luca EK, Seven YB, Watters JJ, Baker TL, Mitchell GS. Microglia regulate motor neuron plasticity via reciprocal fractalkine and adenosine signaling. Nat Commun 2024; 15:10349. [PMID: 39609435 PMCID: PMC11605081 DOI: 10.1038/s41467-024-54619-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
We report an important role for microglia in regulating neuroplasticity within phrenic motor neurons. Brief episodes of low oxygen (acute intermittent hypoxia; AIH) elicit a form of respiratory motor plasticity known as phrenic long-term facilitation (pLTF) that is regulated by the balance of competing serotonin vs adenosine-initiated cellular mechanisms. Serotonin arises from brainstem raphe neurons, but the source of adenosine is unknown. We tested if hypoxic episodes initiate phrenic motor neuron to microglia fractalkine signaling that evokes extracellular adenosine formation using a well-defined neurophysiology preparation in male rats. With moderate AIH, phrenic motor neuron adenosine 2A receptor activation undermines serotonin-dominant pLTF whereas severe AIH induces pLTF by the adenosine-dependent mechanism. Consequently, phrenic motor neuron fractalkine knockdown, microglial fractalkine receptor inhibition, and microglial ablation enhance moderate AIH, but suppress severe AIH-induced pLTF. We conclude, microglia play important roles in healthy spinal cords, regulating plasticity in motor neurons responsible for breathing.
Collapse
Affiliation(s)
- Alexandria B Marciante
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Arash Tadjalli
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Nova Southeastern University, College of Allopathic Medicine (NSU MD) Department of Medical Education, 3200 South University Drive, Fort Lauderdale, FL, USA
| | - Maria Nikodemova
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Kayla A Burrowes
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jose Oberto
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Edward K Luca
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Yasin B Seven
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jyoti J Watters
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, USA
| | - Tracy L Baker
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, USA
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
8
|
Wei Y, Miao Z, Ye H, Wu M, Wei X, Zhang Y, Cai L. The Effect of Caffeine Exposure on Sleep Patterns in Zebrafish Larvae and Its Underlying Mechanism. Clocks Sleep 2024; 6:749-763. [PMID: 39584977 PMCID: PMC11586999 DOI: 10.3390/clockssleep6040048] [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: 08/30/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024] Open
Abstract
The effect of caffeine on the behavior and sleep patterns of zebrafish larvae, as well as its underlying mechanisms, has been a topic of great interest. This study aimed to investigate the impact of caffeine on zebrafish larval sleep/wake behavior and the expression of key regulatory genes such as cAMP-response element binding protein (CREB) and adenosine (ADA) in the sleep pathway. To begin, the study determined the optimal dose and duration of caffeine exposure, with the optimal doses found to be 31.25 μM, 62.5 μM, and 120 μM. Similarly, the optimal exposure time was established as no more than 120 h, ensuring a mortality rate of less than 10%. The confirmation of these conditions was achieved through the assessment of angiogenesis and the inflammatory reaction. As a result, the treatment time point of 24 h post-fertilization (hpf) was selected to examine the effects of caffeine on zebrafish larval sleep rhythm (48 h, with a light cycle of 14:10). Furthermore, the study analyzed the expression of clock genes (bmal1a, per1b, per2, per3, cry2), adenosine receptor genes (adora1a, adora1b, adora2aa, adora2ab, adora2b), and key regulatory factors (CREB and ADA). The research confirmed that caffeine could induce sleep pattern disorders, significantly upregulate adenosine receptor genes (adora1a, adora1b, adora2a, adora2ab, adora2b) (p < 0.05), and markedly decrease the total sleep time and sleep efficiency of the larvae. Additionally, the activity of ADA significantly increased during the exposure (p < 0.001), and the tissue-specific expression of CREB was also significantly increased, as assessed by immunofluorescence. Caffeine may regulate circadian clock genes through the ADA/ADORA/CREB pathway. These findings not only enhance our understanding of the effects of caffeine on zebrafish larvae but also provide valuable insights into the potential impact of caffeine on human behavior and sleep.
Collapse
Affiliation(s)
- Yuanzheng Wei
- Guangdong Provincial Biotechnology Research Institute (Guangdong Provincial Laboratory Animals Monitoring Center), Guangzhou 510663, China; (Y.W.); (Z.M.); (H.Y.); (M.W.)
| | - Zongyu Miao
- Guangdong Provincial Biotechnology Research Institute (Guangdong Provincial Laboratory Animals Monitoring Center), Guangzhou 510663, China; (Y.W.); (Z.M.); (H.Y.); (M.W.)
| | - Huixin Ye
- Guangdong Provincial Biotechnology Research Institute (Guangdong Provincial Laboratory Animals Monitoring Center), Guangzhou 510663, China; (Y.W.); (Z.M.); (H.Y.); (M.W.)
| | - Meihui Wu
- Guangdong Provincial Biotechnology Research Institute (Guangdong Provincial Laboratory Animals Monitoring Center), Guangzhou 510663, China; (Y.W.); (Z.M.); (H.Y.); (M.W.)
| | - Xinru Wei
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China;
| | - Yu Zhang
- Guangdong Provincial Biotechnology Research Institute (Guangdong Provincial Laboratory Animals Monitoring Center), Guangzhou 510663, China; (Y.W.); (Z.M.); (H.Y.); (M.W.)
| | - Lei Cai
- Guangdong Provincial Biotechnology Research Institute (Guangdong Provincial Laboratory Animals Monitoring Center), Guangzhou 510663, China; (Y.W.); (Z.M.); (H.Y.); (M.W.)
| |
Collapse
|
9
|
Narain P, Petković A, Šušić M, Haniffa S, Anwar M, Arnoux M, Drou N, Antonio-Saldi G, Chaudhury D. Nighttime-specific differential gene expression in suprachiasmatic nucleus and habenula is associated with resilience to chronic social stress. Transl Psychiatry 2024; 14:407. [PMID: 39358331 PMCID: PMC11447250 DOI: 10.1038/s41398-024-03100-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024] Open
Abstract
The molecular mechanisms that link stress and biological rhythms still remain unclear. The habenula (Hb) is a key brain region involved in regulating diverse types of emotion-related behaviours while the suprachiasmatic nucleus (SCN) is the body's central clock. To investigate the effects of chronic social stress on transcription patterns, we performed gene expression analysis in the Hb and SCN of stress-naïve and stress-exposed mice. Our analysis revealed a large number of differentially expressed genes and enrichment of synaptic and cell signalling pathways between resilient and stress-naïve mice at zeitgeber 16 (ZT16) in both the Hb and SCN. This transcriptomic signature was nighttime-specific and observed only in stress-resilient mice. In contrast, there were relatively few differences between the stress-susceptible and stress-naïve groups across time points. Our results reinforce the functional link between circadian gene expression patterns and differential responses to stress, thereby highlighting the importance of temporal expression patterns in homoeostatic stress responses.
Collapse
Affiliation(s)
- Priyam Narain
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Aleksa Petković
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Marko Šušić
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Salma Haniffa
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Mariam Anwar
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Marc Arnoux
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Nizar Drou
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | | | - Dipesh Chaudhury
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE.
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE.
- Center for Brain and Health, New York University Abu Dhabi, Abu Dhabi, UAE.
| |
Collapse
|
10
|
Mukherjee U, Sehar U, Brownell M, Reddy PH. Mechanisms, consequences and role of interventions for sleep deprivation: Focus on mild cognitive impairment and Alzheimer's disease in elderly. Ageing Res Rev 2024; 100:102457. [PMID: 39154978 DOI: 10.1016/j.arr.2024.102457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/20/2024]
Abstract
Sleep is established as an essential physiological need that impacts physical, emotional, and cognitive functions profoundly. Physiologically, inadequate sleep weakens immune function, heightening susceptibility to infections and chronic illnesses such as obesity, diabetes, and cardiovascular diseases. Hormonal disruptions due to sleep loss further exacerbate metabolic dysregulation, contributing to weight gain and other health complications. Emotionally, sleep deprivation leads to mood disturbances, including increased irritability, heightened stress responses, and a greater likelihood of mood disorders like depression and anxiety. These effects are compounded by cognitive impairments such as reduced alertness, impaired memory consolidation, and compromised decision-making abilities, akin to the impairments caused by alcohol consumption. Motor skills and coordination also suffer, elevating the risk of accidents, particularly in high-stress environments. For older adults, sleep quality is closely linked to cognitive function and overall longevity. Optimal sleep patterns are associated with slower brain aging and improved health outcomes. However, sleep disorders exacerbate existing conditions such as epilepsy and asthma, necessitating interventions like cognitive behavioral therapy (CBT) and medications such as melatonin to mitigate their impact. Education emerges as a crucial tool in promoting healthier sleep habits across all age groups. Addressing misconceptions about sleep and integrating sleep health into public health policies are essential steps toward improving overall well-being. Additionally, lifestyle factors such as diet and physical activity play significant roles in regulating sleep patterns, further emphasizing the interconnectedness of sleep with broader health outcomes. In summary, the articles underscore the intricate mechanisms through which sleep influences physiological functions and advocate for comprehensive approaches to enhance sleep hygiene and mitigate the adverse effects of sleep deprivation on human health.
Collapse
Affiliation(s)
- Upasana Mukherjee
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Malcolm Brownell
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
11
|
Thomas DC, Somaiya T, Meira E Cruz M, Kodaganallur Pitchumani P, Ardeshna A, Ravi A, Prabhakar S. The enigma of sleep: Implications of sleep neuroscience for the dental clinician and patient. J Am Dent Assoc 2024; 155:735-746. [PMID: 39007793 DOI: 10.1016/j.adaj.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/14/2024] [Accepted: 05/28/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Sleep disturbances have been shown to result in considerable morbidity and mortality. It is important for dental clinicians to understand the neuroscience behind sleep disorders. TYPES OF STUDIES REVIEWED The authors conducted a search of the literature published from January 1990 through March 2024 of sleep medicine-related articles, with a focus on neuroscience. The authors prioritized articles about the science of sleep as related to dental medicine. RESULTS The authors found a proliferation of articles related to sleep neuroscience along with its implications in dental medicine. The authors also found that the intricate neuroscientific principles of sleep medicine are being investigated robustly. The salient features of, and the differences between, central and obstructive sleep apneas have been elucidated. Sleep genes, such as CRY, PER1, PER2, and CLOCK, and their relationship to cancer and neurodegeneration are also additions to this rapidly developing science. CONCLUSIONS AND PRACTICAL IMPLICATIONS The dental clinician has the potential to be the first to screen patients for possible sleep disorders and make prompt referrals to the appropriate medical professionals. This can be lifesaving as well as minimize potential future morbidity for the patient.
Collapse
|
12
|
Tahara Y, Ding J, Ito A, Shibata S. Sweetened caffeine drinking revealed behavioral rhythm independent of the central circadian clock in male mice. NPJ Sci Food 2024; 8:51. [PMID: 39160163 PMCID: PMC11333706 DOI: 10.1038/s41538-024-00295-6] [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: 01/26/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Caffeine consumption is associated with the evening chronotype, and caffeine administration in mice results in prolonged period of the circadian rhythm in locomotor activity. However, as caffeine is bitter, sweetened caffeine is preferred by humans and mice; yet, its impact on the circadian clock has not been explored. In this study, mice were provided with freely available sweetened caffeine to investigate its effects on behavioral rhythms and peripheral clocks. Mice that freely consumed sweetened caffeine shifted from nocturnal to diurnal activity rhythms. In addition to the light-dark entrained behavioral rhythm component, some animals exhibited free-running period longer than 24-h. Intraperitoneal administration of caffeine at the beginning of the light phase also acutely induced diurnal behavior. The behavioral rhythms with long period (26-30 h) due to sweetened caffeine were observed even in mice housed under constant light or with a lesioned central circadian clock located in the suprachiasmatic nucleus of the hypothalamus; however, the rhythmicity was unstable. PER2::LUCIFERASE rhythms in peripheral tissues, such as the kidney, as measured via in vivo whole-body imaging during caffeine consumption, showed reduced amplitude and desynchronized phases among individuals. These results indicate that consumption of sweetened caffeine induces diurnal and long-period behavioral rhythms irrespective of the central clock, causing desynchronization of the clock in the body.
Collapse
Affiliation(s)
- Yu Tahara
- Department of Public Health and Health Policy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-0037, Japan.
- School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 162-0056, Japan.
| | - Jingwei Ding
- Department of Public Health and Health Policy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-0037, Japan
| | - Akito Ito
- School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 162-0056, Japan
| | - Shigenobu Shibata
- Department of Public Health and Health Policy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-0037, Japan
- School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 162-0056, Japan
| |
Collapse
|
13
|
Katsioudi G, Biancolin AD, Jiménez-Sanchez C, Dibner C. Human primary cells can tell body time: Dedicated to Steven A. Brown. Eur J Neurosci 2024; 60:3946-3960. [PMID: 38951126 DOI: 10.1111/ejn.16453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/21/2024] [Accepted: 06/10/2024] [Indexed: 07/03/2024]
Abstract
The field of chronobiology has advanced significantly since ancient observations of natural rhythms. The intricate molecular architecture of circadian clocks, their hierarchical organization within the mammalian body, and their pivotal roles in organ physiology highlight the complexity and significance of these internal timekeeping mechanisms. In humans, circadian phenotypes exhibit considerable variability among individuals and throughout the individual's lifespan. A fundamental challenge in mechanistic studies of human chronobiology arises from the difficulty of conducting serial sampling from most organs. The concept of studying circadian clocks in vitro relies on the groundbreaking discovery by Ueli Schibler and colleagues that nearly every cell in the body harbours autonomous molecular oscillators. The advent of circadian bioluminescent reporters has provided a new perspective for this approach, enabling high-resolution continuous measurements of cell-autonomous clocks in cultured cells, following in vitro synchronization pulse. The work by Steven A. Brown has provided compelling evidence that clock characteristics assessed in primary mouse and human skin fibroblasts cultured in vitro represent a reliable estimation of internal clock properties in vivo. The in vitro approach for studying molecular human clocks in cultured explants and primary cells, pioneered by Steve Brown, represents an invaluable tool for assessing inter-individual differences in circadian characteristics alongside comprehensive genetic, biochemical and functional analyses. In a broader context, this reliable and minimally invasive approach offers a unique perspective for unravelling the functional inputs and outputs of oscillators operative in nearly any human tissue in physiological contexts and across various pathologies.
Collapse
Affiliation(s)
- Georgia Katsioudi
- Department of Surgery, Division of Thoracic and Endocrine Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
| | - Andrew D Biancolin
- Department of Surgery, Division of Thoracic and Endocrine Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
| | - Cecilia Jiménez-Sanchez
- Department of Surgery, Division of Thoracic and Endocrine Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
| | - Charna Dibner
- Department of Surgery, Division of Thoracic and Endocrine Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
| |
Collapse
|
14
|
Foster RG. Introduction and reflections on the comparative physiology of sleep and circadian rhythms. J Comp Physiol B 2024; 194:225-231. [PMID: 38856727 PMCID: PMC11233284 DOI: 10.1007/s00360-024-01567-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 05/17/2024] [Indexed: 06/11/2024]
Abstract
Circadian rhythms and the sleep/wake cycle allows us, and most life on Earth, to function optimally in a dynamic world, adjusting all aspects of biology to the varied and complex demands imposed by the 24-hour rotation of the Earth upon its axis. A key element in understanding these rhythms, and the success of the field in general, has been because researchers have adopted a comparative approach. Across all taxa, fundamental questions relating to the generation and regulation of sleep and circadian rhythms have been address using biochemical, molecular, cellular, system and computer modelling techniques. Furthermore, findings have been placed into an ecological and evolutionary context. By addressing both the "How" - mechanistic, and "Why" - evolutionary questions in parallel, the field has achieved remarkable successes, including how circadian rhythms are generated and regulated by light. Yet many key questions remain. In this special issue on the Comparative Physiology of Sleep and Circadian Rhythms, celebrating the 100th anniversary of the Journal of Comparative Physiology, important new discoveries are detailed. These findings illustrate the power of comparative physiology to address novel questions and demonstrate that sleep and circadian physiology are embedded within the biological framework of an organism.
Collapse
Affiliation(s)
- Russell G Foster
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford, OX1 3QU, UK.
| |
Collapse
|
15
|
Marciante AB, Tadjalli A, Burrowes KA, Oberto JR, Luca EK, Seven YB, Nikodemova M, Watters JJ, Baker TL, Mitchell GS. Microglia regulate motor neuron plasticity via reciprocal fractalkine/adenosine signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.592939. [PMID: 38765982 PMCID: PMC11100694 DOI: 10.1101/2024.05.07.592939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Microglia are innate CNS immune cells that play key roles in supporting key CNS functions including brain plasticity. We now report a previously unknown role for microglia in regulating neuroplasticity within spinal phrenic motor neurons, the neurons driving diaphragm contractions and breathing. We demonstrate that microglia regulate phrenic long-term facilitation (pLTF), a form of respiratory memory lasting hours after repetitive exposures to brief periods of low oxygen (acute intermittent hypoxia; AIH) via neuronal/microglial fractalkine signaling. AIH-induced pLTF is regulated by the balance between competing intracellular signaling cascades initiated by serotonin vs adenosine, respectively. Although brainstem raphe neurons release the relevant serotonin, the cellular source of adenosine is unknown. We tested a model in which hypoxia initiates fractalkine signaling between phrenic motor neurons and nearby microglia that triggers extracellular adenosine accumulation. With moderate AIH, phrenic motor neuron adenosine 2A receptor activation undermines serotonin-dominant pLTF; in contrast, severe AIH drives pLTF by a unique, adenosine-dominant mechanism. Phrenic motor neuron fractalkine knockdown, cervical spinal fractalkine receptor inhibition on nearby microglia, and microglial depletion enhance serotonin-dominant pLTF with moderate AIH but suppress adenosine-dominant pLTF with severe AIH. Thus, microglia play novel functions in the healthy spinal cord, regulating hypoxia-induced neuroplasticity within the motor neurons responsible for breathing.
Collapse
Affiliation(s)
- Alexandria B. Marciante
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida; Gainesville, FL, USA 32610
| | - Arash Tadjalli
- Current Address: Nova Southeastern University, College of Allopathic Medicine (NSU MD), Department of Medical Education, 3200 South University Drive, Fort Lauderdale, FL 33328-2018
| | - Kayla A. Burrowes
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida; Gainesville, FL, USA 32610
| | - Jose R. Oberto
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida; Gainesville, FL, USA 32610
| | - Edward K. Luca
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida; Gainesville, FL, USA 32610
| | - Yasin B. Seven
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida; Gainesville, FL, USA 32610
| | - Maria Nikodemova
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida; Gainesville, FL, USA 32610
| | - Jyoti J. Watters
- Current Address: Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706
| | - Tracy L. Baker
- Current Address: Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706
| | - Gordon S. Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida; Gainesville, FL, USA 32610
| |
Collapse
|
16
|
McPartland M, Stevens S, Bartosova Z, Vardeberg IG, Völker J, Wagner M. Beyond the Nucleus: Plastic Chemicals Activate G Protein-Coupled Receptors. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:4872-4883. [PMID: 38440973 PMCID: PMC10956435 DOI: 10.1021/acs.est.3c08392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 03/06/2024]
Abstract
G protein-coupled receptors (GPCRs) are central mediators of cell signaling and physiological function. Despite their biological significance, GPCRs have not been widely studied in the field of toxicology. Herein, we investigated these receptors as novel targets of plastic chemicals using a high-throughput drug screening assay with 126 human non-olfactory GPCRs. In a first-pass screen, we tested the activity of triphenol phosphate, bisphenol A, and diethyl phthalate, as well as three real-world mixtures of chemicals extracted from plastic food packaging covering all major polymer types. We found 11 GPCR-chemical interactions, of which the chemical mixtures exhibited the most robust activity at adenosine receptor 1 (ADORA1) and melatonin receptor 1 (MTNR1A). We further confirm that polyvinyl chloride and polyurethane products contain ADORA1 or MTNRA1 agonists using a confirmatory secondary screen and pharmacological knockdown experiments. Finally, an analysis of the associated gene ontology terms suggests that ADORA1 and MTNR1A activation may be linked to downstream effects on circadian and metabolic processes. This work highlights that signaling disruption caused by plastic chemicals is broader than that previously believed and demonstrates the relevance of nongenomic pathways, which have, thus far, remained unexplored.
Collapse
Affiliation(s)
- Molly McPartland
- Department of Biology, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Sarah Stevens
- Department of Biology, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Zdenka Bartosova
- Department of Biology, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Ingrid Gisnås Vardeberg
- Department of Biology, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | | | - Martin Wagner
- Department of Biology, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| |
Collapse
|
17
|
Huang L, Zhu W, Li N, Zhang B, Dai W, Li S, Xu H. Functions and mechanisms of adenosine and its receptors in sleep regulation. Sleep Med 2024; 115:210-217. [PMID: 38373361 DOI: 10.1016/j.sleep.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/01/2024] [Accepted: 02/03/2024] [Indexed: 02/21/2024]
Abstract
Sleep is a natural and recurring state of life. Long-term insomnia can lead to physical and mental fatigue, inattention, memory loss, anxiety, depression and other symptoms, imposing immense public health and economic burden worldwide. The sleep and awakening regulation system is composed of many nerve nuclei and neurotransmitters in the brain, and it forms a neural network that interacts and restricts each other to regulate the occurrence and maintenance of sleep-wake. Adenosine (AD) is a neurotransmitter in the central nervous system and a driver of sleep. Meanwhile, the functions and mechanisms underlying sleep-promoting effects of adenosine and its receptors are still not entirely clear. However, in recent years, the increasing evidence indicated that adenosine can promote sleep through inhibiting arousal system and activating sleep-promoting system. At the same time, astrocyte-derived adenosine in modulating sleep homeostasis and sleep loss-induced related cognitive and memory deficits plays an important role. This review, therefore, summarizes the current research on the functions and possible mechanisms of adenosine and its receptors in the regulation of sleep and homeostatic control of sleep. Understanding these aspects will provide us better ideas on clinical problems such as insomnia, hypersomnia and other sleep disorders.
Collapse
Affiliation(s)
- Lishan Huang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Wenwen Zhu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Nanxi Li
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Bin Zhang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Wenbin Dai
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Sen Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, China.
| | - Houping Xu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
18
|
Lei T, Hua H, Du H, Xia J, Xu D, Liu W, Wang Y, Yang T. Molecular mechanisms of artificial light at night affecting circadian rhythm disturbance. Arch Toxicol 2024; 98:395-408. [PMID: 38103071 DOI: 10.1007/s00204-023-03647-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/16/2023] [Indexed: 12/17/2023]
Abstract
Artificial light at night (ALAN) pollution has been regarded as a global environmental concern. More than 80% of the global population is exposed to light pollution. Exacerbating this issue, artificially lit outdoor areas are growing by 2.2% per year, while continuously lit areas have brightened by 2.2% each year due to rapid population growth and expanding urbanization. Furthermore, the increasing prevalence of night shift work and smart device usage contributes to the inescapable influence of ALAN. Studies have shown that ALAN can disrupt endogenous biological clocks, resulting in a disturbance of the circadian rhythm, which ultimately affects various physiological functions. Up until now, scholars have studied various disease mechanisms caused by ALAN that may be related to the response of the circadian system to light. This review outlines the molecular mechanisms by which ALAN causes circadian rhythm abnormalities in sleep disorders, endocrine diseases, cardiovascular disease, cancer, immune impairment, depression, anxiety and cognitive impairments.
Collapse
Affiliation(s)
- Ting Lei
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, 110122, Liaoning, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Hui Hua
- Department of Nutrition, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Huiying Du
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, 110122, Liaoning, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Jie Xia
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, 110122, Liaoning, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Dandan Xu
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, 110122, Liaoning, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Wei Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, 110122, Liaoning, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yutong Wang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China.
| | - Tianyao Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang, 110122, Liaoning, China.
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
19
|
Aeschbach D, Cohen DA, Lockyer BJ, Chellappa SL, Klerman EB. Spontaneous attentional failures reflect multiplicative interactions of chronic sleep loss with acute sleep loss and circadian misalignment. Sleep Health 2024; 10:S89-S95. [PMID: 37689503 DOI: 10.1016/j.sleh.2023.07.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/19/2023] [Accepted: 07/22/2023] [Indexed: 09/11/2023]
Abstract
OBJECTIVES Acute and chronic sleep loss and circadian timing interact such that, depending on their combination, small or very large performance decrements are observed in tasks of attention. Here, we tested whether such nonlinear interactions extend to a physiological measure of spontaneous visual attentional failures, indicating a fundamental principle of sleep-wake regulation. METHODS Nine healthy volunteers completed an in-laboratory 3-week forced desynchrony protocol consisting of 12 consecutive 42.85-hour cycles with a sleep-wake ratio of 1:3.3. The protocol induced increasing chronic sleep loss, while extended wake (32.85 hours) and sleep episodes (10 hours) occurred at multiple circadian phases. Attentional failure rate was quantified from continuous electrooculograms (number of 30-second epochs with slow eye movements/h of wakefulness) as a function of time since scheduled wake (acute sleep loss), week of study (chronic sleep loss), and circadian (melatonin) phase. RESULTS During the first ∼8 hours awake, attentional failure rate was low, irrespective of the week. During the following wake hours, attentional failure rate increased steadily but at a faster rate in weeks 2 and 3 compared to week 1. The effects of acute and chronic sleep loss on attentional failure rate were magnified during the biological night compared to the biological day. CONCLUSIONS A single extended sleep episode can only temporarily reverse attentional impairment associated with chronic sleep loss. Multiplicative effects of acute and chronic sleep loss-further amplified during the biological night-substantiate the interaction of 2 homeostatic response mechanisms and caution against underestimating their disproportionate combined impact on performance, health, and safety.
Collapse
Affiliation(s)
- Daniel Aeschbach
- Department of Sleep and Human Factors Research, Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany; Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Center, Bonn, Germany; Division of Sleep and Circadian Disorders, Department of Medicine, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA; Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA.
| | - Daniel A Cohen
- Department of Neurology, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Brandon J Lockyer
- Division of Sleep and Circadian Disorders, Department of Medicine, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Sarah L Chellappa
- School of Psychology, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Elizabeth B Klerman
- Division of Sleep and Circadian Disorders, Department of Medicine, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA; Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA; Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Figarella K, Kim J, Ruan W, Mills T, Eltzschig HK, Yuan X. Hypoxia-adenosine axis as therapeutic targets for acute respiratory distress syndrome. Front Immunol 2024; 15:1328565. [PMID: 38312838 PMCID: PMC10835146 DOI: 10.3389/fimmu.2024.1328565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/03/2024] [Indexed: 02/06/2024] Open
Abstract
The human respiratory and circulatory systems collaborate intricately to ensure oxygen delivery to all cells, which is vital for ATP production and maintaining physiological functions and structures. During limited oxygen availability, hypoxia-inducible factors (HIFs) are stabilized and play a fundamental role in maintaining cellular processes for hypoxia adaptation. First discovered during investigations of erythropoietin production regulation, HIFs influence physiological and pathological processes, including development, inflammation, wound healing, and cancer. HIFs promote extracellular adenosine signaling by enhancing adenosine generation and receptor signaling, representing an endogenous feedback mechanism that curbs excessive inflammation, supports injury resolution, and enhances hypoxia tolerance. This is especially important for conditions that involve tissue hypoxia, such as acute respiratory distress syndrome (ARDS), which globally poses significant health challenges without specific treatment options. Consequently, pharmacological strategies to amplify HIF-mediated adenosine production and receptor signaling are of great importance.
Collapse
Affiliation(s)
- Katherine Figarella
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jieun Kim
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Wei Ruan
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tingting Mills
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Holger Klaus Eltzschig
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Xiaoyi Yuan
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
21
|
Wang S, Liu L, Liang S, Yang J, Zhang Y, Liu X. Effects of BXSMD on ESR1 and ESR2 expression in CSD female mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116973. [PMID: 37517566 DOI: 10.1016/j.jep.2023.116973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
ETHNIC PHARMACOLOGICAL RELEVANCE Due to the rapid pace of modern society, chronic insomnia has become universal phenomenon. In China, Banxia Shumi Decoction (BXSMD) has been used in treating chronic insomnia for thousands of years, but its chemical composition and action mechanism are still unknown. AIM OF THE STUDY This study aims to explore the chemical composition of BXSMD and its effects on Estrogen receptor 1 (ESR1) and Estrogen receptor 2 (ESR2) in mice with chronic sleep deprivation (CSD). MATERIALS AND METHODS UHPLC-Q-Orbitrap-MS/MS was applied in determining the chemical composition of BXSMD. After 21-day sleep deprivation (SD) in platform water environment, CSD mice model was prepared. By conducting open field test, 24-h autonomic diurnal and nocturnal activity of mice in each group was detected. ELISA was employed to measure the contents of 5-HT, DA, NE, GABA, Glu, and MT. With RT-PCR, Western blot (WB), and immunohistochemistry (IHC), mRNA and protein expressions of ESR1 and ESR2 in the hypothalamus and hippocampus were tested. RESULTS BXSMD included ferulic acid, kaverol, daidzein, apigenin, berberine, adenosine, aesculin, vanillin, naringin, and glycine, which might constitute the material basis forthe improvement of chronic insomnia. With BXSMD, the total moving distance and the rest time in dark period of CSD mice were shortened, while its rest time in light period was increased. Besides, BXSMD enhanced the contents of 5-HT, GABA, and MT in CSD mice, and decreased the contents of Glu, NE, and DA. BXSMD elevated the mRNA of Esr1 and Esr2, and elevated the protein expressions of ESR1 and ESR2 in the hypothalamus and hippocampus of CSD mice. CONCLUSIONS BXSMD contains various chemical components for sleep-wake regulation, with the mechanism of stimulating estrogen signaling pathway by regulating the expressions of ESR1 and ESR2, ultimately realizing the regulation to sleep-wake disorder caused by CSD.
Collapse
Affiliation(s)
- Shujun Wang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong Province, PR China
| | - Leilei Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong Province, PR China
| | - Shuzhi Liang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong Province, PR China
| | - Jinni Yang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong Province, PR China
| | - Yan Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong Province, PR China.
| | - Xijian Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong Province, PR China.
| |
Collapse
|
22
|
Le L, Miyanishi K, Tanaka J, Majewska AK. Microglial Regulation of Sleep and Wakefulness. ADVANCES IN NEUROBIOLOGY 2024; 37:243-260. [PMID: 39207696 DOI: 10.1007/978-3-031-55529-9_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Sleep serves a multitude of roles in brain maturation and function. Although the neural networks involved in sleep regulation have been extensively characterized, it is increasingly recognized that neurons are not the sole conductor orchestrating the rhythmic cycle of sleep and wakefulness. In the central nervous system, microglia have emerged as an important player in sleep regulation. Within the last two decades, microglia have gained substantial attention for carrying out numerous nonimmune tasks that are crucial for brain development and function by co-opting similar mechanisms used in their conventional immune functions. Here, we highlight the importance of microglia in sleep regulation with recent findings reporting an arrhythmic sleep/wake cycle in the absence of microglia. Although the underlying mechanisms for such regulation are still being uncovered, it is likely that microglial contributions to the rhythmic control of the sleep/wake cycle come from their influence on synaptic strength and neuronal activity. We review the current literature to provide speculative signaling pathways and suggest key questions for future research. Advancing our knowledge of the mechanistic contribution of microglia to sleep regulation will not only further our insight into this critical biological process but also be instrumental in providing novel therapeutic strategies for sleep disorders.
Collapse
Affiliation(s)
- Linh Le
- Department of Neuroscience, Del Monte Institute of Neuroscience, Center for Visual Science, Neuroscience Graduate Program, University of Rochester, Rochester, NY, USA
| | - Kazuya Miyanishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon, Japan.
| | - Ania K Majewska
- Department of Neuroscience, Del Monte Institute of Neuroscience, Center for Visual Science, Neuroscience Graduate Program, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
23
|
Witt RM, Byars KC, Decker K, Dye TJ, Riley JM, Simmons D, Smith DF. Current Considerations in the Diagnosis and Treatment of Circadian Rhythm Sleep-Wake Disorders in Children. Semin Pediatr Neurol 2023; 48:101091. [PMID: 38065634 PMCID: PMC10710539 DOI: 10.1016/j.spen.2023.101091] [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: 09/15/2023] [Accepted: 09/29/2023] [Indexed: 12/18/2023]
Abstract
Circadian Rhythm Sleep-Wake Disorders (CRSWDs) are important sleep disorders whose unifying feature is a mismatch between the preferred or required times for sleep and wakefulness and the endogenous circadian drives for these. Their etiology, presentation, and treatment can be different in pediatric patients as compared to adults. Evaluation of these disorders must be performed while viewed through the lens of a patient's comorbid conditions. Newer methods of assessment promise to provide greater diagnostic clarity and critical insights into how circadian physiology affects overall health and disease states. Effective clinical management of CRSWDs is multimodal, requiring an integrated approach across disciplines. Therapeutic success depends upon appropriately timed nonpharmacologic and pharmacologic interventions. A better understanding of the genetic predispositions for and causes of CRSWDs has led to novel clinical opportunities for diagnosis and improved therapeutics.
Collapse
Affiliation(s)
- Rochelle M Witt
- Division of Child Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Pulmonary Medicine and the Sleep Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Center for Circadian Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Kelly C Byars
- Division of Pulmonary Medicine and the Sleep Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Center for Circadian Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH; Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Kristina Decker
- Division of Pulmonary Medicine and the Sleep Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Center for Circadian Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH; Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Thomas J Dye
- Division of Child Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Pulmonary Medicine and the Sleep Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Center for Circadian Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Jessica M Riley
- Center for Circadian Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Danielle Simmons
- Division of Pulmonary Medicine and the Sleep Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Center for Circadian Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH; Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - David F Smith
- Division of Pulmonary Medicine and the Sleep Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Center for Circadian Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Otolaryngology- Head and Neck Surgery, University of Cincinnati College of Medicine, Cincinnati, OH.
| |
Collapse
|
24
|
Zhang Y, Li X, Liu Z, Zhao X, Chen L, Hao G, Ye X, Meng S, Xiao G, Mu J, Mu X, Qiu J, Qian Y. The neurobehavioral impacts of typical antibiotics toward zebrafish larvae. CHEMOSPHERE 2023; 340:139829. [PMID: 37598953 DOI: 10.1016/j.chemosphere.2023.139829] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/26/2023] [Accepted: 08/13/2023] [Indexed: 08/22/2023]
Abstract
Due to the widely usage in livestock, aquaculture and clinics, antibiotic residues are existed in aqueous environments and their potential toxicity to aquatic organisms is concerning. Here, we used zebrafish as the model to investigate the neurotoxicity and involved mechanism of seven antibiotics that were frequently detected in surface waters. The results revealed that the short-term exposure to clarithromycin (CLA), chlortetracycline (CTC) and roxithromycin (ROX) induced behavioral effects, with effective concentration of 1 μg/L (CTC and ROX) and 100 μg/L (CLA, CTC and ROX) respectively. A significant decrease in the travel distance and velocity as well as an increase in turn angle was measured. TUNEL assay identified increased cell apoptosis in brain sections of larvae exposed to three neurotoxic antibiotics, which raised the possibility that the behavioral symptoms were associated with neural damage. Transcriptome sequencing showed that the three antibiotics could affect the nervous system of zebrafish including the alteration of synaptogenesis and neurotransmission. Additionally, ROX and CTC affected pathways involved in mitochondrial stress response and endocrine system in zebrafish larvae. Besides, BDNF, ASCL1, and CREBBP are potential upstream regulatory factors that mediated these impacts. These findings indicated that exposure of CTC, ROX and CLA may cause abnormal behavior toward zebrafish larvae under environmental relevant concentration and revealed the potential role of neural cell apoptosis and synaptogenesis signaling in mediating this effect.
Collapse
Affiliation(s)
- Yining Zhang
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xue Li
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China; Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, China
| | - Zaiteng Liu
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaoyu Zhao
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lu Chen
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Guijie Hao
- Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture and Rural Affairs; Key Laboratory of Fish Health and Nutrition of Zhejiang Province; Zhejiang Institute of Freshwater Fisheries, Huzhou, China
| | - Xueping Ye
- Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture and Rural Affairs; Key Laboratory of Fish Health and Nutrition of Zhejiang Province; Zhejiang Institute of Freshwater Fisheries, Huzhou, China
| | - Shunlong Meng
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Jiangsu Province, China.
| | - Guohua Xiao
- Hebei Ocean and Fisheries Science Research Institute, Qinhuangdao, China; Hebei Marine Living Resources and Environment Key Laboratory, Qinhuangdao, China
| | - Jiandong Mu
- Hebei Ocean and Fisheries Science Research Institute, Qinhuangdao, China; Hebei Marine Living Resources and Environment Key Laboratory, Qinhuangdao, China
| | - Xiyan Mu
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China.
| | - Jing Qiu
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yongzhong Qian
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
25
|
Simões AP, Portes MAM, Lopes CR, Vanz F, Lourenço VS, Pliássova A, Gaspar IL, Silva HB, Tomé ÂR, Canas PM, Prediger RD, Cunha RA. Adenosine A 2A receptors control generalization of contextual fear in rats. Transl Psychiatry 2023; 13:316. [PMID: 37828000 PMCID: PMC10570294 DOI: 10.1038/s41398-023-02613-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/14/2023] Open
Abstract
Fear learning is essential to survival, but traumatic events may lead to abnormal fear consolidation and overgeneralization, triggering fear responses in safe environments, as occurs in post-traumatic stress disorder (PTSD). Adenosine A2A receptors (A2AR) control emotional memory and fear conditioning, but it is not known if they affect the consolidation and generalization of fear, which was now investigated. We now report that A2AR blockade through systemic administration of the A2AR antagonist SCH58261 immediately after contextual fear conditioning (within the consolidation window), accelerated fear generalization. Conversely, A2AR activation with CGS21680 decreased fear generalization. Ex vivo electrophysiological recordings of field excitatory post-synaptic potentials (fEPSPs) in CA3-CA1 synapses and of population spikes in the lateral amygdala (LA), showed that the effect of SCH58261 is associated with a reversion of fear conditioning-induced decrease of long-term potentiation (LTP) in the dorsal hippocampus (DH) and with increased amplitude of LA LTP in conditioned animals. These data suggest that A2AR are engaged during contextual fear consolidation, controlling long-term potentiation mechanisms in both DH and LA during fear consolidation, impacting on fear generalization; this supports targeting A2AR during fear consolidation to control aberrant fear processing in PTSD and other fear-related disorders.
Collapse
Affiliation(s)
- Ana P Simões
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Marina A M Portes
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
- Department of Pharmacology, Graduate Program in Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Cátia R Lopes
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Felipe Vanz
- Department of Pharmacology, Graduate Program in Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Vanessa S Lourenço
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Anna Pliássova
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Ingride L Gaspar
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Henrique B Silva
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Ângelo R Tomé
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
- Faculty of Science and Technology, Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Paula M Canas
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Rui D Prediger
- Department of Pharmacology, Graduate Program in Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Rodrigo A Cunha
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal.
- Multidisciplinary Institute of Aging (MIA-Portugal), University of Coimbra, 3004-504, Coimbra, Portugal.
| |
Collapse
|
26
|
Vincenzi F, Pasquini S, Contri C, Cappello M, Nigro M, Travagli A, Merighi S, Gessi S, Borea PA, Varani K. Pharmacology of Adenosine Receptors: Recent Advancements. Biomolecules 2023; 13:1387. [PMID: 37759787 PMCID: PMC10527030 DOI: 10.3390/biom13091387] [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: 08/14/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Adenosine receptors (ARs) are widely acknowledged pharmacological targets yet are still underutilized in clinical practice. Their ubiquitous distribution in almost all cells and tissues of the body makes them, on the one hand, excellent candidates for numerous diseases, and on the other hand, intrinsically challenging to exploit selectively and in a site-specific manner. This review endeavors to comprehensively depict the substantial advancements witnessed in recent years concerning the development of drugs that modulate ARs. Through preclinical and clinical research, it has become evident that the modulation of ARs holds promise for the treatment of numerous diseases, including central nervous system disorders, cardiovascular and metabolic conditions, inflammatory and autoimmune diseases, and cancer. The latest studies discussed herein shed light on novel mechanisms through which ARs exert control over pathophysiological states. They also introduce new ligands and innovative strategies for receptor activation, presenting compelling evidence of efficacy along with the implicated signaling pathways. Collectively, these emerging insights underscore a promising trajectory toward harnessing the therapeutic potential of these multifaceted targets.
Collapse
Affiliation(s)
- Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Silvia Pasquini
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Chiara Contri
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Martina Cappello
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Manuela Nigro
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Alessia Travagli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Stefania Merighi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Stefania Gessi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | | | - Katia Varani
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| |
Collapse
|
27
|
Wang Y, Beukeboom LW, Wertheim B, Hut RA. Transcriptomic Analysis of Light-Induced Genes in Nasonia vitripennis: Possible Implications for Circadian Light Entrainment Pathways. BIOLOGY 2023; 12:1215. [PMID: 37759614 PMCID: PMC10525998 DOI: 10.3390/biology12091215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/31/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023]
Abstract
Circadian entrainment to the environmental day-night cycle is essential for the optimal use of environmental resources. In insects, opsin-based photoreception in the compound eye and ocelli and CRYPTOCHROME1 (CRY1) in circadian clock neurons are thought to be involved in sensing photic information, but the genetic regulation of circadian light entrainment in species without light-sensitive CRY1 remains unclear. To elucidate a possible CRY1-independent light transduction cascade, we analyzed light-induced gene expression through RNA-sequencing in Nasonia vitripennis. Entrained wasps were subjected to a light pulse in the subjective night to reset the circadian clock, and light-induced changes in gene expression were characterized at four different time points in wasp heads. We used co-expression, functional annotation, and transcription factor binding motif analyses to gain insight into the molecular pathways in response to acute light stimulus and to form hypotheses about the circadian light-resetting pathway. Maximal gene induction was found after 2 h of light stimulation (1432 genes), and this included the opsin gene opblue and the core clock genes cry2 and npas2. Pathway and cluster analyses revealed light activation of glutamatergic and GABA-ergic neurotransmission, including CREB and AP-1 transcription pathway signaling. This suggests that circadian photic entrainment in Nasonia may require pathways that are similar to those in mammals. We propose a model for hymenopteran circadian light-resetting that involves opsin-based photoreception, glutamatergic neurotransmission, and gene induction of cry2 and npas2 to reset the circadian clock.
Collapse
Affiliation(s)
- Yifan Wang
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, 9712 CP Groningen, The Netherlands; (L.W.B.); (R.A.H.)
| | | | - Bregje Wertheim
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, 9712 CP Groningen, The Netherlands; (L.W.B.); (R.A.H.)
| | | |
Collapse
|
28
|
Aframian K, Yousef Yengej D, Nwaobi S, Raman S, Faas GC, Charles A. Effects of chronic caffeine on patterns of brain blood flow and behavior throughout the sleep-wake cycle in freely behaving mice. PNAS NEXUS 2023; 2:pgad303. [PMID: 37780231 PMCID: PMC10538474 DOI: 10.1093/pnasnexus/pgad303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/05/2023] [Indexed: 10/03/2023]
Abstract
Caffeine has significant effects on neurovascular activity and behavior throughout the sleep-wake cycle. We used a minimally invasive microchip/video system to continuously record effects of caffeine in the drinking water of freely behaving mice. Chronic caffeine shifted both rest and active phases by up to 2 h relative to the light-dark cycle in a dose-dependent fashion. There was a particular delay in the onset of rapid eye movement (REM) sleep as compared with non-REM sleep during the rest phase. Chronic caffeine increased wakefulness during the active phase and consolidated sleep during the rest phase; overall, there was no net change in the amount of time spent in the wake, sleep, or REM sleep states during caffeine administration. Despite these effects on wakefulness and sleep, chronic caffeine decreased mean cerebral blood volume (CBV) during the active phase and increased mean CBV during the rest phase. Chronic caffeine also increased heart rate variability in both the sleep and wake states. These results provide new insight into the effects of caffeine on the biology of the sleep-wake cycle. Increased blood flow during sleep caused by chronic caffeine may have implications for its potential neuroprotective effects through vascular mechanisms of brain waste clearance.
Collapse
Affiliation(s)
- Kimiya Aframian
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Dmitri Yousef Yengej
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Sinifunanya Nwaobi
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Shrayes Raman
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Guido C Faas
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Andrew Charles
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| |
Collapse
|
29
|
Gangitano E, Baxter M, Voronkov M, Lenzi A, Gnessi L, Ray D. The interplay between macronutrients and sleep: focus on circadian and homeostatic processes. Front Nutr 2023; 10:1166699. [PMID: 37680898 PMCID: PMC10482045 DOI: 10.3389/fnut.2023.1166699] [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: 02/15/2023] [Accepted: 08/04/2023] [Indexed: 09/09/2023] Open
Abstract
Sleep disturbances are an emerging risk factor for metabolic diseases, for which the burden is particularly worrying worldwide. The importance of sleep for metabolic health is being increasingly recognized, and not only the amount of sleep plays an important role, but also its quality. In this review, we studied the evidence in the literature on macronutrients and their influence on sleep, focusing on the mechanisms that may lay behind this interaction. In particular, we focused on the effects of macronutrients on circadian and homeostatic processes of sleep in preclinical models, and reviewed the evidence of clinical studies in humans. Given the importance of sleep for health, and the role of circadian biology in healthy sleep, it is important to understand how macronutrients regulate circadian clocks and sleep homeostasis.
Collapse
Affiliation(s)
- Elena Gangitano
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Matthew Baxter
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Maria Voronkov
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucio Gnessi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - David Ray
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
30
|
Marciante AB, Seven YB, Kelly MN, Perim RR, Mitchell GS. Magnitude and Mechanism of Phrenic Long-term Facilitation Shift Between Daily Rest Versus Active Phase. FUNCTION 2023; 4:zqad041. [PMID: 37753182 PMCID: PMC10519274 DOI: 10.1093/function/zqad041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 09/28/2023] Open
Abstract
Plasticity is a fundamental property of the neural system controlling breathing. One key example of respiratory motor plasticity is phrenic long-term facilitation (pLTF), a persistent increase in phrenic nerve activity elicited by acute intermittent hypoxia (AIH). pLTF can arise from distinct cell signaling cascades initiated by serotonin versus adenosine receptor activation, respectively, and interact via powerful cross-talk inhibition. Here, we demonstrate that the daily rest/active phase and the duration of hypoxic episodes within an AIH protocol have profound impact on the magnitude and mechanism of pLTF due to shifts in serotonin/adenosine balance. Using the historical "standard" AIH protocol (3, 5-min moderate hypoxic episodes), we demonstrate that pLTF magnitude is unaffected by exposure in the midactive versus midrest phase, yet the mechanism driving pLTF shifts from serotonin-dominant (midrest) to adenosine-dominant (midactive). This mechanistic "flip" results from combined influences of hypoxia-evoked adenosine release and daily fluctuations in basal spinal adenosine. Since AIH evokes less adenosine with shorter (15, 1-min) hypoxic episodes, midrest pLTF is amplified due to diminished adenosine constraint on serotonin-driven plasticity; in contrast, elevated background adenosine during the midactive phase suppresses serotonin-dominant pLTF. These findings demonstrate the importance of the serotonin/adenosine balance in regulating the amplitude and mechanism of AIH-induced pLTF. Since AIH is emerging as a promising therapeutic modality to restore respiratory and nonrespiratory movements in people with spinal cord injury or ALS, knowledge of how time-of-day and hypoxic episode duration impact the serotonin/adenosine balance and the magnitude and mechanism of pLTF has profound biological, experimental, and translational implications.
Collapse
Affiliation(s)
- Alexandria B Marciante
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Yasin B Seven
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Mia N Kelly
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Raphael R Perim
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
31
|
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
|
32
|
Schamilow S, Santonja I, Weitzer J, Strohmaier S, Klösch G, Seidel S, Schernhammer E, Papantoniou K. Time Spent Outdoors and Associations with Sleep, Optimism, Happiness and Health before and during the COVID-19 Pandemic in Austria. Clocks Sleep 2023; 5:358-372. [PMID: 37489436 PMCID: PMC10366917 DOI: 10.3390/clockssleep5030027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/06/2023] [Accepted: 06/14/2023] [Indexed: 07/26/2023] Open
Abstract
Social restriction measures (SRM) implemented during the COVID-19 pandemic led to a reduction in time spent outdoors (TSO). The aim of this study was to describe TSO and evaluate its association with sleep outcomes, optimism, happiness and health-status before and during SRM. Two online surveys were conducted in 2017 (N = 1004) and 2020, during SRM (N = 1010), in samples representative of the age, sex and region of the Austrian population. Information on the duration of TSO, sleep, optimism, happiness and health-status was collected. Multivariable-adjusted logistic regression models were used to study the association of TSO with chronic insomnia, short sleep, late chronotype, optimism, happiness and self-rated health-status. The mean TSO was 3.6 h (SD: 2.18) in 2017 and 2.6 h (SD: 1.87) during times of SRM. Men and participants who were older, married or in a partnership and lived in a rural area reported longer TSO. Participants who spent less time outdoors were more likely to report short sleep or a late chronotype in both surveys and, in 2020, also chronic insomnia. Less TSO was associated with lower happiness and optimism levels and poor health-status. Our findings suggest that TSO may be a protective factor for sleep, mood and health, particularly during stressful and uncertain times.
Collapse
Affiliation(s)
- Simon Schamilow
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, 1090 Vienna, Austria
| | - Isabel Santonja
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, 1090 Vienna, Austria
| | - Jakob Weitzer
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, 1090 Vienna, Austria
- Department of Health Promotion and Prevention, Federal Ministry of the Republic of Austria for Social Affairs, Health, Care and Consumer Protection, 1030 Vienna, Austria
| | - Susanne Strohmaier
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, 1090 Vienna, Austria
| | - Gerhard Klösch
- Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Stefan Seidel
- Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Eva Schernhammer
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, 1090 Vienna, Austria
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Kyriaki Papantoniou
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
33
|
Marciante AB, Mitchell GS. Increased spinal adenosine impairs phrenic long-term facilitation in aging rats. J Appl Physiol (1985) 2023; 134:1537-1548. [PMID: 37167263 PMCID: PMC10281789 DOI: 10.1152/japplphysiol.00197.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/26/2023] [Accepted: 05/08/2023] [Indexed: 05/13/2023] Open
Abstract
Moderate acute intermittent hypoxia (mAIH) elicits a form of spinal, respiratory motor plasticity known as phrenic long-term facilitation (pLTF). In middle-aged male and geriatric female rats, mAIH-induced pLTF is attenuated through unknown mechanisms. In young adults, mAIH activates competing intracellular signaling cascades, initiated by serotonin 2 and adenosine 2A (A2A) receptors, respectively. Spinal A2A receptor inhibition enhances mAIH-induced pLTF, meaning, serotonin dominates, and adenosine constrains mAIH-induced plasticity in the daily rest phase. Thus, we hypothesized elevated basal adenosine levels in the ventral cervical spinal cord of aged rats shifts this balance, undermining mAIH-induced pLTF. A selective A2A receptor antagonist (MSX-3) or vehicle was delivered intrathecally at C4 in anesthetized young (3-6 mo) and aged (20-22 mo) Sprague-Dawley rats before mAIH (3,5-min episodes; arterial Po2 = 45-55 mmHg). In young males, spinal A2A receptor inhibition enhanced pLTF (119 ± 5%) vs. vehicle (55 ± 9%), consistent with prior reports. In old males, pLTF was reduced to 25 ± 11%, but A2A receptor inhibition increased pLTF to levels greater than in young males (186 ± 19%). Basal adenosine levels in ventral C3-C5 homogenates are elevated two- to threefold in old vs. young males. These findings advance our understanding of age as a biological variable in phrenic motor plasticity and will help guide translation of mAIH as a therapeutic modality to restore respiratory and nonrespiratory movements in older populations afflicted with clinical disorders that compromise movement.NEW & NOTEWORTHY Advanced age undermines respiratory motor plasticity, specifically phrenic long-term facilitation (pLTF) following moderate acute intermittent hypoxia (mAIH). We report that spinal adenosine increases in aged male rats, undermining mAIH-induced pLTF via adenosine 2A (A2A) receptor activation, an effect reversed by selective spinal adenosine 2A receptor inhibition. These findings advance our understanding of mechanisms that impair neuroplasticity, and the ability to compensate for the onset of lung or neural injury with age, and may guide efforts to harness mAIH as a treatment for clinical disorders that compromise breathing and other movements.
Collapse
Affiliation(s)
- Alexandria B Marciante
- Department of Physical Therapy & McKnight Brain Institute, Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, United States
| | - Gordon S Mitchell
- Department of Physical Therapy & McKnight Brain Institute, Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
34
|
Duo L, Yu X, Hu R, Duan X, Zhou J, Wang K. Sleep disorders in chronic pain and its neurochemical mechanisms: a narrative review. Front Psychiatry 2023; 14:1157790. [PMID: 37324825 PMCID: PMC10267346 DOI: 10.3389/fpsyt.2023.1157790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
Chronic pain (CP) is a prevalent problem, and more than half of patients with CP have sleep disorders. CP comorbidity with sleep disorders imposes immense suffering and seriously affects the patient's quality of life, which is a challenging issue encountered by clinicians. Although the reciprocal interactions between pain and sleep have been studied to some degree, there is still a lack of awareness and comprehensive description of CP comorbidity with sleep disorders. In this narrative review article, we summarize the current knowledge about the present estimates of the prevalence of comorbid sleep disorders in CP patients, sleep detection methods, sleep characterization in CP, and the effect of sleep disorders on CP and current therapies. We also summarize current knowledge of the neurochemical mechanisms of CP comorbidity with sleep disorders. In conclusion, insufficient attention has been paid to the role of sleep disorders in CP patients, and CP patients should be screened for sleep disorders in the clinic. Special attention should be given to a possible risk of drug-drug interaction when using two types of drugs targeting pain and sleep simultaneously. The current insight into the neurobiological mechanisms underlying CP comorbidity with sleep disorders is still rather limited.
Collapse
|
35
|
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
|
36
|
Liu H, Fu R, Zhang Y, Mao L, Zhu L, Zhang L, Liu X, Jiang H. Integrate transcriptomic and metabolomic analysis reveals the underlying mechanisms of behavioral disorders in zebrafish (Danio rerio) induced by imidacloprid. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 870:161541. [PMID: 36731560 DOI: 10.1016/j.scitotenv.2023.161541] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/24/2022] [Accepted: 01/07/2023] [Indexed: 06/18/2023]
Abstract
Imidacloprid, a widely used neonicotinoid insecticide, poses a significant threat to aquatic ecosystems. Behavior is a functional indicator of the net sensory, motor, and integrative processes of the nervous system and is presumed to be more sensitive in detecting toxicity. In the present study, we investigated the behavioral effects of imidacloprid at the level of environmental concentrations (1, 10 and 100 μg/L) for a constant exposure to zebrafish adults, and performed the integrated transcriptomic and metabolomic analysis to analyze the molecular mechanism underlying behavioral effects of imidacloprid. Our results show that imidacloprid exposure significantly induce behavioral disruptions characterized by anxiety, depression, and reduced physiological function including exploratory, decision, social interaction and locomotor activity. Integrated transcriptomic and metabolomic analysis indicate that the disruption of circadian rhythm, metabolic imbalance of arginine and proline, and neurotransmitter disorder are the underlying molecular mechanisms of behavioral impairment induced by imidacloprid. The "gene-metabolite-disease" network consisted by 11 metabolites and 15 genes is associated human disease Alzheimer's disease (AD) and schizophrenia. Our results confirm the behavioral impairment induced by imidacloprid at environmental concentrations for constant exposure. The identified genes and metabolites can be used not only to illustrate the underlying mechanisms, but also can be developed as biomarkers in determining the ecological risk of imidacloprid to aquatic organisms even Homo sapiens.
Collapse
Affiliation(s)
- Hongli Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ruiqiang Fu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yanning Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Liangang Mao
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lizhen Zhu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lan Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Xingang Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Hongyun Jiang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
37
|
Skapetze L, Owino S, Lo EH, Arai K, Merrow M, Harrington M. Rhythms in barriers and fluids: Circadian clock regulation in the aging neurovascular unit. Neurobiol Dis 2023; 181:106120. [PMID: 37044366 DOI: 10.1016/j.nbd.2023.106120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 04/14/2023] Open
Abstract
The neurovascular unit is where two very distinct physiological systems meet: The central nervous system (CNS) and the blood. The permeability of the barriers separating these systems is regulated by time, including both the 24 h circadian clock and the longer processes of aging. An endogenous circadian rhythm regulates the transport of molecules across the blood-brain barrier and the circulation of the cerebrospinal fluid and the glymphatic system. These fluid dynamics change with time of day, and with age, and especially in the context of neurodegeneration. Factors may differ depending on brain region, as can be highlighted by consideration of circadian regulation of the neurovascular niche in white matter. As an example of a potential target for clinical applications, we highlight chaperone-mediated autophagy as one mechanism at the intersection of circadian dysregulation, aging and neurodegenerative disease. In this review we emphasize key areas for future research.
Collapse
Affiliation(s)
- Lea Skapetze
- Institute of Medical Psychology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sharon Owino
- Neuroscience Program, Smith College, Northampton, MA 01060, United States of America
| | - Eng H Lo
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Martha Merrow
- Institute of Medical Psychology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Mary Harrington
- Neuroscience Program, Smith College, Northampton, MA 01060, United States of America.
| |
Collapse
|
38
|
Marciante AB, Mitchell GS. Mild inflammation impairs acute intermittent hypoxia-induced phrenic long-term facilitation by a spinal adenosine-dependent mechanism. J Neurophysiol 2023; 129:799-806. [PMID: 36883762 PMCID: PMC10069977 DOI: 10.1152/jn.00035.2023] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/23/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Inflammation undermines neuroplasticity, including serotonin-dependent phrenic long-term facilitation (pLTF) following moderate acute intermittent hypoxia (mAIH: 3, 5-min episodes, arterial Po2: 40-50 mmHg; 5-min intervals). Mild inflammation elicited by a low dose of the TLR-4 receptor agonist, lipopolysaccharide (LPS; 100 µg/kg, ip), abolishes mAIH-induced pLTF by unknown mechanisms. In the central nervous system, neuroinflammation primes glia, triggering ATP release and extracellular adenosine accumulation. As spinal adenosine 2 A (A2A) receptor activation impairs mAIH-induced pLTF, we hypothesized that spinal adenosine accumulation and A2A receptor activation are necessary in the mechanism whereby LPS impairs pLTF. We report that 24 h after LPS injection in adult male Sprague Dawley rats: 1) adenosine levels increase in ventral spinal segments containing the phrenic motor nucleus (C3-C5; P = 0.010; n = 7/group) and 2) cervical spinal A2A receptor inhibition (MSX-3, 10 µM, 12 µL intrathecal) rescues mAIH-induced pLTF. In LPS vehicle-treated rats (saline, ip), MSX-3 enhanced pLTF versus controls (LPS: 110 ± 16% baseline; controls: 53 ± 6%; P = 0.002; n = 6/group). In LPS-treated rats, pLTF was abolished as expected (4 ± 6% baseline; n = 6), but intrathecal MSX-3 restored pLTF to levels equivalent to MSX-3-treated control rats (120 ± 14% baseline; P < 0.001; n = 6; vs. LPS controls with MSX-3: P = 0.539). Thus, inflammation abolishes mAIH-induced pLTF by a mechanism that requires increased spinal adenosine levels and A2A receptor activation. As repetitive mAIH is emerging as a treatment to improve breathing and nonrespiratory movements in people with spinal cord injury or ALS, A2A inhibition may offset undermining effects of neuroinflammation associated with these neuromuscular disorders.NEW & NOTEWORTHY Mild inflammation undermines motor plasticity elicited by mAIH. In a model of mAIH-induced respiratory motor plasticity (phrenic long-term facilitation; pLTF), we report that inflammation induced by low-dose lipopolysaccharide undermines mAIH-induced pLTF by a mechanism requiring increased cervical spinal adenosine and adenosine 2 A receptor activation. This finding advances the understanding of mechanisms impairing neuroplasticity, potentially undermining the ability to compensate for the onset of lung/neural injury or to harness mAIH as a therapeutic modality.
Collapse
Affiliation(s)
- Alexandria B Marciante
- Breathing Research and Therapeutics Center, Department of Physical Therapy & McKnight Brain Institute, University of Florida, Gainesville, Florida, United States
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy & McKnight Brain Institute, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
39
|
Chen J, Xiao L, Chen Y, Li W, Liu Y, Zhou Y, Tan H. YT521-B homology domain containing 1 ameliorates mitochondrial damage and ferroptosis in sleep deprivation by activating the sirtuin 1/nuclear factor erythroid-derived 2-like 2/heme oxygenase 1 pathway. Brain Res Bull 2023; 197:1-12. [PMID: 36935054 DOI: 10.1016/j.brainresbull.2023.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/01/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023]
Abstract
In sleep deprivation (SD) models, ferroptosis is increased. SIRT1 alleviates cognitive impairment in SD, and SIRT1/NRF2/HO1 pathway depresses ferroptosis in different diseases. Moreover, YTHDC1 can regulate SIRT1 mRNA stability. Therefore, our study explored effects of the YTHDC1/SIRT1/NRF2/HO1 axis on neuronal damage and ferroptosis in SD. The SD mouse model was established through a modified multi-platform water environment method and a cell model of ferroptosis was constructed with Erastin, followed by gain- and loss-of-function assays. In mice, the cognitive impairment and CLOCK and BMAL1 levels in hippocampal tissues were assessed. In cells, viability was measured. In mice and cells, mitochondrial ultrastructure, the content of reactive oxygen species (ROS), malondialdehyde (MDA), glutathione (GSH), and iron, and the expression of GPX4 and ACSL4 were detected. The potential relationships among YTHDC1, SIRT1, and NRF2 were analyzed. SD mice had downregulated YTHDC1, SIRT1, NRF2, and HO1 protein expression in hippocampal tissues and increased ferroptosis. Mechanically, SIRT1 activated the NRF2/HO1 pathway through deacetylation, and YTHDC1 increased SIRT1 mRNA stability. YTHDC1 overexpression diminished mitochondrial damage, the content of ROS, iron, and MDA, and the expression of ACSL4 while enhancing GSH contents and GPX4 expression in hippocampal tissues of SD mice and Erastin-induced HT22 cells. Additionally, YTHDC1 overexpression elevated viability in Erastin-induced HT22 cells. SIRT1 or NRF2 overexpression ameliorated Erastin-induced mitochondrial damage and ferroptosis in HT22 cells. Silencing SIRT1 abolished the impact of YTHDC1 overexpression on SD mice and Erastin-induced HT22 cells. Collectively, YTHDC1 ameliorates mitochondrial damage and ferroptosis after SD by activating the SIRT1/NRF2/HO1 pathway.
Collapse
Affiliation(s)
- Juan Chen
- Department of Neurology, the First Hospital of Changsha, Changsha, Hunan 410005, P.R. China.
| | - Lijun Xiao
- Department of Neurology, the First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Ying Chen
- Department of Neurology, the First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Wei Li
- Department of Neurology, the First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Yinan Liu
- Department of Neurology, the First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Ying Zhou
- Department of Neurology, the First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Hong Tan
- Department of Neurology, the First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| |
Collapse
|
40
|
Astrocyte strategies in the energy-efficient brain. Essays Biochem 2023; 67:3-16. [PMID: 36350053 DOI: 10.1042/ebc20220077] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/10/2022]
Abstract
Astrocytes generate ATP through glycolysis and mitochondrion respiration, using glucose, lactate, fatty acids, amino acids, and ketone bodies as metabolic fuels. Astrocytic mitochondria also participate in neuronal redox homeostasis and neurotransmitter recycling. In this essay, we aim to integrate the multifaceted evidence about astrocyte bioenergetics at the cellular and systems levels, with a focus on mitochondrial oxidation. At the cellular level, the use of fatty acid β-oxidation and the existence of molecular switches for the selection of metabolic mode and fuels are examined. At the systems level, we discuss energy audits of astrocytes and how astrocytic Ca2+ signaling might contribute to the higher performance and lower energy consumption of the brain as compared to engineered circuits. We finish by examining the neural-circuit dysregulation and behavior impairment associated with alterations of astrocytic mitochondria. We conclude that astrocytes may contribute to brain energy efficiency by coupling energy, redox, and computational homeostasis in neural circuits.
Collapse
|
41
|
Taylor L, Von Lendenfeld F, Ashton A, Sanghani H, Di Pretoro S, Usselmann L, Veretennikova M, Dallmann R, McKeating JA, Vasudevan S, Jagannath A. Sleep and circadian rhythm disruption alters the lung transcriptome to predispose to viral infection. iScience 2023; 26:105877. [PMID: 36590897 PMCID: PMC9788990 DOI: 10.1016/j.isci.2022.105877] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 10/11/2022] [Accepted: 12/21/2022] [Indexed: 12/26/2022] Open
Abstract
Sleep and circadian rhythm disruption (SCRD), as encountered during shift work, increases the risk of respiratory viral infection including SARS-CoV-2. However, the mechanism(s) underpinning higher rates of respiratory viral infection following SCRD remain poorly characterized. To address this, we investigated the effects of acute sleep deprivation on the mouse lung transcriptome. Here we show that sleep deprivation profoundly alters the transcriptional landscape of the lung, causing the suppression of both innate and adaptive immune systems, disrupting the circadian clock, and activating genes implicated in SARS-CoV-2 replication, thereby generating a lung environment that could promote viral infection and associated disease pathogenesis. Our study provides a mechanistic explanation of how SCRD increases the risk of respiratory viral infections including SARS-CoV-2 and highlights possible therapeutic avenues for the prevention and treatment of respiratory viral infection.
Collapse
Affiliation(s)
- Lewis Taylor
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, New Biochemistry Building, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Felix Von Lendenfeld
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, New Biochemistry Building, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Anna Ashton
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, New Biochemistry Building, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Harshmeena Sanghani
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Simona Di Pretoro
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, New Biochemistry Building, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Laura Usselmann
- Division of Biomedical Sciences, Warwick Medical School, Interdisciplinary Biomedical Research Building, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK
| | - Maria Veretennikova
- Zeeman Institute for Systems Biology & Infectious Disease Epidemiology Research, Department of Mathematics, Mathematical Sciences Building, University of Warwick, Coventry CV4 7AL, UK
| | - Robert Dallmann
- Division of Biomedical Sciences, Warwick Medical School, Interdisciplinary Biomedical Research Building, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK
| | - Jane A. McKeating
- Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford OX3 7BN, UK
- Chinese Academy of Medical Sciences (CAMS) Oxford Institute (COI), University of Oxford, Old Road Campus, Oxford OX3 7BN, UK
| | - Sridhar Vasudevan
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Aarti Jagannath
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, New Biochemistry Building, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| |
Collapse
|
42
|
Raciti L, Raciti G, Militi D, Tonin P, Quartarone A, Calabrò RS. Sleep in Disorders of Consciousness: A Brief Overview on a Still under Investigated Issue. Brain Sci 2023; 13:275. [PMID: 36831818 PMCID: PMC9954700 DOI: 10.3390/brainsci13020275] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/29/2023] [Accepted: 02/06/2023] [Indexed: 02/09/2023] Open
Abstract
Consciousness is a multifaceted concept, involving both wakefulness, i.e., a condition of being alert that is regulated by the brainstem, and awareness, a subjective experience of any thoughts or perception or emotion. Recently, the European Academy of Neurology has published international guidelines for a better diagnosis of coma and other disorders of consciousness (DOC) through the investigation of sleep patterns, such as slow-wave and REM, and the study of the EEG using machine learning methods and artificial intelligence. The management of sleep disorders in DOC patients is an increasingly hot topic and deserves careful diagnosis, to allow for the most accurate prognosis and the best medical treatment possible. The aim of this review was to investigate the anatomo-physiological basis of the sleep/wake cycle, as well as the main sleep patterns and sleep disorders in patients with DOC. We found that the sleep characteristics in DOC patients are still controversial. DOC patients often present a theta/delta pattern, while epileptiform activity, as well as other sleep elements, have been reported as correlating with outcomes in patients with coma and DOC. The absence of spindles, as well as REM and K-complexes of NREM sleep, have been used as poor predictors for early awakening in DOC patients, especially in UWS patients. Therefore, sleep could be considered a marker of DOC recovery, and effective treatments for sleep disorders may either indirectly or directly favor recovery of consciousness.
Collapse
Affiliation(s)
| | | | - David Militi
- IRCCS Centro Neurolesi Bonino Pulejo, 98121 Messina, Italy
| | | | | | | |
Collapse
|
43
|
Ono D, Wang H, Hung CJ, Wang HT, Kon N, Yamanaka A, Li Y, Sugiyama T. Network-driven intracellular cAMP coordinates circadian rhythm in the suprachiasmatic nucleus. SCIENCE ADVANCES 2023; 9:eabq7032. [PMID: 36598978 PMCID: PMC11318661 DOI: 10.1126/sciadv.abq7032] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 11/23/2022] [Indexed: 06/17/2023]
Abstract
The mammalian central circadian clock, located in the suprachiasmatic nucleus (SCN), coordinates the timing of physiology and behavior to local time cues. In the SCN, second messengers, such as cAMP and Ca2+, are suggested to be involved in the input and/or output of the molecular circadian clock. However, the functional roles of second messengers and their dynamics in the SCN remain largely unclear. In the present study, we visualized the spatiotemporal patterns of circadian rhythms of second messengers and neurotransmitter release in the SCN. Here, we show that neuronal activity regulates the rhythmic release of vasoactive intestinal peptides from the SCN, which drives the circadian rhythms of intracellular cAMP in the SCN. Furthermore, optical manipulation of intracellular cAMP levels in the SCN shifts molecular and behavioral circadian rhythms. Together, our study demonstrates that intracellular cAMP is a key molecule in the organization of the SCN circadian neuronal network.
Collapse
Affiliation(s)
- Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Huan Wang
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Chi Jung Hung
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hsin-tzu Wang
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
- Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
- Department of Biological Sciences, School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Naohiro Kon
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
- Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Akihiro Yamanaka
- Chinese Institute for Brain Research (CIBR), Beijing, 102206, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Takashi Sugiyama
- Advanced Optics and Biological Engineering, Evident Corporation, Tokyo, Japan
| |
Collapse
|
44
|
Lee J, Kang J, Kim Y, Lee S, Oh CM, Kim T. Integrated analysis of the microbiota-gut-brain axis in response to sleep deprivation and diet-induced obesity. Front Endocrinol (Lausanne) 2023; 14:1117259. [PMID: 36896179 PMCID: PMC9990496 DOI: 10.3389/fendo.2023.1117259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
INTRODUCTION Sleep deprivation (SD) and obesity are common in modern societies. SD and obesity frequently coexist, but research on the combined consequences of SD and obesity has been limited. In this study, we investigated the gut microbiota and host responses to SD and high-fat diet (HFD)-induced obesity. In addition, we attempted to identify key mediators of the microbiota-gut-brain axis. METHODS C57BL/6J mice were divided into four groups based on whether they were sleep deprived and whether they were fed a standard chow diet (SCD) or HFD. We then performed fecal microbiome shotgun sequencing, gut transcriptome analysis using RNA sequencing, and brain mRNA expression analysis using the nanoString nCounter Mouse Neuroinflammation Panel. RESULTS The gut microbiota was significantly altered by the HFD, whereas the gut transcriptome was primarily influenced by SD. Sleep and diet are both important in the inflammatory system of the brain. When SD and the HFD were combined, the inflammatory system of the brain was severely disrupted. In addition, inosine-5' phosphate may be the gut microbial metabolite that mediates microbiota-gut-brain interactions. To identify the major drivers of this interaction, we analyzed the multi-omics data. Integrative analysis revealed two driver factors that were mostly composed of the gut microbiota. We discovered that the gut microbiota may be the primary driver of microbiota-gut-brain interactions. DISCUSSION These findings imply that healing gut dysbiosis may be a viable therapeutic target for enhancing sleep quality and curing obesity-related dysfunction.
Collapse
Affiliation(s)
- Jibeom Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jiseung Kang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Yumin Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Sunjae Lee
- Department of School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
- *Correspondence: Sunjae Lee, ; Chang-Myung Oh, ; Tae Kim,
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
- *Correspondence: Sunjae Lee, ; Chang-Myung Oh, ; Tae Kim,
| | - Tae Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
- *Correspondence: Sunjae Lee, ; Chang-Myung Oh, ; Tae Kim,
| |
Collapse
|
45
|
Szczepkowska A, Bochenek J, Wójcik M, Tomaszewska-Zaremba D, Antushevich H, Tomczyk M, Skipor J, Herman A. Effect of caffeine on adenosine and ryanodine receptor
gene expression in the hypothalamus, pituitary, and choroid
plexus in ewes under basal and LPS challenge conditions. JOURNAL OF ANIMAL AND FEED SCIENCES 2022. [DOI: 10.22358/jafs/156762/2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
46
|
Harmsen JF, van Weeghel M, Parsons R, Janssens GE, Wefers J, van Moorsel D, Hansen J, Hoeks J, Hesselink MKC, Houtkooper RH, Schrauwen P. Divergent remodeling of the skeletal muscle metabolome over 24 h between young, healthy men and older, metabolically compromised men. Cell Rep 2022; 41:111786. [PMID: 36516749 DOI: 10.1016/j.celrep.2022.111786] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/28/2022] [Accepted: 11/15/2022] [Indexed: 12/15/2022] Open
Abstract
24 h whole-body substrate metabolism and the circadian clock within skeletal muscle are both compromised upon metabolic disease in humans. Here, we assessed the 24 h muscle metabolome by serial muscle sampling performed under 24 h real-life conditions in young, healthy (YH) men versus older, metabolically compromised (OMC) men. We find that metabolites associated with the initial steps of glycolysis and hexosamine biosynthesis are higher in OMC men around the clock, whereas metabolites associated with glutamine-alpha-ketoglutarate, ketone, and redox metabolism are lower in OMC men. The night period shows the largest number of differently expressed metabolites. Both groups demonstrate 24 h rhythmicity in half of the metabolome, but rhythmic metabolites only partially overlap. Specific metabolites are only rhythmic in YH men (adenosine), phase shifted in OMC men (cis-aconitate, flavin adenine dinucleotide [FAD], and uridine diphosphate [UDP]), or have a reduced 24 h amplitude in OMC men (hydroxybutyrate and hippuric acid). Our data highlight the plasticity of the skeletal muscle metabolome over 24 h and large divergence across the metabolic health spectrum.
Collapse
Affiliation(s)
- Jan-Frieder Harmsen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6200 MD Maastricht, the Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Core Facility Metabolomics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rex Parsons
- Australian Centre for Health Services Innovation and Centre for Healthcare Transformation, School of Public Health and Social Work, Faculty of Health, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - Georges E Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Jakob Wefers
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6200 MD Maastricht, the Netherlands
| | - Dirk van Moorsel
- Division of Endocrinology, Department of Internal Medicine, Maastricht University Medical Center, 6200 MD Maastricht, the Netherlands
| | - Jan Hansen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6200 MD Maastricht, the Netherlands
| | - Joris Hoeks
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6200 MD Maastricht, the Netherlands
| | - Matthijs K C Hesselink
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6200 MD Maastricht, the Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6200 MD Maastricht, the Netherlands.
| |
Collapse
|
47
|
Klerman EB, Brager A, Carskadon MA, Depner CM, Foster R, Goel N, Harrington M, Holloway PM, Knauert MP, LeBourgeois MK, Lipton J, Merrow M, Montagnese S, Ning M, Ray D, Scheer FAJL, Shea SA, Skene DJ, Spies C, Staels B, St‐Onge M, Tiedt S, Zee PC, Burgess HJ. Keeping an eye on circadian time in clinical research and medicine. Clin Transl Med 2022; 12:e1131. [PMID: 36567263 PMCID: PMC9790849 DOI: 10.1002/ctm2.1131] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Daily rhythms are observed in humans and almost all other organisms. Most of these observed rhythms reflect both underlying endogenous circadian rhythms and evoked responses from behaviours such as sleep/wake, eating/fasting, rest/activity, posture changes and exercise. For many research and clinical purposes, it is important to understand the contribution of the endogenous circadian component to these observed rhythms. CONTENT The goal of this manuscript is to provide guidance on best practices in measuring metrics of endogenous circadian rhythms in humans and promote the inclusion of circadian rhythms assessments in studies of health and disease. Circadian rhythms affect all aspects of physiology. By specifying minimal experimental conditions for studies, we aim to improve the quality, reliability and interpretability of research into circadian and daily (i.e., time-of-day) rhythms and facilitate the interpretation of clinical and translational findings within the context of human circadian rhythms. We describe protocols, variables and analyses commonly used for studying human daily rhythms, including how to assess the relative contributions of the endogenous circadian system and other daily patterns in behaviours or the environment. We conclude with recommendations for protocols, variables, analyses, definitions and examples of circadian terminology. CONCLUSION Although circadian rhythms and daily effects on health outcomes can be challenging to distinguish in practice, this distinction may be important in many clinical settings. Identifying and targeting the appropriate underlying (patho)physiology is a medical goal. This review provides methods for identifying circadian effects to aid in the interpretation of published work and the inclusion of circadian factors in clinical research and practice.
Collapse
Affiliation(s)
- Elizabeth B. Klerman
- Department of NeurologyMassachusetts General Hospital, Brigham and Women's HospitalBostonMassachusettsUSA
- Division of Sleep MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Allison Brager
- PlansAnalysis, and FuturesJohn F. Kennedy Special Warfare Center and SchoolFort BraggNorth CarolinaUSA
| | - Mary A. Carskadon
- Alpert Medical School of Brown UniversityDepartment of Psychiatry and Human BehaviorEP Bradley HospitalChronobiology and Sleep ResearchProvidenceRhode IslandUSA
| | | | - Russell Foster
- Sir Jules Thorn Sleep and Circadian Neuroscience InstituteNuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Namni Goel
- Biological Rhythms Research LaboratoryDepartment of Psychiatry and Behavioral SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Mary Harrington
- Neuroscience ProgramSmith CollegeNorthamptonMassachusettsUSA
| | | | - Melissa P. Knauert
- Section of PulmonaryCritical Care, and Sleep MedicineDepartment of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Monique K. LeBourgeois
- Sleep and Development LaboratoryDepartment of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Jonathan Lipton
- Boston Children's Hospital and Kirby Neurobiology CenterBostonMassachusettsUSA
| | - Martha Merrow
- Institute of Medical PsychologyFaculty of MedicineLMUMunichGermany
| | - Sara Montagnese
- Department of MedicineUniversity of PadovaPadovaItaly
- ChronobiologyFaculty of Health and Medical SciencesUniversity of SurreyGuildfordUK
| | - Mingming Ning
- Clinical Proteomics Research Center and Cardio‐Neurology DivisionMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - David Ray
- NIHR Oxford Biomedical Research CentreJohn Radcliffe HospitalOxfordUK
- Oxford Centre for DiabetesEndocrinology and MetabolismUniversity of OxfordOxfordUK
| | - Frank A. J. L. Scheer
- Division of Sleep MedicineHarvard Medical SchoolBostonMassachusettsUSA
- Medical Chronobiology ProgramDivision of Sleep and Circadian DisordersDepartments of Medicine and NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Steven A. Shea
- Oregon Institute of Occupational Health SciencesOregon Health and Science UniversityPortlandOregonUSA
| | - Debra J. Skene
- ChronobiologyFaculty of Health and Medical SciencesUniversity of SurreyGuildfordUK
| | - Claudia Spies
- Department of Anesthesiology and Intensive Care MedicineCharité – Universitaetsmedizin BerlinBerlinGermany
| | - Bart Staels
- UnivLilleInsermCHU LilleInstitut Pasteur de LilleU1011‐EGIDLilleFrance
| | - Marie‐Pierre St‐Onge
- Division of General Medicine and Center of Excellence for Sleep and Circadian ResearchDepartment of MedicineColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Steffen Tiedt
- Institute for Stroke and Dementia ResearchUniversity HospitalLMUMunichGermany
| | - Phyllis C. Zee
- Center for Circadian and Sleep MedicineDivision of Sleep MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Helen J. Burgess
- Sleep and Circadian Research LaboratoryDepartment of PsychiatryUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
48
|
Dai HR, Guo HL, Hu YH, Xu J, Ding XS, Cheng R, Chen F. Precision caffeine therapy for apnea of prematurity and circadian rhythms: New possibilities open up. Front Pharmacol 2022; 13:1053210. [PMID: 36532766 PMCID: PMC9753576 DOI: 10.3389/fphar.2022.1053210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/18/2022] [Indexed: 09/10/2024] Open
Abstract
Caffeine is the globally consumed psychoactive substance and the drug of choice for the treatment of apnea of prematurity (AOP), but its therapeutic effects are highly variable among preterm infants. Many of the molecular underpinnings of the marked individual response have remained elusive yet. Interestingly, the significant association between Clock gene polymorphisms and the response to caffeine therapy offers an opportunity to advance our understanding of potential mechanistic pathways. In this review, we delineate the functions and mechanisms of human circadian rhythms. An up-to-date advance of the formation and ontogeny of human circadian rhythms during the perinatal period are concisely discussed. Specially, we summarize and discuss the characteristics of circadian rhythms in preterm infants. Second, we discuss the role of caffeine consumption on the circadian rhythms in animal models and human, especially in neonates and preterm infants. Finally, we postulate how circadian-based therapeutic initiatives could open new possibilities to promote precision caffeine therapy for the AOP management in preterm infants.
Collapse
Affiliation(s)
- Hao-Ran Dai
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hong-Li Guo
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Ya-Hui Hu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Xu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Xuan-Sheng Ding
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Rui Cheng
- Neonatal Intensive Care Unit, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Feng Chen
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
49
|
Lu M, He X, Jiao Z, Hu Z, Guo Z, Dai S, Wang H, Xu D. The upregulation of glutamate decarboxylase 67 against hippocampal excitability damage in male fetal rats by prenatal caffeine exposure. ENVIRONMENTAL TOXICOLOGY 2022; 37:2703-2717. [PMID: 35917217 DOI: 10.1002/tox.23630] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/25/2022] [Accepted: 07/13/2022] [Indexed: 06/15/2023]
Abstract
As a kind of xanthine alkaloid, caffeine is widely present in beverages, food, and analgesic drugs. Our previous studies have shown that prenatal caffeine exposure (PCE) can induce programmed hypersensitivity of the hypothalamic-pituitary-adrenal (HPA) axis in offspring rats, which is involved in developing many chronic adult diseases. The present study further examined the potential molecular mechanism and toxicity targets of hippocampal dysfunction, which might mediate the programmed hypersensitivity of the HPA axis in offspring. Pregnant rats were intragastrically administered with 0, 30, and 120 mg/kg/day caffeine from gestational days (GD) 9-20, and the fetal rats were extracted at GD20. Rat fetal hippocampal H19-7/IGF1R cell line was treated with caffeine, adenosine A2A receptor (A2AR) agonist (CGS-21680) or adenylate cyclase agonist (forskolin) plus caffeine. Compared with the control group, hippocampal neurons of male fetal rats by PCE displayed increased apoptosis and reduced synaptic plasticity, whereas glutamate decarboxylase 67 (GAD67) expression was increased. Moreover, the expression of A2AR was down-regulated, PCE inhibited the cAMP/PKA/CREB/BDNF/TrkB pathway. Furthermore, the results in vitro were consistent with the in vivo study. Both CGS21680 and forskolin could reverse the above alteration caused by caffeine. These results indicated that PCE inhibits the BDNF pathway and mediates the hippocampus's glutamate (Glu) excitotoxicity. The compensatory up-regulation of GAD67 unbalanced the Glu/gamma-aminobutyric acid (GABA)ergic output, leading to the impaired negative feedback to the hypothalamus and hypersensitivity of the HPA axis.
Collapse
Affiliation(s)
- Mengxi Lu
- Department of Pharmacology, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Xia He
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Zhexiao Jiao
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Zewen Hu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Zijing Guo
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Shiyun Dai
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Hui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Dan Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| |
Collapse
|
50
|
A Pattern to Link Adenosine Signaling, Circadian System, and Potential Final Common Pathway in the Pathogenesis of Major Depressive Disorder. Mol Neurobiol 2022; 59:6713-6723. [PMID: 35999325 PMCID: PMC9525429 DOI: 10.1007/s12035-022-03001-3] [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: 02/15/2022] [Accepted: 08/07/2022] [Indexed: 11/18/2022]
Abstract
Several studies have reported separate roles of adenosine receptors and circadian clockwork in major depressive disorder. While less evidence exists for regulation of the circadian clock by adenosine signaling, a small number of studies have linked the adenosinergic system, the molecular circadian clock, and mood regulation. In this article, we review relevant advances and propose that adenosine receptor signaling, including canonical and other alternative downstream cellular pathways, regulates circadian gene expression, which in turn may underlie the pathogenesis of mood disorders. Moreover, we summarize the convergent point of these signaling pathways and put forward a pattern by which Homer1a expression, regulated by both cAMP-response element binding protein (CREB) and circadian clock genes, may be the final common pathogenetic mechanism in depression.
Collapse
|