1
|
Chen H, Xing Y, Wan C, Zhang Z, Shi Z, Liang Y, Jin C, Chen Y, Zhou X, Xu J, Ptáček LJ, Fu YH, Shi G. The SIK3-N783Y mutation is associated with the human natural short sleep trait. Proc Natl Acad Sci U S A 2025; 122:e2500356122. [PMID: 40324078 DOI: 10.1073/pnas.2500356122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/26/2025] [Indexed: 05/07/2025] Open
Abstract
Sleep is an essential component of our daily life. A mutation in human salt induced kinase 3 (hSIK3), which is critical for regulating sleep duration and depth in rodents, is associated with natural short sleep (NSS), a condition characterized by reduced daily sleep duration in human subjects. This NSS hSIK3-N783Y mutation results in diminished kinase activity in vitro. In a mouse model, the presence of the NSS hSIK3-N783Y mutation leads to a decrease in sleep time and an increase in electroencephalogram delta power. At the phosphoproteomic level, the SIK3-N783Y mutation induces substantial changes predominantly at synaptic sites. Bioinformatic analysis has identified several sleep-related kinase alterations triggered by the SIK3-N783Y mutation, including changes in protein kinase A and mitogen-activated protein kinase. These findings underscore the conserved function of SIK3 as a critical gene in human sleep regulation and provide insights into the kinase regulatory network governing sleep.
Collapse
Affiliation(s)
- Hongmin Chen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528451, China
| | - Ye Xing
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528451, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200003, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Chunyan Wan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zheng Zhang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528451, China
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhu Shi
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yutao Liang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528451, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Chunlai Jin
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528451, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200003, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Yating Chen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528451, China
| | - Xia Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528451, China
| | - Junyu Xu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528451, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200003, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Louis J Ptáček
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143
- Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA 94143
| | - Ying-Hui Fu
- Department of Neurology, University of California San Francisco, San Francisco, CA 94143
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143
- Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA 94143
| | - Guangsen Shi
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528451, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200003, China
- University of Chinese Academy of Sciences, Beijing 100190, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
2
|
Prakash BA, Shah I, Ni G, Vasudevan S, Jagannath A, Foster RG. Dreaming of Better Treatments: Advances in Drug Development for Sleep Medicine and Chronotherapy. J Sleep Res 2025:e70087. [PMID: 40346938 DOI: 10.1111/jsr.70087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/12/2025]
Abstract
Throughout history, the development of new sleep medicines has been driven by progress in our understanding of the mechanisms underlying sleep. Ancient civilisations used their understanding of the sedative nature of natural herbs and compounds to induce sleep. The discovery of barbiturates and bromides heralded a new era of synthetic sleep medicine in the 19th century. This was followed by the development of benzodiazepines that were used to inhibit signalling throughout the brain by promoting gamma-amino butyric acid release and thereby produce loss of consciousness. As our understanding of sleep has deepened, newer therapies have more specifically targeted the wake-inducing neurotransmitter orexin with fewer side effects. Given the newly highlighted role of kinases in sleep/wake regulation, we predict that the next breakthroughs in sleep medicine will likely target these kinases. Given the fundamental role that sleep plays in maintaining brain health through processes such as glymphatic clearance, sleep medicine has therapeutic potential beyond just sleep. Recent evidence suggests that sleep disruptions directly contribute to the build-up of pathological neuronal proteins in neurodegenerative disorders. Therefore, sleep medicine could improve prognosis in disorders such as these. Great attention must be paid to the mechanism of action of each sleep medicine, however, as sleep medicines which do not fully mimic sleep could actually worsen disease progression.
Collapse
Affiliation(s)
| | - Ishani Shah
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Guohao Ni
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | | - Aarti Jagannath
- Nuffield Department of Clinical Neurosciences, Sleep and Circadian Neuroscience Institute (SCNi), University of Oxford, Oxford, UK
| | - Russell G Foster
- Nuffield Department of Clinical Neurosciences, Sleep and Circadian Neuroscience Institute (SCNi), University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Wang W, Zhao L, He Z, Zhao Y, Jiang G, Gong C, Zhang Y, Yu J, Liang T, Guo L. Decoding Multifaceted Roles of Sleep-Related Genes as Molecular Bridges in Chronic Disease Pathogenesis. Int J Mol Sci 2025; 26:2872. [PMID: 40243466 PMCID: PMC11988575 DOI: 10.3390/ijms26072872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/01/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Sleep is a fundamental process essential for all organisms. Sleep deprivation can lead to significant detrimental effects, contributing to various physiological disorders and elevating the risk of several diseases. Investigating the relationship between sleep and human diseases offers valuable insights into the molecular mechanisms governing sleep regulation, potentially guiding the development of more effective treatments for sleep disorders and associated diseases. This study explored the roles of sleep-related genes in biological processes and their associations with chronic diseases, mainly including neurological, metabolic, cardiovascular diseases, and cancer. Additionally, an analysis on the sleep-related genes was also performed to understand the potential role in tumorigenesis. This review aims to enhance the understanding of the link between sleep-related genes and chronic diseases, contributing to the development of novel therapeutic approaches targeting sleep and circadian rhythm-related chronic diseases.
Collapse
Affiliation(s)
- Wenyuan Wang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Chemistry and Life Sciences, Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (W.W.); (L.Z.); (Z.H.); (Y.Z.); (C.G.); (Y.Z.)
| | - Linjie Zhao
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Chemistry and Life Sciences, Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (W.W.); (L.Z.); (Z.H.); (Y.Z.); (C.G.); (Y.Z.)
| | - Zhiheng He
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Chemistry and Life Sciences, Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (W.W.); (L.Z.); (Z.H.); (Y.Z.); (C.G.); (Y.Z.)
| | - Yang Zhao
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Chemistry and Life Sciences, Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (W.W.); (L.Z.); (Z.H.); (Y.Z.); (C.G.); (Y.Z.)
| | - Guijie Jiang
- School of Life Science, Nanjing Normal University, Nanjing 210023, China;
| | - Chengjun Gong
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Chemistry and Life Sciences, Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (W.W.); (L.Z.); (Z.H.); (Y.Z.); (C.G.); (Y.Z.)
| | - Yan Zhang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Chemistry and Life Sciences, Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (W.W.); (L.Z.); (Z.H.); (Y.Z.); (C.G.); (Y.Z.)
| | - Jiafeng Yu
- Shandong Provincial Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, China;
| | - Tingming Liang
- School of Life Science, Nanjing Normal University, Nanjing 210023, China;
| | - Li Guo
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Chemistry and Life Sciences, Nanjing University of Posts & Telecommunications, Nanjing 210023, China; (W.W.); (L.Z.); (Z.H.); (Y.Z.); (C.G.); (Y.Z.)
| |
Collapse
|
4
|
Savy V, Stein P, Delker D, Estermann MA, Papas BN, Xu Z, Radonova L, Williams CJ. Calcium signals shape metabolic control of H3K27ac and H3K18la to regulate EGA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643362. [PMID: 40161793 PMCID: PMC11952514 DOI: 10.1101/2025.03.14.643362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The use of assisted reproductive technologies (ART) has enabled the birth of over 9 million babies; but it is associated with increased risks of negative metabolic outcomes in offspring. Yet, the underlying mechanism remains unknown. Calcium (Ca2+) signals, which initiate embryo development at fertilization, are frequently disrupted in human ART. In mice, abnormal Ca2+ signals at fertilization impair embryo development and adult offspring metabolism. Changes in intracellular Ca2+ drive mitochondrial activity and production of metabolites used by the epigenetic machinery. For example, acetyl-CoA (derived mainly from pyruvate) and lactyl-CoA (derived from lactate) are used for writing H3K27ac and H3K18la marks that orchestrate initiation of development. Using both a genetic mouse model and treatment with ionomycin to raise intracellular Ca2+ of wild-type fertilized eggs, we found that excess Ca2+ at fertilization changes metabolic substrate availability, causing epigenetic changes that impact embryo development and offspring health. Specifically, increased Ca2+ exposure at fertilization led to increased H3K27ac levels and decreased H3K18la levels at the 1-cell (1C) stage, that persisted until the 2-cell (2C) stage. Ultralow input CUT&Tag revealed significant differences in H3K27ac and H3K18la genomic profiles between control and ionomycin groups. In addition, increased Ca2+ exposure resulted in a marked reduction in global transcription at the 1C stage that persisted through the 2C stage due to diminished activity of RNA polymerase I. Excess Ca2+ following fertilization increased pyruvate dehydrogenase activity (enzyme that converts pyruvate to acetyl-CoA) and decreased total lactate levels. Provision of exogenous lactyl-CoA before ionomycin treatment restored H3K18la levels at the 1C and 2C stages and rescued global transcription to control levels. Our findings demonstrate conclusively that Ca2+ dynamics drive metabolic regulation of epigenetic reprogramming at fertilization and alter EGA.
Collapse
Affiliation(s)
- Virginia Savy
- Reproductive Medicine Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Paula Stein
- Reproductive Medicine Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Don Delker
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Martín A. Estermann
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Brian N. Papas
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Zongli Xu
- Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Lenka Radonova
- Reproductive Medicine Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Carmen J. Williams
- Reproductive Medicine Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| |
Collapse
|
5
|
Zhang Y, Yang G, Jin Q, Shi T, Chen X, Zhang R, Wang C, Li L. In Situ Mass Spectrometry Imaging to Elucidate the Effects of an Adenosine A 2A Receptor Agonist and Alprazolam on Sleep Regulation. ACS Pharmacol Transl Sci 2025; 8:841-853. [PMID: 40109753 PMCID: PMC11915180 DOI: 10.1021/acsptsci.4c00707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 03/22/2025]
Abstract
Alprazolam (Alp), a commonly used sleep medication in clinical practice, has several potential limitations, including a narrow therapeutic dosage range and a delayed sleep onset. CGS21680 (CGS), a selective agonist of the adenosine A2A receptor, exhibits neuroinhibitory properties. This study aimed to evaluate the effects of CGS on the sleep properties of Alp. The sleep-inducing effects of Alp were assessed through the righting reflex, while the sedative effects of CGS were evaluated by spontaneous activity detection. The synergistic effect of CGS on Alp was evaluated by using electroencephalography and electromyography. The results indicate that we optimized and selected ED5 dose of Alp and ED50 dose of CGS for coadministration. CGS reduced the sleep latency induced by Alp and extended the sleep duration. The distribution of Alp in the brain was assessed through mass spectrometry imaging (MSI). The blood-brain barrier (BBB) model was established to evaluate the impact of CGS on the transmittance of Alp. The results indicated that CGS influenced the distribution of Alp across various brain regions and increased Alp's transmittance across the BBB. The metabolic pathways of GABA, glutamate, and glutamine were assessed through MSI and enzyme activity verification. The coadministration of Alp and CGS resulted in the regulation of GABA, glutamate, and glutamine during the sleep latency and sleep maintenance periods, respectively. In conclusion, the potentiating effect of CGS on the sleep-inducing properties of Alp is attributed to its ability to modulate the distribution of Alp in the brain by enhancing BBB permeability and its influence on Alp-induced neurotransmitter release.
Collapse
Affiliation(s)
- Yi Zhang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Guixiang Yang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Qian Jin
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Tong Shi
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Xuejun Chen
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Ruihua Zhang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Chen Wang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Liqin Li
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| |
Collapse
|
6
|
Ye M, Diering GH. Calcineurin goes to sleep. Proc Natl Acad Sci U S A 2025; 122:e2500910122. [PMID: 40030038 PMCID: PMC11912360 DOI: 10.1073/pnas.2500910122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Affiliation(s)
- Michael Ye
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Graham H. Diering
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Carolina Institute for Developmental Disabilities, Carrboro, NC27510
| |
Collapse
|
7
|
Xie J, Dong R, Zhu J, Lin H, Wang S, Lai L. MMFuncPhos: A Multi-Modal Learning Framework for Identifying Functional Phosphorylation Sites and Their Regulatory Types. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410981. [PMID: 39804866 PMCID: PMC11884596 DOI: 10.1002/advs.202410981] [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: 09/08/2024] [Revised: 12/05/2024] [Indexed: 01/16/2025]
Abstract
Protein phosphorylation plays a crucial role in regulating a wide range of biological processes, and its dysregulation is strongly linked to various diseases. While many phosphorylation sites have been identified so far, their functionality and regulatory effects are largely unknown. Here, a deep learning model MMFuncPhos, based on a multi-modal deep learning framework, is developed to predict functional phosphorylation sites. MMFuncPhos outperforms existing functional phosphorylation site prediction approaches. EFuncType is further developed based on transfer learning to predict whether phosphorylation of a residue upregulates or downregulates enzyme activity for the first time. The functional phosphorylation sites predicted by MMFuncPhos and the regulatory types predicted by EFuncType align with experimental findings from several newly reported protein phosphorylation studies. The study contributes to the understanding of the functional regulatory mechanism of phosphorylation and provides valuable tools for precision medicine, enzyme engineering, and drug discovery. For user convenience, these two prediction models are integrated into a web server which can be accessed at http://pkumdl.cn:8000/mmfuncphos.
Collapse
Affiliation(s)
- Juan Xie
- Center for Quantitative BiologyAcademy for Advanced Interdisciplinary StudiesPeking UniversityBeijing100871China
| | - Ruihan Dong
- PTN Graduate ProgramAcademy for Advanced Interdisciplinary StudiesPeking UniversityBeijing100871China
| | - Jintao Zhu
- Center for Quantitative BiologyAcademy for Advanced Interdisciplinary StudiesPeking UniversityBeijing100871China
| | - Haoyu Lin
- Peking‐Tsinghua Center for Life Sciences at BNLMSCollege of Chemistry and Molecular EngineeringPeking UniversityBeijing100871China
| | - Shiwei Wang
- Peking University Chengdu Academy for Advanced Interdisciplinary BiotechnologiesChengduSichuan610213China
| | - Luhua Lai
- Center for Quantitative BiologyAcademy for Advanced Interdisciplinary StudiesPeking UniversityBeijing100871China
- Peking‐Tsinghua Center for Life Sciences at BNLMSCollege of Chemistry and Molecular EngineeringPeking UniversityBeijing100871China
- Peking University Chengdu Academy for Advanced Interdisciplinary BiotechnologiesChengduSichuan610213China
- Research Unit of Drug Design MethodChinese Academy of Medical Sciences (2021RU014)Beijing100871China
| |
Collapse
|
8
|
Sato TR, Ode KL, Kinoshita FL, Ueda HR. A design principle for neuronal firing with up-down oscillation through Na + dynamics. iScience 2025; 28:111904. [PMID: 40028276 PMCID: PMC11869597 DOI: 10.1016/j.isci.2025.111904] [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: 08/05/2024] [Revised: 12/10/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
Nonrapid eye movement sleep is characterized by high-amplitude and low-frequency electroencephalography signals. These signals are thought to be produced by the synchronized activity of cortical neurons, demonstrating the alternating bursting (up) and resting (down) states. Here, such an activity is referred to as up-down oscillation (UDO). Previously, we discussed the importance of the Ca2+-dependent hyperpolarization pathway in the generation of UDO by simulating neuronal activity based on the Hodgkin-Huxley-type model. We herein focus on intracellular Na+ dynamics. The Na+-centered model indicates that the activation of voltage-gated Na+ channels leads to intracellular Na+ accumulation, which in turn activates Na+-dependent K+ (KNa) channels or Na+/K+ ATPases, resulting in the down state. Activation kinetics of voltage-gated Na+ channels are important in shaping the UDO firing. Therefore, our model demonstrates that voltage-gated Na+ and KNa channels or Na+/K+ ATPases are candidate pathways for UDO induction.
Collapse
Affiliation(s)
- Tomohide R. Sato
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Koji L. Ode
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Fukuaki L. Kinoshita
- Department of Neurology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hiroki R. Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
- Department of Systems Biology, Institute of Life Science, Kurume University, 67 Asahimachi, Kurume, Fukuoka 830-0011, Japan
| |
Collapse
|
9
|
Nakata S, Fujiyama T, Asano F, Komiya H, Hotta-Hirashima N, Juichi M, Komine D, Kakizaki M, Ikkyu A, Mizuno S, Takahashi S, Miyoshi C, Funato H, Yanagisawa M. Partial activation of salt-inducible kinase 3 delays the onset of wakefulness and alleviates hypersomnia due to the lack of protein kinase A-phosphorylation site. Sleep 2025; 48:zsae279. [PMID: 39656684 PMCID: PMC11807893 DOI: 10.1093/sleep/zsae279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 10/23/2024] [Indexed: 12/17/2024] Open
Abstract
STUDY OBJECTIVES Sleep/wakefulness is regulated by intracellular signaling pathways composed of protein kinases such as salt-inducible kinase 3 (Sik3). Sik3-deficiency in neurons decreases nonrapid eye movement (NREM) sleep time and electroencephalogram (EEG) delta power during NREM sleep, while Sik3Slp mice lacking a protein kinase A (PKA)-phosphorylation site, S551, show hypersomnia phenotype. In this study, we examined how a phosphomimetic mutation of the 221st threonine residue (T221E), which provides a partial (weak) constitutive activity of the kinase, affects sleep/wakefulness and circadian behavior. We also examined the effect of T221E substitution on the hypersomnia phenotype of Sik3Slp mice. METHODS We examined the sleep/wake behavior of heterozygous and homozygous Sik3T221E mice and Sik3T221E;Slp mice using EEG and electromyogram recording. We also examined the circadian behavior of Sik3T221E mice using a running wheel under the light-dark cycle and constant darkness. RESULTS Heterozygous and homozygous Sik3T221E mice showed normal sleep time and sleep homeostatic responses. Homozygous Sik3T221E mice exhibited a delayed onset of wakefulness at the early dark phase and longer circadian periods. Sik3T221E;Slp mice showed decreased NREM sleep time and homeostatic responses compared to Sik3Slp mice. CONCLUSIONS Our results suggest that the peak onset of wakefulness is sensitive to disturbed kinase activity of SIK3, and the relationship between phosphorylation at T221 and S551 is critical for regulating sleep need.
Collapse
Affiliation(s)
- Shinya Nakata
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Tomoyuki Fujiyama
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Fuyuki Asano
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Haruna Komiya
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Noriko Hotta-Hirashima
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Motoki Juichi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Daiki Komine
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Miyo Kakizaki
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Aya Ikkyu
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center and Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center and Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Chika Miyoshi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiromasa Funato
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Anatomy, Graduate School of Medicine, Toho University, Tokyo, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
10
|
Yin X, Zhang Z, Zhou R, Zuo P, Sang D, Zhou S, Shi B, Chen L, Wu C, Guo Y, Wang F, Zhang EE, Li Q, Yanagisawa M, Liu Q. Calcineurin governs baseline and homeostatic regulations of non-rapid eye movement sleep in mice. Proc Natl Acad Sci U S A 2025; 122:e2418317122. [PMID: 39847332 PMCID: PMC11789068 DOI: 10.1073/pnas.2418317122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Sleep need accumulates during waking and dissipates during sleep to maintain sleep homeostasis (process S). Besides the regulation of daily (baseline) sleep amount, homeostatic sleep regulation commonly refers to the universal phenomenon that sleep deprivation (SD) causes an increase of sleep need, hence, the amount and intensity of subsequent recovery sleep. The central regulators and signaling pathways that govern the baseline and homeostatic sleep regulations in mammals remain unclear. Here, we report that enhanced activity of calcineurin Aα (CNAα)-a catalytic subunit of calcineurin-in the mouse brain neurons sharply increases the amount (to ~17-h/d) and delta power-a measure of intensity-of non-rapid eye movement sleep (NREMS). Knockout of the regulatory (CnB1) or catalytic (CnAα and CnAβ) subunits of calcineurin diminishes the amount (to ~4-h/d) and delta power of baseline NREMS, but also nearly abrogates the homeostatic recovery NREMS following SD. Accordingly, mathematical modeling of process S reveals an inability to accumulate sleep need during spontaneous or forced wakefulness in calcineurin deficient mice. Moreover, calcineurin promotes baseline NREMS by antagonizing wake-promoting protein kinase A and, in part, by activating sleep-promoting kinase SIK3. Together, these results indicate that calcineurin is an important regulator of sleep need and governs both baseline and homeostatic regulations of NREMS in mice.
Collapse
Affiliation(s)
- Xin Yin
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Peking University, Beijing100871, China
- National Institute of Biological Sciences, Beijing102206, China
| | - Zihan Zhang
- National Institute of Biological Sciences, Beijing102206, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing100730, China
| | - Rui Zhou
- National Institute of Biological Sciences, Beijing102206, China
| | - Peng Zuo
- National Institute of Biological Sciences, Beijing102206, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Di Sang
- National Institute of Biological Sciences, Beijing102206, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing100730, China
| | - Shuang Zhou
- National Institute of Biological Sciences, Beijing102206, China
- College of Life Sciences, Beijing Normal University, Beijing100875, China
| | - Bihan Shi
- National Institute of Biological Sciences, Beijing102206, China
| | - Lin Chen
- National Institute of Biological Sciences, Beijing102206, China
| | - Chongyang Wu
- National Institute of Biological Sciences, Beijing102206, China
| | - Ying Guo
- National Institute of Biological Sciences, Beijing102206, China
| | - Fengchao Wang
- National Institute of Biological Sciences, Beijing102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| | - Eric Erquan Zhang
- National Institute of Biological Sciences, Beijing102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| | - Qi Li
- National Institute of Biological Sciences, Beijing102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Qinghua Liu
- National Institute of Biological Sciences, Beijing102206, China
- New Cornerstone Science Laboratory, Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| |
Collapse
|
11
|
Yu J, Liu H, Gao R, Wang TV, Li C, Liu Y, Yang L, Xu Y, Cui Y, Jia C, Huang J, Chen PR, Rao Y. Calcineurin: An essential regulator of sleep revealed by biochemical, chemical biological, and genetic approaches. Cell Chem Biol 2025; 32:157-173.e7. [PMID: 39740665 DOI: 10.1016/j.chembiol.2024.12.003] [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: 07/09/2024] [Revised: 10/29/2024] [Accepted: 12/09/2024] [Indexed: 01/02/2025]
Abstract
Research into mechanisms underlying sleep traditionally relies on electrophysiology and genetics. Because sleep can only be measured on whole animals by behavioral observations and physical means, no sleep research was initiated by biochemical and chemical biological approaches. We used phosphorylation sites of kinases important for sleep as targets for biochemical and chemical biological approaches. Sleep was increased in mice carrying a threonine-to-alanine substitution at residue T469 of salt-inducible kinase 3 (SIK3). Our biochemical purification and photo-crosslinking revealed calcineurin (CaN) dephosphorylation, both in vitro and in vivo, of SIK3 at T469 and S551, but not T221. Knocking down CaN regulatory subunit reduced daily sleep by more than 5 h, exceeding all known mouse mutants. Our work uncovered a critical physiological role for CaN in sleep and pioneered biochemical purification and chemical biology as effective approaches to study sleep.
Collapse
Affiliation(s)
- Jianjun Yu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Huijie Liu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Rui Gao
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Tao V Wang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Chenggang Li
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Yuxiang Liu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Lu Yang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Ying Xu
- National Center for Protein Sciences Phoenix, Beijing, China
| | - Yunfeng Cui
- Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Chenxi Jia
- National Center for Protein Sciences Phoenix, Beijing, China
| | - Juan Huang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Peng R Chen
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Yi Rao
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China.
| |
Collapse
|
12
|
Zhang H, Sun H, Li J, Lv Z, Tian Y, Lei X. Gene expression is associated with brain function of insomnia disorder, rather than brain structure. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111209. [PMID: 39617164 DOI: 10.1016/j.pnpbp.2024.111209] [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: 06/23/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Previous research has found brain structural and functional abnormalities in patients with insomnia disorder (ID). However, the relationship between brain abnormalities in ID and brain gene expression is unclear. This study explored the relationship between gene expression and brain structural or functional abnormalities in ID, and we validated the reliability of the results with two independent datasets (discover dataset: healthy control (HC) = 129, ID = 264; validation dataset: HC = 160, ID = 115). Brain imaging results show that ID has abnormal resting-state spontaneous activity, regional homogeneity, and widespread gray matter volume reduction compared to HC. The association analysis results with gene expression further revealed that brain function abnormalities in ID were significantly associated with gene expression, but structural abnormalities were not. This study establishes a link between transcriptional changes and brain functional abnormalities in ID, revealing a genetic basis that may involve several biological pathways. Specifically, these pathways include hormonal regulation of the hypothalamic-pituitary-adrenal (HPA) axis, which plays a crucial role in stress response and sleep regulation; ion transport across membranes, vital for neuronal communication; and inhibitory neuronal regulation, essential for maintaining normal brain function. Furthermore, the ID-related genes are enriched for brain tissue and cortical cells, emphasizing their relevance in understanding the biological underpinnings of ID.
Collapse
Affiliation(s)
- Haobo Zhang
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing 400715, China
| | - Haonan Sun
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing 400715, China
| | - Jiatao Li
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing 400715, China
| | - Zhangwei Lv
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing 400715, China
| | - Yun Tian
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing 400715, China
| | - Xu Lei
- Sleep and NeuroImaging Center, Faculty of Psychology, Southwest University, Chongqing 400715, China; Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, Chongqing 400715, China.
| |
Collapse
|
13
|
Yang W, Shi J, Li C, Yang J, Yu J, Huang J, Rao Y. Calcium/calmodulin-dependent protein kinase II α and β differentially regulate mammalian sleep. Commun Biol 2025; 8:11. [PMID: 39757286 DOI: 10.1038/s42003-024-07449-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/30/2024] [Indexed: 01/07/2025] Open
Abstract
While sleep is important, our understanding of its molecular mechanisms is limited. Over the last two decades, protein kinases including Ca2+/calmodulin-dependent protein kinase II (CaMKII) α and β have been implicated in sleep regulation. Of all the known mouse genetic mutants, the biggest changes in sleep is reported to be observed in adult mice with sgRNAs for Camk2b injected into their embryos: sleep is reduced by approximately 120 min (mins) over 24 h (hrs). We have reexamined the sleep phenotype in mice with either Camk2a or Camk2b gene knocked-out by conventional gene targetting. While the basal sleep is reduced in Camk2a knockout mice, it remains unaltered in Camk2b mutants. Knockout of either Camk2a or Camk2b reduces sleep rebound after deprivation, indicating their roles in sleep homeostasis. These results indicate the involvement of CaMKIIα in both basal sleep and sleep homeostasis while CaMKIIβ is mainly required physiologically for sleep homeostasis, serving as a stimulus for rigorous studies in the future.
Collapse
Affiliation(s)
- Weiwen Yang
- Chinese Institute of Brain Research, Beijing (CIBR), and Chinese Institutes for Medical Research, Beijing (CIMR), Capital Medical University, Beijing, China
- Laboratory of Neurochemical Biology, Peking-Tsinghua-NIBS (PTN) Graduate Program, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences at the Health Sciences Center, Peking University, Beijing, China
| | - Jingyi Shi
- Laboratory of Neurochemical Biology, Peking-Tsinghua-NIBS (PTN) Graduate Program, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences at the Health Sciences Center, Peking University, Beijing, China
| | - Chenggang Li
- Chinese Institute of Brain Research, Beijing (CIBR), and Chinese Institutes for Medical Research, Beijing (CIMR), Capital Medical University, Beijing, China
- Laboratory of Neurochemical Biology, Peking-Tsinghua-NIBS (PTN) Graduate Program, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences at the Health Sciences Center, Peking University, Beijing, China
| | - Jingqun Yang
- Chinese Institute of Brain Research, Beijing (CIBR), and Chinese Institutes for Medical Research, Beijing (CIMR), Capital Medical University, Beijing, China
| | - Jianjun Yu
- Chinese Institute of Brain Research, Beijing (CIBR), and Chinese Institutes for Medical Research, Beijing (CIMR), Capital Medical University, Beijing, China
- Laboratory of Neurochemical Biology, Peking-Tsinghua-NIBS (PTN) Graduate Program, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences at the Health Sciences Center, Peking University, Beijing, China
| | - Juan Huang
- Chinese Institute of Brain Research, Beijing (CIBR), and Chinese Institutes for Medical Research, Beijing (CIMR), Capital Medical University, Beijing, China
- Laboratory of Neurochemical Biology, Peking-Tsinghua-NIBS (PTN) Graduate Program, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences at the Health Sciences Center, Peking University, Beijing, China
| | - Yi Rao
- Chinese Institute of Brain Research, Beijing (CIBR), and Chinese Institutes for Medical Research, Beijing (CIMR), Capital Medical University, Beijing, China.
- Laboratory of Neurochemical Biology, Peking-Tsinghua-NIBS (PTN) Graduate Program, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences at the Health Sciences Center, Peking University, Beijing, China.
| |
Collapse
|
14
|
Zheng Y, Yu X, Wei L, Chen Q, Xu Y, Ni P, Deng W, Guo W, Hu X, Qi X, Li T. LT-102, an AMPA receptor potentiator, alleviates depression-like behavior and synaptic plasticity impairments in prefrontal cortex induced by sleep deprivation. J Affect Disord 2024; 367:18-30. [PMID: 39214374 DOI: 10.1016/j.jad.2024.08.176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Sleep loss is closely related to the onset and development of depression, and the mechanisms involved may include impaired synaptic plasticity. Considering the important role of glutamate α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptors (AMPARs) in synaptic plasticity as well as depression, we introduce LT-102, a novel AMPARs potentiator, to evaluate the potential of LT-102 in treating sleep deprivation-induced depression-like behaviors. METHODS We conducted a comprehensive behavioral assessment to evaluate the effects of LT-102 on depression-like symptoms in male C57BL/6J mice. This assessment included the open field test to measure general locomotor activity and anxiety-like behavior, the forced swimming test and tail suspension test to assess despair behaviors indicative of depressive states, and the sucrose preference test to quantify anhedonia, a core symptom of depression. Furthermore, to explore the impact of LT-102 on synaptic plasticity, we utilized a combination of Western blot analysis to detect protein expression levels, Golgi-Cox staining to visualize neuronal morphology, and immunofluorescence to examine the localization of synaptic proteins. Additionally, we utilized primary cortical neurons to delineate the signaling pathway modulated by LT-102. RESULTS Treatment with LT-102 significantly reduced depression-like behaviors associated with sleep deprivation. Quantitative Western blot (WB) analysis revealed a significant increase in GluA1 phosphorylation in the prefrontal cortex (PFC), triggering the Ca2+/calmodulin-dependent protein kinase II/cAMP response element-binding protein/brain-derived neurotrophic factor (CaMKII/CREB/BDNF) and forkhead box protein P2/postsynaptic density protein 95 (FoxP2/PSD95) signaling pathways. Immunofluorescence imaging confirmed that LT-102 treatment increased spine density and co-labeling of PSD95 and vesicular glutamate transporter 1 (VGLUT1) in the PFC, reversing the reductions typically observed following sleep deprivation. Golgi staining further validated these results, showing a substantial increase in neuronal dendritic spine density in sleep-deprived mice treated with LT-102. Mechanistically, application of LT-102 to primary cortical neurons, resulted in elevated levels of phosphorylated AKT (p-AKT) and phosphorylated glycogen synthase kinase-3 beta (p-GSK3β), key downstream molecules in the BDNF signaling pathway, which in turn upregulated FoxP2 and PSD95 expression. LIMITATIONS In our study, we chose to exclusively use male mice to eliminate potential influences of the estrous cycle on behavior and physiology. As there is no widely accepted positive drug control for sleep deprivation studies, we did not include one in our research. CONCLUSION Our results suggest that LT-102 is a promising therapeutic agent for counteracting depression-like behaviors and synaptic plasticity deficits induced by sleep deprivation, primarily through the activation of CaMKII/CREB/BDNF and AKT/GSK3β/FoxP2/PSD95 signaling pathways.
Collapse
Affiliation(s)
- Yanghao Zheng
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China
| | - Xueli Yu
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Long Wei
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qiyuan Chen
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yan Xu
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Peiyan Ni
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Wei Deng
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Wanjun Guo
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Xun Hu
- The Clinical Research Center and Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xueyu Qi
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China.
| | - Tao Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
15
|
Zhang Y, Wu J, Zheng Y, Xu Y, Yu Z, Ping Y. Voltage Gated Ion Channels and Sleep. J Membr Biol 2024; 257:269-280. [PMID: 39354150 DOI: 10.1007/s00232-024-00325-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Ion channels are integral components of the nervous system, playing a pivotal role in shaping membrane potential, neuronal excitability, synaptic transmission and plasticity. Dysfunction in these channels, such as improper expression or localization, can lead to irregular neuronal excitability and synaptic communication, which may manifest as various behavioral abnormalities, including disrupted rest-activity cycles. Research has highlighted the significant impact of voltage gated ion channels on sleep parameters, influencing sleep latency, duration and waveforms. Furthermore, these ion channels have been implicated in the vulnerability to, and the pathogenesis of, several neurological and psychiatric disorders, including epilepsy, autism, schizophrenia, and Alzheimer's disease (AD). In this comprehensive review, we aim to provide a summary of the regulatory role of three predominant types of voltage-gated ion channels-calcium (Ca2+), sodium (Na+), and potassium (K+)-in sleep across species, from flies to mammals. We will also discuss the association of sleep disorders with various human diseases that may arise from the dysfunction of these ion channels, thereby underscoring the potential therapeutic benefits of targeting specific ion channel subtypes for sleep disturbance treatment.
Collapse
Affiliation(s)
- Yan Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jiawen Wu
- Faculty of Brain Sciences, University College London, London, UK
| | - Yuxian Zheng
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yangkun Xu
- Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Ziqi Yu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yong Ping
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
16
|
Wang Y, Cao S, Tone D, Fujishima H, Yamada RG, Ohno RI, Shi S, Matsuzawa K, Yada S, Kaneko M, Sakamoto H, Onishi T, Ukai-Tadenuma M, Ukai H, Hanashima C, Hirose K, Kiyonari H, Sumiyama K, Ode KL, Ueda HR. Postsynaptic competition between calcineurin and PKA regulates mammalian sleep-wake cycles. Nature 2024; 636:412-421. [PMID: 39506111 DOI: 10.1038/s41586-024-08132-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 09/27/2024] [Indexed: 11/08/2024]
Abstract
The phosphorylation of synaptic proteins is a significant biochemical reaction that controls the sleep-wake cycle in mammals1-3. Protein phosphorylation in vivo is reversibly regulated by kinases and phosphatases. In this study, we investigate a pair of kinases and phosphatases that reciprocally regulate sleep duration. First, we perform a comprehensive screen of protein kinase A (PKA) and phosphoprotein phosphatase (PPP) family genes by generating 40 gene knockout mouse lines using prenatal and postnatal CRISPR targeting. We identify a regulatory subunit of PKA (Prkar2b), a regulatory subunit of protein phosphatase 1 (PP1; Pppr1r9b) and catalytic and regulatory subunits of calcineurin (also known as PP2B) (Ppp3ca and Ppp3r1) as sleep control genes. Using adeno-associated virus (AAV)-mediated stimulation of PKA and PP1-calcineurin activities, we show that PKA is a wake-promoting kinase, whereas PP1 and calcineurin function as sleep-promoting phosphatases. The importance of these phosphatases in sleep regulation is supported by the marked changes in sleep duration associated with their increased and decreased activities, ranging from approximately 17.3 h per day (PP1 expression) to 4.3 h per day (postnatal CRISPR targeting of calcineurin). Localization signals to the excitatory post-synapse are necessary for these phosphatases to exert their sleep-promoting effects. Furthermore, the wake-promoting effect of PKA localized to the excitatory post-synapse negated the sleep-promoting effect of PP1-calcineurin. These findings indicate that PKA and PP1-calcineurin have competing functions in sleep regulation at excitatory post-synapses.
Collapse
Affiliation(s)
- Yimeng Wang
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Siyu Cao
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Daisuke Tone
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Hiroshi Fujishima
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Department of Systems Biology, Institute of Life Science, Kurume University, Kurume, Fukuoka, Japan
| | - Rikuhiro G Yamada
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Department of Systems Biology, Institute of Life Science, Kurume University, Kurume, Fukuoka, Japan
| | - Rei-Ichiro Ohno
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shoi Shi
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kyoko Matsuzawa
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Saori Yada
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Mari Kaneko
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Hirokazu Sakamoto
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Taichi Onishi
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Maki Ukai-Tadenuma
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hideki Ukai
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Carina Hanashima
- Department of Biology, Faculty of Education and Integrated Arts and Sciences, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Kenzo Hirose
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Kenta Sumiyama
- Laboratory for Mouse Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Department of Animal Sciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, Japan
| | - Koji L Ode
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Hiroki R Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan.
- Department of Systems Biology, Institute of Life Science, Kurume University, Kurume, Fukuoka, Japan.
| |
Collapse
|
17
|
Yamada RG, Matsuzawa K, Ode KL, Ueda HR. An automated sleep staging tool based on simple statistical features of mice electroencephalography (EEG) and electromyography (EMG) data. Eur J Neurosci 2024; 60:5467-5486. [PMID: 39072800 DOI: 10.1111/ejn.16465] [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: 03/29/2024] [Revised: 06/04/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024]
Abstract
Electroencephalogram (EEG) and electromyogram (EMG) are fundamental tools in sleep research. However, investigations into the statistical properties of rodent EEG/EMG signals in the sleep-wake cycle have been limited. The lack of standard criteria in defining sleep stages forces researchers to rely on human expertise to inspect EEG/EMG. The recent increasing demand for analysing large-scale and long-term data has been overwhelming the capabilities of human experts. In this study, we explored the statistical features of EEG signals in the sleep-wake cycle. We found that the normalized EEG power density profile changes its lower and higher frequency powers to a comparable degree in the opposite direction, pivoting around 20-30 Hz between the NREM sleep and the active brain state. We also found that REM sleep has a normalized EEG power density profile that overlaps with wakefulness and a characteristic reduction in the EMG signal. Based on these observations, we proposed three simple statistical features that could span a 3D space. Each sleep-wake stage formed a separate cluster close to a normal distribution in the 3D space. Notably, the suggested features are a natural extension of the conventional definition, making it useful for experts to intuitively interpret the EEG/EMG signal alterations caused by genetic mutations or experimental treatments. In addition, we developed an unsupervised automatic staging algorithm based on these features. The developed algorithm is a valuable tool for expediting the quantitative evaluation of EEG/EMG signals so that researchers can utilize the recent high-throughput genetic or pharmacological methods for sleep research.
Collapse
Affiliation(s)
- Rikuhiro G Yamada
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
- Department of Systems Biology, Institute of Life Science, Kurume University, Fukuoka, Japan
| | - Kyoko Matsuzawa
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
| | - Koji L Ode
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroki R Ueda
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
- Department of Systems Biology, Institute of Life Science, Kurume University, Fukuoka, Japan
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
18
|
Nakata S, Iwasaki K, Funato H, Yanagisawa M, Ozaki H. Neuronal subtype-specific transcriptomic changes in the cerebral neocortex associated with sleep pressure. Neurosci Res 2024; 207:13-25. [PMID: 38537682 DOI: 10.1016/j.neures.2024.03.004] [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: 02/20/2024] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/07/2024]
Abstract
Sleep is homeostatically regulated by sleep pressure, which increases during wakefulness and dissipates during sleep. Recent studies have suggested that the cerebral neocortex, a six-layered structure composed of various layer- and projection-specific neuronal subtypes, is involved in the representation of sleep pressure governed by transcriptional regulation. Here, we examined the transcriptomic changes in neuronal subtypes in the neocortex upon increased sleep pressure using single-nucleus RNA sequencing datasets and predicted the putative intracellular and intercellular molecules involved in transcriptome alterations. We revealed that sleep deprivation (SD) had the greatest effect on the transcriptome of layer 2 and 3 intratelencephalic (L2/3 IT) neurons among the neocortical glutamatergic neuronal subtypes. The expression of mutant SIK3 (SLP), which is known to increase sleep pressure, also induced profound changes in the transcriptome of L2/3 IT neurons. We identified Junb as a candidate transcription factor involved in the alteration of the L2/3 IT neuronal transcriptome by SD and SIK3 (SLP) expression. Finally, we inferred putative intercellular ligands, including BDNF, LSAMP, and PRNP, which may be involved in SD-induced alteration of the transcriptome of L2/3 IT neurons. We suggest that the transcriptome of L2/3 IT neurons is most impacted by increased sleep pressure among neocortical glutamatergic neuronal subtypes and identify putative molecules involved in such transcriptional alterations.
Collapse
Affiliation(s)
- Shinya Nakata
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kanako Iwasaki
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiromasa Funato
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan; Department of Anatomy, Graduate School of Medicine, Toho University, Tokyo, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan; Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | - Haruka Ozaki
- Bioinformatics Laboratory, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan; Center for Artificial Intelligence Research, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
19
|
Sawada T, Iino Y, Yoshida K, Okazaki H, Nomura S, Shimizu C, Arima T, Juichi M, Zhou S, Kurabayashi N, Sakurai T, Yagishita S, Yanagisawa M, Toyoizumi T, Kasai H, Shi S. Prefrontal synaptic regulation of homeostatic sleep pressure revealed through synaptic chemogenetics. Science 2024; 385:1459-1465. [PMID: 39325885 DOI: 10.1126/science.adl3043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 06/28/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024]
Abstract
Sleep is regulated by homeostatic processes, yet the biological basis of sleep pressure that accumulates during wakefulness, triggers sleep, and dissipates during sleep remains elusive. We explored a causal relationship between cellular synaptic strength and electroencephalography delta power indicating macro-level sleep pressure by developing a theoretical framework and a molecular tool to manipulate synaptic strength. The mathematical model predicted that increased synaptic strength promotes the neuronal "down state" and raises the delta power. Our molecular tool (synapse-targeted chemically induced translocation of Kalirin-7, SYNCit-K), which induces dendritic spine enlargement and synaptic potentiation through chemically induced translocation of protein Kalirin-7, demonstrated that synaptic potentiation of excitatory neurons in the prefrontal cortex (PFC) increases nonrapid eye movement sleep amounts and delta power. Thus, synaptic strength of PFC excitatory neurons dictates sleep pressure in mammals.
Collapse
Affiliation(s)
- Takeshi Sawada
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yusuke Iino
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kensuke Yoshida
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Hitoshi Okazaki
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shinnosuke Nomura
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Chika Shimizu
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Tomoki Arima
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Motoki Juichi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Siqi Zhou
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | | | - Takeshi Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Molecular Behavioral Physiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Sho Yagishita
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Taro Toyoizumi
- RIKEN Center for Brain Science, Wako, Saitama, Japan
- Department of Mathematical Informatics, Graduate School of Information Science and Technology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Haruo Kasai
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shoi Shi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
20
|
ElGrawani W, Sun G, Kliem FP, Sennhauser S, Pierre-Ferrer S, Rosi-Andersen A, Boccalaro I, Bethge P, Heo WD, Helmchen F, Adamantidis AR, Forger DB, Robles MS, Brown SA. BDNF-TrkB signaling orchestrates the buildup process of local sleep. Cell Rep 2024; 43:114500. [PMID: 39046880 DOI: 10.1016/j.celrep.2024.114500] [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: 12/11/2023] [Revised: 05/15/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
Sleep debt accumulates during wakefulness, leading to increased slow wave activity (SWA) during sleep, an encephalographic marker for sleep need. The use-dependent demands of prior wakefulness increase sleep SWA locally. However, the circuitry and molecular identity of this "local sleep" remain unclear. Using pharmacology and optogenetic perturbations together with transcriptomics, we find that cortical brain-derived neurotrophic factor (BDNF) regulates SWA via the activation of tyrosine kinase B (TrkB) receptor and cAMP-response element-binding protein (CREB). We map BDNF/TrkB-induced sleep SWA to layer 5 (L5) pyramidal neurons of the cortex, independent of neuronal firing per se. Using mathematical modeling, we here propose a model of how BDNF's effects on synaptic strength can increase SWA in ways not achieved through increased firing alone. Proteomic analysis further reveals that TrkB activation enriches ubiquitin and proteasome subunits. Together, our study reveals that local SWA control is mediated by BDNF-TrkB-CREB signaling in L5 excitatory cortical neurons.
Collapse
Affiliation(s)
- Waleed ElGrawani
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland.
| | - Guanhua Sun
- Department of Mathematics, University of Michigan, Ann Arbor, MI, USA
| | - Fabian P Kliem
- Institute of Medical Psychology and Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Germany
| | - Simon Sennhauser
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Sara Pierre-Ferrer
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
| | - Alex Rosi-Andersen
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
| | - Ida Boccalaro
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, Bern, Switzerland
| | - Philipp Bethge
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland; Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Won Do Heo
- Department of Biological Science, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Fritjof Helmchen
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland; Brain Research Institute, University of Zurich, Zurich, Switzerland; University Research Priority Program (URPP), Adaptive Brain Circuits in Development and Learning, University of Zurich, Zurich, Switzerland
| | - Antoine R Adamantidis
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, Bern, Switzerland.
| | - Daniel B Forger
- Department of Mathematics, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| | - Maria S Robles
- Institute of Medical Psychology and Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Germany.
| | - Steven A Brown
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
21
|
Kon K, Ode KL, Mano T, Fujishima H, Takahashi RR, Tone D, Shimizu C, Shiono S, Yada S, Matsuzawa K, Yoshida SY, Yoshida Garçon J, Kaneko M, Shinohara Y, Yamada RG, Shi S, Miyamichi K, Sumiyama K, Kiyonari H, Susaki EA, Ueda HR. Cortical parvalbumin neurons are responsible for homeostatic sleep rebound through CaMKII activation. Nat Commun 2024; 15:6054. [PMID: 39025867 PMCID: PMC11258272 DOI: 10.1038/s41467-024-50168-5] [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: 03/27/2023] [Accepted: 07/01/2024] [Indexed: 07/20/2024] Open
Abstract
The homeostatic regulation of sleep is characterized by rebound sleep after prolonged wakefulness, but the molecular and cellular mechanisms underlying this regulation are still unknown. In this study, we show that Ca2+/calmodulin-dependent protein kinase II (CaMKII)-dependent activity control of parvalbumin (PV)-expressing cortical neurons is involved in homeostatic regulation of sleep in male mice. Prolonged wakefulness enhances cortical PV-neuron activity. Chemogenetic suppression or activation of cortical PV neurons inhibits or induces rebound sleep, implying that rebound sleep is dependent on increased activity of cortical PV neurons. Furthermore, we discovered that CaMKII kinase activity boosts the activity of cortical PV neurons, and that kinase activity is important for homeostatic sleep rebound. Here, we propose that CaMKII-dependent PV-neuron activity represents negative feedback inhibition of cortical neural excitability, which serves as the distributive cortical circuits for sleep homeostatic regulation.
Collapse
Affiliation(s)
- Kazuhiro Kon
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Kennedy Krieger Institute, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Koji L Ode
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
| | - Tomoyuki Mano
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- Department of Information Physics and Computing, Graduate School of Information Science and Technology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Computational Neuroethology Unit, Okinawa Institute of Science and Technology, Onna, Okinawa, Japan
| | - Hiroshi Fujishima
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- Department of Systems Biology, Institute of Life Science, Kurume University, Kurume, Fukuoka, Japan
| | - Riina R Takahashi
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Daisuke Tone
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
| | - Chika Shimizu
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
| | - Shinnosuke Shiono
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Saori Yada
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kyoko Matsuzawa
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
| | - Shota Y Yoshida
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
| | - Junko Yoshida Garçon
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- Department of Molecular Embryology, Research Institute, Osaka Women's and Children's Hospital, Izumi, Osaka, Japan
| | - Mari Kaneko
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research (BDR), Chuou-ku, Kobe, Hyogo, Japan
| | - Yuta Shinohara
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Rikuhiro G Yamada
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- Department of Systems Biology, Institute of Life Science, Kurume University, Kurume, Fukuoka, Japan
| | - Shoi Shi
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazunari Miyamichi
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research (BDR), Chuou-ku, Kobe, Hyogo, Japan
| | - Kenta Sumiyama
- Laboratory for Mouse Genetic Engineering, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- Laboratory of Animal Genetics and Breeding, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa-ku, Nagoya, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research (BDR), Chuou-ku, Kobe, Hyogo, Japan
| | - Etsuo A Susaki
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Hiroki R Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan.
- Department of Information Physics and Computing, Graduate School of Information Science and Technology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
- Department of Systems Biology, Institute of Life Science, Kurume University, Kurume, Fukuoka, Japan.
| |
Collapse
|
22
|
Yoshikawa T, Honma KI, Shigeyoshi Y, Yamagata Y, Honma S. A critical role of Ca 2+/calmodulin-dependent protein kinase II in coupling between evening and morning circadian oscillators in the suprachiasmatic nucleus. Eur J Neurosci 2024; 60:3828-3842. [PMID: 38571281 DOI: 10.1111/ejn.16316] [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: 10/21/2022] [Revised: 02/16/2024] [Accepted: 02/28/2024] [Indexed: 04/05/2024]
Abstract
Ca2+/calmodulin-dependent protein kinase IIα (CaMKIIα) is widely expressed in the brain and is involved in various functions, including memory formation, mood and sleep. We previously reported that CaMKIIα is involved in the circadian molecular clock. Mice lacking functional CaMKIIα (K42R mice) exhibited a gradual increase in activity time (α decompression) of running-wheel (RW) activity due to a lengthened circadian period (τ) of activity offset under constant darkness (DD). In the present study, to investigate the functional roles of CaMKIIα in behavioural rhythms, we measured RW and general movements simultaneously under prolonged DD. Tau became longer as the relative intensity of behaviour activity within an activity time shifted from activity onset towards activity offset. In some K42R mice, α was gradually expanded with a marked reduction of RW activity, while general movements persisted without noticeable decline, which was followed by an abrupt shortening of α (α compression) with differential phase shifts of the activity onset and offset and recovery of RW activity. These results suggest that an internal coupling between the oscillators controlling activity onset and offset is bidirectional but with different strengths. The α compression occurred recurrently in 38% of K42R mice examined with an average interval of 37 days in association with attenuation of RW activity but never in the wild-type (WT) mice. Consistent with behavioural rhythms, the circadian period of the PER2::LUC rhythm in the cultured suprachiasmatic nucleus (SCN) slice was significantly longer in K42R than in WT. These findings are best interpreted by assuming that a loss of functional CaMKIIα attenuates the coupling between the onset and offset oscillators.
Collapse
Affiliation(s)
- Tomoko Yoshikawa
- Photonic Bioimaging Section, Research Center for Cooperative Project, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Department of Chronomedicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Department of Anatomy and Neurobiology, Kindai University Faculty of Medicine, Osakasayama, Japan
- Organization for International Education and Exchange, University of Toyama, Toyama, Japan
| | - Ken-Ichi Honma
- Department of Chronomedicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Research and Education Center for Brain Science, Hokkaido University, Sapporo, Japan
| | - Yasufumi Shigeyoshi
- Department of Anatomy and Neurobiology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Yoko Yamagata
- Section of Multilayer Physiology, National Institute for Physiological Sciences, Okazaki, Japan
- The Graduate University for Advanced Studies, Hayama, Japan
| | - Sato Honma
- Department of Chronomedicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Research and Education Center for Brain Science, Hokkaido University, Sapporo, Japan
| |
Collapse
|
23
|
Ding F, Sun Q, Long C, Rasmussen RN, Peng S, Xu Q, Kang N, Song W, Weikop P, Goldman SA, Nedergaard M. Dysregulation of extracellular potassium distinguishes healthy ageing from neurodegeneration. Brain 2024; 147:1726-1739. [PMID: 38462589 PMCID: PMC11068329 DOI: 10.1093/brain/awae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 03/12/2024] Open
Abstract
Progressive neuronal loss is a hallmark feature distinguishing neurodegenerative diseases from normal ageing. However, the underlying mechanisms remain unknown. Extracellular K+ homeostasis is a potential mediator of neuronal injury as K+ elevations increase excitatory activity. The dysregulation of extracellular K+ and potassium channel expressions during neurodegeneration could contribute to this distinction. Here we measured the cortical extracellular K+ concentration ([K+]e) in awake wild-type mice as well as murine models of neurodegeneration using K+-sensitive microelectrodes. Unexpectedly, aged wild-type mice exhibited significantly lower cortical [K+]e than young mice. In contrast, cortical [K+]e was consistently elevated in Alzheimer's disease (APP/PS1), amyotrophic lateral sclerosis (ALS) (SOD1G93A) and Huntington's disease (R6/2) models. Cortical resting [K+]e correlated inversely with neuronal density and the [K+]e buffering rate but correlated positively with the predicted neuronal firing rate. Screening of astrocyte-selective genomic datasets revealed a number of potassium channel genes that were downregulated in these disease models but not in normal ageing. In particular, the inwardly rectifying potassium channel Kcnj10 was downregulated in ALS and Huntington's disease models but not in normal ageing, while Fxyd1 and Slc1a3, each of which acts as a negative regulator of potassium uptake, were each upregulated by astrocytes in both Alzheimer's disease and ALS models. Chronic elevation of [K+]e in response to changes in gene expression and the attendant neuronal hyperexcitability may drive the neuronal loss characteristic of these neurodegenerative diseases. These observations suggest that the dysregulation of extracellular K+ homeostasis in a number of neurodegenerative diseases could be due to aberrant astrocytic K+ buffering and as such, highlight a fundamental role for glial dysfunction in neurodegeneration.
Collapse
Affiliation(s)
- Fengfei Ding
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Pharmacology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qian Sun
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Pharmacology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Carter Long
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Rune Nguyen Rasmussen
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, Neurology Department, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sisi Peng
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Qiwu Xu
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ning Kang
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Wei Song
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Pia Weikop
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, Neurology Department, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, Neurology Department, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, Neurology Department, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
24
|
Andersen JV, Westi EW, Griem-Krey N, Skotte NH, Schousboe A, Aldana BI, Wellendorph P. Deletion of CaMKIIα disrupts glucose metabolism, glutamate uptake, and synaptic energetics in the cerebral cortex. J Neurochem 2024; 168:704-718. [PMID: 36949663 DOI: 10.1111/jnc.15814] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/28/2023] [Accepted: 03/20/2023] [Indexed: 03/24/2023]
Abstract
Ca2+/calmodulin-dependent protein kinase II alpha (CaMKIIα) is a key regulator of neuronal signaling and synaptic plasticity. Synaptic activity and neurotransmitter homeostasis are closely coupled to the energy metabolism of both neurons and astrocytes. However, whether CaMKIIα function is implicated in brain energy and neurotransmitter metabolism remains unclear. Here, we explored the metabolic consequences of CaMKIIα deletion in the cerebral cortex using a genetic CaMKIIα knockout (KO) mouse. Energy and neurotransmitter metabolism was functionally investigated in acutely isolated cerebral cortical slices using stable 13C isotope tracing, whereas the metabolic function of synaptosomes was assessed by the rates of glycolytic activity and mitochondrial respiration. The oxidative metabolism of [U-13C]glucose was extensively reduced in cerebral cortical slices of the CaMKIIα KO mice. In contrast, metabolism of [1,2-13C]acetate, primarily reflecting astrocyte metabolism, was unaffected. Cellular uptake, and subsequent metabolism, of [U-13C]glutamate was decreased in cerebral cortical slices of CaMKIIα KO mice, whereas uptake and metabolism of [U-13C]GABA were unaffected, suggesting selective metabolic impairments of the excitatory system. Synaptic metabolic function was maintained during resting conditions in isolated synaptosomes from CaMKIIα KO mice, but both the glycolytic and mitochondrial capacities became insufficient when the synaptosomes were metabolically challenged. Collectively, this study shows that global deletion of CaMKIIα significantly impairs cellular energy and neurotransmitter metabolism, particularly of neurons, suggesting a metabolic role of CaMKIIα signaling in the brain.
Collapse
Affiliation(s)
- Jens V Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emil W Westi
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nane Griem-Krey
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels H Skotte
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Petrine Wellendorph
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Chen P, Wang W, Ban W, Zhang K, Dai Y, Yang Z, You Y. Deciphering Post-Stroke Sleep Disorders: Unveiling Neurological Mechanisms in the Realm of Brain Science. Brain Sci 2024; 14:307. [PMID: 38671959 PMCID: PMC11047862 DOI: 10.3390/brainsci14040307] [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: 02/21/2024] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 04/28/2024] Open
Abstract
Sleep disorders are the most widespread mental disorders after stroke and hurt survivors' functional prognosis, response to restoration, and quality of life. This review will address an overview of the progress of research on the biological mechanisms associated with stroke-complicating sleep disorders. Extensive research has investigated the negative impact of stroke on sleep. However, a bidirectional association between sleep disorders and stroke exists; while stroke elevates the risk of sleep disorders, these disorders also independently contribute as a risk factor for stroke. This review aims to elucidate the mechanisms of stroke-induced sleep disorders. Possible influences were examined, including functional changes in brain regions, cerebrovascular hemodynamics, neurological deficits, sleep ion regulation, neurotransmitters, and inflammation. The results provide valuable insights into the mechanisms of stroke complicating sleep disorders.
Collapse
Affiliation(s)
- Pinqiu Chen
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, Yantai 264005, China; (P.C.)
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Wenyan Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, Yantai 264005, China; (P.C.)
| | - Weikang Ban
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Kecan Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Yanan Dai
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Zhihong Yang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Yuyang You
- School of Automation, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
26
|
Xing L, Zou X, Yin C, Webb JM, Shi G, Ptáček LJ, Fu YH. Diverse roles of pontine NPS-expressing neurons in sleep regulation. Proc Natl Acad Sci U S A 2024; 121:e2320276121. [PMID: 38381789 PMCID: PMC10907243 DOI: 10.1073/pnas.2320276121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/17/2024] [Indexed: 02/23/2024] Open
Abstract
Neuropeptide S (NPS) was postulated to be a wake-promoting neuropeptide with unknown mechanism, and a mutation in its receptor (NPSR1) causes the short sleep duration trait in humans. We investigated the role of different NPS+ nuclei in sleep/wake regulation. Loss-of-function and chemogenetic studies revealed that NPS+ neurons in the parabrachial nucleus (PB) are wake-promoting, whereas peri-locus coeruleus (peri-LC) NPS+ neurons are not important for sleep/wake modulation. Further, we found that a NPS+ nucleus in the central gray of the pons (CGPn) strongly promotes sleep. Fiber photometry recordings showed that NPS+ neurons are wake-active in the CGPn and wake/REM-sleep active in the PB and peri-LC. Blocking NPS-NPSR1 signaling or knockdown of Nps supported the function of the NPS-NPSR1 pathway in sleep/wake regulation. Together, these results reveal that NPS and NPS+ neurons play dichotomous roles in sleep/wake regulation at both the molecular and circuit levels.
Collapse
Affiliation(s)
- Lijuan Xing
- Department of Neurology, University of California San Francisco, San Francisco, CA94143
| | - Xianlin Zou
- Department of Neurology, University of California San Francisco, San Francisco, CA94143
| | - Chen Yin
- Department of Neurology, University of California San Francisco, San Francisco, CA94143
| | - John M. Webb
- Department of Neurology, University of California San Francisco, San Francisco, CA94143
| | - Guangsen Shi
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan528400, China
| | - Louis J. Ptáček
- Department of Neurology, University of California San Francisco, San Francisco, CA94143
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA94143
- Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA94143
- Institute of Human Genetics, University of California San Francisco, San Francisco, CA94143
| | - Ying-Hui Fu
- Department of Neurology, University of California San Francisco, San Francisco, CA94143
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA94143
- Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA94143
- Institute of Human Genetics, University of California San Francisco, San Francisco, CA94143
| |
Collapse
|
27
|
Ma L, Huo Y, Peng T, Liu Z, Ye J, Chen L, Wu D, Du W, Chen J. Assessing the journey of calcium supplementation: A mendelian randomization study on the causal link between calcium levels and sleep disorders. Clin Nutr ESPEN 2024; 59:1-8. [PMID: 38220361 DOI: 10.1016/j.clnesp.2023.10.039] [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: 07/18/2023] [Revised: 10/12/2023] [Accepted: 10/29/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND & AIM Sleep disorder is a growing concern, and calcium supplementation is often recommended as a potential intervention for sleep disorders. However, the causal relationship between calcium levels and the incidence of sleep disorders remains unclear. Mendelian randomization techniques utilizing genetic variants that affect calcium levels, can provide valuable insights into causality. This study aims to examine the association between calcium levels and sleep disorders in a diverse population that includes both adolescents and adults, and investigate the effects of calcium levels on sleep disorders. METHODS Mendelian randomization analysis was conducted using data from UK Biobank and FinnGen datasets. The inverse-variance weighting (IVW) was selected as the primary method. In addition, traditional mediation analysis was performed on a subset of the NHANES data spanning from 2007 to 2018. RESULTS Our findings provide evidence supporting a causal relationship between calcium intake and reduced risk of sleep disorders (beta = -0.079, SE = 0.0395, P = 0.0457). While not reaching statistical significance, other MR methods such as weighted median and Mr-Egger exhibited similar directional trends. Analysis of the NHANES cohort revealed a negative association between calcium levels and the prevalence of sleep disorders in male, black, and physically active populations. However, this association was not observed in other demographic groups. CONCLUSION Our results suggested that there is no significant correlation between calcium levels and sleep disorder in non-exercise populations. This raises concerns about the long-term high-dose calcium supplementation in clinical practice, which requires further investigation.
Collapse
Affiliation(s)
- Ling Ma
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanyan Huo
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Peng
- Department of Neonatology, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai, China
| | - Zhongling Liu
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiangfeng Ye
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Lingyan Chen
- Department of Occupational Therapy Science, Nagasaki University Graduate School of Biomedical Science, 1-7-1 Sakamoto, Nagasaki 852-8520, Japan
| | - Dan Wu
- Department of Cognitive Neuroscience, Donders Center for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Wenchong Du
- NTU Psychology, Nottingham Trent University, Burton Street, Nottingham, NG1 4BU, United Kingdom.
| | - Jinjin Chen
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Children's Hospital Dipro Medical Research Center, China.
| |
Collapse
|
28
|
Yufu L, Qiumei L, Tiantian Z, Jiansheng C, Xu T, Yanfei W, Xiaoting M, Shenxiang H, Yinxia L, You L, Tingyu L, Jian Q, Zhiyong Z. Association between multiple metals exposure and sleep disorders in a Chinese population: A mixture-based approach. CHEMOSPHERE 2023; 343:140213. [PMID: 37742758 DOI: 10.1016/j.chemosphere.2023.140213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/27/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023]
Abstract
OBJECTIVE Previous studies have suggested a possible association between metals and sleep disorders. This study aimed to explore the association between Zn, Cu, Se, Mg and Ca and sleep disorders in single and multi-metal co-exposure models. METHODS Logistic regression models, restricted cubic spline model (RCS), Quantile g computation (Q-gcomp), Weighted Quantile Sum (WQS), and Bayesian kernel machine regression (BKMR) models were used to investigate the association between metal levels and sleep disorders. RESULTS Logistic regression showed that in the total population, the second, third, and fourth quartile Zn concentration exhibited a lower risk of sleep disorders compared with the first quartile, with odds ratios (ORs) of 0.783, 0.711, and 0.704, respectively. Compared with Zn/Cu and Zn/Se in the first quartile, the third and fourth quartiles showed a lower risk of sleep disorders. In the 30-59 years group, the risk of sleep disorders was 0.699 times greater for the fourth quartile Mg concentration than that for the first quartile. The risk of sleep disorders in Mg/Ca concentration in the third quartile was 0.737 times higher than in the first quartile. Q-gcomp, WQS, and BKMR model analysis showed the negative overall effect of mixtures of the five metals on sleep disorders, with Zn being the largest contributor. CONCLUSION Our study showed that plasma Zn, Mg, Zn/Cu, Zn/Se, and Mg/Ca reduced the risk of sleep disorders, and the combined effect of multiple metals was negatively associated with the risk of sleep disorders, with Zn being the largest contributor to this relationship.
Collapse
Affiliation(s)
- Lu Yufu
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning, 530021, Guangxi, China
| | - Liu Qiumei
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning, 530021, Guangxi, China
| | - Zhang Tiantian
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning, 530021, Guangxi, China
| | - Cai Jiansheng
- School of Public Health, Guilin Medical University, 20 Lequn Road, Guilin, Guangxi, China
| | - Tang Xu
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning, 530021, Guangxi, China
| | - Wei Yanfei
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning, 530021, Guangxi, China
| | - Mo Xiaoting
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning, 530021, Guangxi, China
| | - Huang Shenxiang
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning, 530021, Guangxi, China
| | - Lin Yinxia
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning, 530021, Guangxi, China
| | - Li You
- School of Public Health, Guilin Medical University, 20 Lequn Road, Guilin, Guangxi, China
| | - Luo Tingyu
- School of Public Health, Guilin Medical University, 20 Lequn Road, Guilin, Guangxi, China
| | - Qin Jian
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning, 530021, Guangxi, China; Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, China.
| | - Zhang Zhiyong
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning, 530021, Guangxi, China; School of Public Health, Guilin Medical University, 20 Lequn Road, Guilin, Guangxi, China.
| |
Collapse
|
29
|
Barone I, Gilette NM, Hawks-Mayer H, Handy J, Zhang KJ, Chifamba FF, Mostafa E, Johnson-Venkatesh EM, Sun Y, Gibson JM, Rotenberg A, Umemori H, Tsai PT, Lipton JO. Synaptic BMAL1 phosphorylation controls circadian hippocampal plasticity. SCIENCE ADVANCES 2023; 9:eadj1010. [PMID: 37878694 PMCID: PMC10599629 DOI: 10.1126/sciadv.adj1010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/25/2023] [Indexed: 10/27/2023]
Abstract
The time of day strongly influences adaptive behaviors like long-term memory, but the correlating synaptic and molecular mechanisms remain unclear. The circadian clock comprises a canonical transcription-translation feedback loop (TTFL) strictly dependent on the BMAL1 transcription factor. We report that BMAL1 rhythmically localizes to hippocampal synapses in a manner dependent on its phosphorylation at Ser42 [pBMAL1(S42)]. pBMAL1(S42) regulates the autophosphorylation of synaptic CaMKIIα and circadian rhythms of CaMKIIα-dependent molecular interactions and LTP but not global rest/activity behavior. Therefore, our results suggest a model in which repurposing of the clock protein BMAL1 to synapses locally gates the circadian timing of plasticity.
Collapse
Affiliation(s)
- Ilaria Barone
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Nicole M. Gilette
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Hannah Hawks-Mayer
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Jonathan Handy
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Kevin J. Zhang
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Fortunate F. Chifamba
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Engie Mostafa
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Erin M. Johnson-Venkatesh
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Yan Sun
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Jennifer M. Gibson
- Departments of Neurology, Neuroscience, Pediatrics, and Psychiatry, University of Texas at Southwestern, Dallas, TX 75390, USA
| | - Alexander Rotenberg
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Hisashi Umemori
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Peter T. Tsai
- Departments of Neurology, Neuroscience, Pediatrics, and Psychiatry, University of Texas at Southwestern, Dallas, TX 75390, USA
| | - Jonathan O. Lipton
- Department of Neurology and F.M. Kirby Center for Neurobiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Neurology and Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
30
|
Millius A, Yamada RG, Fujishima H, Maeda K, Standley DM, Sumiyama K, Perrin D, Ueda HR. Circadian ribosome profiling reveals a role for the Period2 upstream open reading frame in sleep. Proc Natl Acad Sci U S A 2023; 120:e2214636120. [PMID: 37769257 PMCID: PMC10556633 DOI: 10.1073/pnas.2214636120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 08/29/2023] [Indexed: 09/30/2023] Open
Abstract
Many mammalian proteins have circadian cycles of production and degradation, and many of these rhythms are altered posttranscriptionally. We used ribosome profiling to examine posttranscriptional control of circadian rhythms by quantifying RNA translation in the liver over a 24-h period from circadian-entrained mice transferred to constant darkness conditions and by comparing ribosome binding levels to protein levels for 16 circadian proteins. We observed large differences in ribosome binding levels compared to protein levels, and we observed delays between peak ribosome binding and peak protein abundance. We found extensive binding of ribosomes to upstream open reading frames (uORFs) in circadian mRNAs, including the core clock gene Period2 (Per2). An increase in the number of uORFs in the 5'UTR was associated with a decrease in ribosome binding in the main coding sequence and a reduction in expression of synthetic reporter constructs. Mutation of the Per2 uORF increased luciferase and fluorescence reporter expression in 3T3 cells and increased luciferase expression in PER2:LUC MEF cells. Mutation of the Per2 uORF in mice increased Per2 mRNA expression, enhanced ribosome binding on Per2, and reduced total sleep time compared to that in wild-type mice. These results suggest that uORFs affect mRNA posttranscriptionally, which can impact physiological rhythms and sleep.
Collapse
Affiliation(s)
- Arthur Millius
- Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, Suita, Osaka565-0871, Japan
- Laboratory for Host Defense, Immunology Frontier Research Center, Suita, Osaka565-0871, Japan
- Laboratory for Systems Immunology, Immunology Frontier Research Center, Suita, Osaka565-0871, Japan
| | - Rikuhiro G. Yamada
- Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, Suita, Osaka565-0871, Japan
| | - Hiroshi Fujishima
- Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, Suita, Osaka565-0871, Japan
| | - Kazuhiko Maeda
- Laboratory for Host Defense, Immunology Frontier Research Center, Suita, Osaka565-0871, Japan
| | - Daron M. Standley
- Laboratory for Systems Immunology, Immunology Frontier Research Center, Suita, Osaka565-0871, Japan
| | - Kenta Sumiyama
- Laboratory of Animal Genetics and Breeding, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya464-8601, Japan
| | - Dimitri Perrin
- School of Computer Science, Queensland University of Technology, BrisbaneQLD 4000, Australia
- Centre for Data Science, Queensland University of Technology, BrisbaneQLD 4000, Australia
| | - Hiroki R. Ueda
- Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, Suita, Osaka565-0871, Japan
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo113-0033, Japan
| |
Collapse
|
31
|
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
|
32
|
Jung J, Kang J, Kim T. Attenuation of homeostatic sleep response and rest-activity circadian rhythm in vitamin D deficient mice. Chronobiol Int 2023; 40:1097-1110. [PMID: 37661839 DOI: 10.1080/07420528.2023.2253299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/09/2023] [Accepted: 08/24/2023] [Indexed: 09/05/2023]
Abstract
The link between vitamin D deficiency (VDD) and sleep disturbances has long been suggested. However, the direct causality between VDD, sleep disturbances, and circadian rhythm remains unclear. We aimed to characterize sleep-wake behavior and circadian rhythms in an animal model of VDD. VDD was induced by feeding vitamin D-deficient chow, and we analyzed sleep and circadian rhythm parameters. During light period, VDD mice exhibited reduced wake with more frequent wake bouts and increased NREM sleep time. However, during dark period, the wake EEG power spectrum peaked at theta band frequency, and slow-wave energy was suppressed in mice with VDD. Rest-activity analyses revealed increased circadian period, lower wheel counts, and more frequent and short activity bouts during VDD. Combining sleep and circadian data, we found significantly suppressed activities during the hours with a wake duration shorter than 30 minutes. Moreover, mice in VDD state exhibited a negative correlation between wake theta power and hourly wheel-running counts during dark period. Our data point to a direct link between VDD and disturbances in sleep and rest-activity circadian rhythm, featuring frequent wake bouts during the sleeping phase, reduced sleep pressure build-up in dark period, and reduced activity levels due to increased susceptibility to sleepiness.
Collapse
Affiliation(s)
- Jieun Jung
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Jiseung Kang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Tae Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| |
Collapse
|
33
|
Untiet V, Beinlich FRM, Kusk P, Kang N, Ladrón-de-Guevara A, Song W, Kjaerby C, Andersen M, Hauglund N, Bojarowska Z, Sigurdsson B, Deng S, Hirase H, Petersen NC, Verkhratsky A, Nedergaard M. Astrocytic chloride is brain state dependent and modulates inhibitory neurotransmission in mice. Nat Commun 2023; 14:1871. [PMID: 37015909 PMCID: PMC10073105 DOI: 10.1038/s41467-023-37433-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/15/2023] [Indexed: 04/06/2023] Open
Abstract
Information transfer within neuronal circuits depends on the balance and recurrent activity of excitatory and inhibitory neurotransmission. Chloride (Cl-) is the major central nervous system (CNS) anion mediating inhibitory neurotransmission. Astrocytes are key homoeostatic glial cells populating the CNS, although the role of these cells in regulating excitatory-inhibitory balance remains unexplored. Here we show that astrocytes act as a dynamic Cl- reservoir regulating Cl- homoeostasis in the CNS. We found that intracellular chloride concentration ([Cl-]i) in astrocytes is high and stable during sleep. In awake mice astrocytic [Cl-]i is lower and exhibits large fluctuation in response to both sensory input and motor activity. Optogenetic manipulation of astrocytic [Cl-]i directly modulates neuronal activity during locomotion or whisker stimulation. Astrocytes thus serve as a dynamic source of extracellular Cl- available for GABAergic transmission in awake mice, which represents a mechanism for modulation of the inhibitory tone during sustained neuronal activity.
Collapse
Affiliation(s)
- Verena Untiet
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark.
| | - Felix R M Beinlich
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Peter Kusk
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Ning Kang
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Antonio Ladrón-de-Guevara
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Wei Song
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Celia Kjaerby
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Mie Andersen
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Natalie Hauglund
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Zuzanna Bojarowska
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Björn Sigurdsson
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Saiyue Deng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P.R. China
| | - Hajime Hirase
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Nicolas C Petersen
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Alexei Verkhratsky
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark.
- Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Rd, Manchester, M13 9PL, UK.
- Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, 48009, Bilbao, Spain.
| | - Maiken Nedergaard
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark.
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
34
|
Eacret D, Manduchi E, Noreck J, Tyner E, Fenik P, Dunn AD, Schug J, Veasey SC, Blendy JA. Mu-opioid receptor-expressing neurons in the paraventricular thalamus modulate chronic morphine-induced wake alterations. Transl Psychiatry 2023; 13:78. [PMID: 36869037 PMCID: PMC9984393 DOI: 10.1038/s41398-023-02382-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023] Open
Abstract
Disrupted sleep is a symptom of many psychiatric disorders, including substance use disorders. Most drugs of abuse, including opioids, disrupt sleep. However, the extent and consequence of opioid-induced sleep disturbance, especially during chronic drug exposure, is understudied. We have previously shown that sleep disturbance alters voluntary morphine intake. Here, we examine the effects of acute and chronic morphine exposure on sleep. Using an oral self-administration paradigm, we show that morphine disrupts sleep, most significantly during the dark cycle in chronic morphine, with a concomitant sustained increase in neural activity in the Paraventricular Nucleus of the Thalamus (PVT). Morphine binds primarily to Mu Opioid Receptors (MORs), which are highly expressed in the PVT. Translating Ribosome Affinity Purification (TRAP)-Sequencing of PVT neurons that express MORs showed significant enrichment of the circadian entrainment pathway. To determine whether MOR + cells in the PVT mediate morphine-induced sleep/wake properties, we inhibited these neurons during the dark cycle while mice were self-administering morphine. This inhibition decreased morphine-induced wakefulness but not general wakefulness, indicating that MORs in the PVT contribute to opioid-specific wake alterations. Overall, our results suggest an important role for PVT neurons that express MORs in mediating morphine-induced sleep disturbance.
Collapse
Affiliation(s)
- Darrell Eacret
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elisabetta Manduchi
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Julia Noreck
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emma Tyner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Polina Fenik
- Center for Sleep and Circadian Neurobiology and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Amelia D Dunn
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan Schug
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sigrid C Veasey
- Center for Sleep and Circadian Neurobiology and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Julie A Blendy
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Carbone EA, Menculini G, de Filippis R, D’Angelo M, De Fazio P, Tortorella A, Steardo L. Sleep Disturbances in Generalized Anxiety Disorder: The Role of Calcium Homeostasis Imbalance. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4431. [PMID: 36901441 PMCID: PMC10002427 DOI: 10.3390/ijerph20054431] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/16/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
Patients with a generalized anxiety disorder (GAD) often report preeminent sleep disturbances. Recently, calcium homeostasis gained interest because of its role in the regulation of sleep-wake rhythms and anxiety symptoms. This cross-sectional study aimed at investigating the association between calcium homeostasis imbalance, anxiety, and quality of sleep in patients with GAD. A total of 211 patients were assessed using the Hamilton Rating Scale for Anxiety (HAM-A), Pittsburgh Sleep Quality Index questionnaire (PSQI) and Insomnia Severity Index (ISI) scales. Calcium, vitamin D, and parathyroid hormone (PTH) levels were evaluated in blood samples. A correlation and linear regression analysis were run to evaluate the association of HAM-A, PSQI, and ISI scores with peripheral markers of calcium homeostasis imbalance. Significant correlations emerged between HAM-A, PSQI, ISI, PTH, and vitamin D. The regression models showed that patients with GAD displaying low levels of vitamin D and higher levels of PTH exhibit a poor subjective quality of sleep and higher levels of anxiety, underpinning higher psychopathological burden. A strong relationship between peripheral biomarkers of calcium homeostasis imbalance, insomnia, poor sleep quality, and anxiety symptomatology was underlined. Future studies could shed light on the causal and temporal relationship between calcium metabolism imbalance, anxiety, and sleep.
Collapse
Affiliation(s)
- Elvira Anna Carbone
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
| | - Giulia Menculini
- Department of Psychiatry, University of Perugia, Piazzale Lucio Severi, 1, 06132 Perugia, Italy
| | - Renato de Filippis
- Department of Health Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy
| | - Martina D’Angelo
- Department of Health Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy
| | - Pasquale De Fazio
- Department of Health Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy
| | - Alfonso Tortorella
- Department of Psychiatry, University of Perugia, Piazzale Lucio Severi, 1, 06132 Perugia, Italy
| | - Luca Steardo
- Department of Health Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy
| |
Collapse
|
36
|
Hazuga MA, Grant SFA. Awakening new sleep biology with machine learning. Sleep 2023; 46:zsac284. [PMID: 36422063 PMCID: PMC9905772 DOI: 10.1093/sleep/zsac284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Indexed: 11/27/2022] Open
Affiliation(s)
- Mary Ann Hazuga
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Struan F A Grant
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Diabetes and Endocrinology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
37
|
Lee YY, Endale M, Wu G, Ruben MD, Francey LJ, Morris AR, Choo NY, Anafi RC, Smith DF, Liu AC, Hogenesch JB. Integration of genome-scale data identifies candidate sleep regulators. Sleep 2023; 46:zsac279. [PMID: 36462188 PMCID: PMC9905783 DOI: 10.1093/sleep/zsac279] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/02/2022] [Indexed: 12/05/2022] Open
Abstract
STUDY OBJECTIVES Genetics impacts sleep, yet, the molecular mechanisms underlying sleep regulation remain elusive. In this study, we built machine learning models to predict sleep genes based on their similarity to genes that are known to regulate sleep. METHODS We trained a prediction model on thousands of published datasets, representing circadian, immune, sleep deprivation, and many other processes, using a manually curated list of 109 sleep genes. RESULTS Our predictions fit with prior knowledge of sleep regulation and identified key genes and pathways to pursue in follow-up studies. As an example, we focused on the NF-κB pathway and showed that chronic activation of NF-κB in a genetic mouse model impacted the sleep-wake patterns. CONCLUSION Our study highlights the power of machine learning in integrating prior knowledge and genome-wide data to study genetic regulation of complex behaviors such as sleep.
Collapse
Affiliation(s)
- Yin Yeng Lee
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Mehari Endale
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Gang Wu
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Marc D Ruben
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Lauren J Francey
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Andrew R Morris
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Natalie Y Choo
- Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ron C Anafi
- Department of Medicine, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David F Smith
- Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Division of Pulmonary Medicine and the Sleep Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Center for Circadian Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Otolaryngology - Head and Neck Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Andrew C Liu
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - John B Hogenesch
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Center for Circadian Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
38
|
Pinto MJ, Cottin L, Dingli F, Laigle V, Ribeiro LF, Triller A, Henderson F, Loew D, Fabre V, Bessis A. Microglial TNFα orchestrates protein phosphorylation in the cortex during the sleep period and controls homeostatic sleep. EMBO J 2023; 42:e111485. [PMID: 36385434 PMCID: PMC9811617 DOI: 10.15252/embj.2022111485] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 10/04/2022] [Accepted: 10/11/2022] [Indexed: 11/18/2022] Open
Abstract
Sleep intensity is adjusted by the length of previous awake time, and under tight homeostatic control by protein phosphorylation. Here, we establish microglia as a new cellular component of the sleep homeostasis circuit. Using quantitative phosphoproteomics of the mouse frontal cortex, we demonstrate that microglia-specific deletion of TNFα perturbs thousands of phosphorylation sites during the sleep period. Substrates of microglial TNFα comprise sleep-related kinases such as MAPKs and MARKs, and numerous synaptic proteins, including a subset whose phosphorylation status encodes sleep need and determines sleep duration. As a result, microglial TNFα loss attenuates the build-up of sleep need, as measured by electroencephalogram slow-wave activity and prevents immediate compensation for loss of sleep. Our data suggest that microglia control sleep homeostasis by releasing TNFα which acts on neuronal circuitry through dynamic control of phosphorylation.
Collapse
Affiliation(s)
- Maria J Pinto
- Institut de Biologie de l'École normale supérieure (IBENS), École normale supérieure, CNRS, INSERMUniversité PSLParisFrance
| | - Léa Cottin
- Institut de Biologie de l'École normale supérieure (IBENS), École normale supérieure, CNRS, INSERMUniversité PSLParisFrance
| | - Florent Dingli
- Centre de Recherche, Laboratoire de Spectrométrie de Masse ProtéomiqueInstitut Curie, PSL Research UniversityParisFrance
| | - Victor Laigle
- Centre de Recherche, Laboratoire de Spectrométrie de Masse ProtéomiqueInstitut Curie, PSL Research UniversityParisFrance
| | - Luís F Ribeiro
- Center for Neuroscience and Cell Biology (CNC), Institute for Interdisciplinary Research (IIIUC)University of CoimbraCoimbraPortugal
| | - Antoine Triller
- Institut de Biologie de l'École normale supérieure (IBENS), École normale supérieure, CNRS, INSERMUniversité PSLParisFrance
| | - Fiona Henderson
- Sorbonne Université, CNRS UMR 8246, INSERM U1130, Neuroscience Paris Seine – Institut de Biologie Paris Seine (NPS – IBPS)ParisFrance
| | - Damarys Loew
- Centre de Recherche, Laboratoire de Spectrométrie de Masse ProtéomiqueInstitut Curie, PSL Research UniversityParisFrance
| | - Véronique Fabre
- Sorbonne Université, CNRS UMR 8246, INSERM U1130, Neuroscience Paris Seine – Institut de Biologie Paris Seine (NPS – IBPS)ParisFrance
| | - Alain Bessis
- Institut de Biologie de l'École normale supérieure (IBENS), École normale supérieure, CNRS, INSERMUniversité PSLParisFrance
| |
Collapse
|
39
|
Alfonsa H, Burman RJ, Brodersen PJN, Newey SE, Mahfooz K, Yamagata T, Panayi MC, Bannerman DM, Vyazovskiy VV, Akerman CJ. Intracellular chloride regulation mediates local sleep pressure in the cortex. Nat Neurosci 2023; 26:64-78. [PMID: 36510112 PMCID: PMC7614036 DOI: 10.1038/s41593-022-01214-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 10/25/2022] [Indexed: 12/14/2022]
Abstract
Extended wakefulness is associated with reduced performance and the build-up of sleep pressure. In the cortex, this manifests as changes in network activity. These changes show local variation depending on the waking experience, and their underlying mechanisms represent targets for overcoming the effects of tiredness. Here, we reveal a central role for intracellular chloride regulation, which sets the strength of postsynaptic inhibition via GABAA receptors in cortical pyramidal neurons. Wakefulness results in depolarizing shifts in the equilibrium potential for GABAA receptors, reflecting local activity-dependent processes during waking and involving changes in chloride cotransporter activity. These changes underlie electrophysiological and behavioral markers of local sleep pressure within the cortex, including the levels of slow-wave activity during non-rapid eye movement sleep and low-frequency oscillatory activity and reduced performance levels in the sleep-deprived awake state. These findings identify chloride regulation as a crucial link between sleep-wake history, cortical activity and behavior.
Collapse
Affiliation(s)
- Hannah Alfonsa
- Department of Pharmacology, University of Oxford, Oxford, UK.
| | | | | | - Sarah E Newey
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Kashif Mahfooz
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Tomoko Yamagata
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Marios C Panayi
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | | | - Colin J Akerman
- Department of Pharmacology, University of Oxford, Oxford, UK.
| |
Collapse
|
40
|
Liu S, Ba Y, Li C, Xu G. Inactivation of CACNA1H induces cell apoptosis by initiating endoplasmic reticulum stress in glioma. Transl Neurosci 2023; 14:20220285. [PMID: 37250140 PMCID: PMC10224624 DOI: 10.1515/tnsci-2022-0285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/04/2023] [Accepted: 04/16/2023] [Indexed: 05/31/2023] Open
Abstract
Background Ca2+ channels are abnormally expressed in various tumor cells and are involved in the progression of human glioma. Here, we explored the role of a calcium channel, voltage-dependent, T-type, alpha 1H subunit (CACNA1H), which encodes T-type Ca2+ channel Cav3.2 in glioma cells. Methods Cell viability and apoptosis were detected using cell-counting kit-8 and flow cytometry, respectively. The expression of target protein was determined using western blot analysis. Results Cell viability of U251 cells was inhibited significantly after the knockdown of CACNA1H. The apoptosis of U251 cells was enhanced significantly after the knockdown of CACNA1H. Importantly, knockdown of CACNA1H decreased the levels of p-PERK, GRP78, CHOP, and ATF6, indicating that CACNA1H knockdown activated endoplasmic reticulum stress (ERS) in U251 cells. In addition, T-type Ca2+ channel inhibitor NNC55-0396 also induced apoptosis through the activation of ERS in U251 cells. ERS inhibitor UR906 could block CACNA1H inhibitor ABT-639-induced apoptosis. Conclusion Suppression of CACNA1H activated the ERS and thus induced apoptosis in glioma cells. T-type Ca2+ channel inhibitors ABT-639 and NNC55-0396 also induced apoptosis through ERS in glioma cells. Our data highlighted the effect of CACNA1H as an oncogenic gene in human glioma.
Collapse
Affiliation(s)
- Sheng Liu
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Ying Ba
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Chenglong Li
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Guangming Xu
- Department of Neurosurgery, Shandong Provincial Hospital, Shandong University, No. 324, Jingwuweiqi Road, Jinan, 250021, China
| |
Collapse
|
41
|
Diering GH. Remembering and forgetting in sleep: Selective synaptic plasticity during sleep driven by scaling factors Homer1a and Arc. Neurobiol Stress 2022; 22:100512. [PMID: 36632309 PMCID: PMC9826981 DOI: 10.1016/j.ynstr.2022.100512] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 12/01/2022] [Accepted: 12/29/2022] [Indexed: 01/02/2023] Open
Abstract
Sleep is a conserved and essential process that supports learning and memory. Synapses are a major target of sleep function and a locus of sleep need. Evidence in the literature suggests that the need for sleep has a cellular or microcircuit level basis, and that sleep need can accumulate within localized brain regions as a function of waking activity. Activation of sleep promoting kinases and accumulation of synaptic phosphorylation was recently shown to be part of the molecular basis for the localized sleep need. A prominent hypothesis in the field suggests that some benefits of sleep are mediated by a broad but selective weakening, or scaling-down, of synaptic strength during sleep in order to offset increased excitability from synaptic potentiation during wake. The literature also shows that synapses can be strengthened during sleep, raising the question of what molecular mechanisms may allow for selection of synaptic plasticity types during sleep. Here I describe mechanisms of action of the scaling factors Arc and Homer1a in selective plasticity and links with sleep need. Arc and Homer1a are induced in neurons in response to waking neuronal activity and accumulate with time spent awake. I suggest that during sleep, Arc and Homer1a drive broad weakening of synapses through homeostatic scaling-down, but in a manner that is sensitive to the plasticity history of individual synapses, based on patterned phosphorylation of synaptic proteins. Therefore, Arc and Homer1a may offer insights into the intricate links between a cellular basis of sleep need and memory consolidation during sleep.
Collapse
Affiliation(s)
- Graham H. Diering
- Department of Cell Biology and Physiology and the UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Carolina Institute for Developmental Disabilities, USA,111 Mason Farm Road, 5200 Medical and Biomolecular Research Building, Chapel Hill, NC, 27599-7545, USA.
| |
Collapse
|
42
|
Offline neuronal activity and synaptic plasticity during sleep and memory consolidation. Neurosci Res 2022; 189:29-36. [PMID: 36584924 DOI: 10.1016/j.neures.2022.12.021] [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: 08/22/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/29/2022]
Abstract
After initial formation during learning, memories are further processed in the brain during subsequent days for long-term consolidation, with sleep playing a key role in this process. Studies have shown that neuronal activity patterns during the awake period are repeated in the hippocampus during sleep, which may coordinate brain-wide reactivation leading to memory consolidation. Consistently, perturbation of this activity blocks the formation of long-term memory. This 'replay' of activity during sleep likely triggers plastic changes in synaptic transmission, a cellular substrate of memory, in multiple brain regions, which likely plays a critical role in long-term memory. Two forms of synaptic plasticity, potentiation and depression of synaptic transmission, are induced in parallel during sleep and is termed "offline synaptic plasticity", as opposed to the "online synaptic plasticity" that occurs immediately following a memory event.
Collapse
|
43
|
Wang Y, Minami Y, Ode KL, Ueda HR. The role of calcium and CaMKII in sleep. Front Syst Neurosci 2022; 16:1059421. [PMID: 36618010 PMCID: PMC9815122 DOI: 10.3389/fnsys.2022.1059421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Sleep is an evolutionarily conserved phenotype shared by most of the animals on the planet. Prolonged wakefulness will result in increased sleep need or sleep pressure. However, its mechanisms remain elusive. Recent findings indicate that Ca2+ signaling, known to control diverse physiological functions, also regulates sleep. This review intends to summarize research advances in Ca2+ and Ca2+/calmodulin-dependent protein kinase II (CaMKII) in sleep regulation. Significant changes in sleep phenotype have been observed through calcium-related channels, receptors, and pumps. Mathematical modeling for neuronal firing patterns during NREM sleep suggests that these molecules compose a Ca2+-dependent hyperpolarization mechanism. The intracellular Ca2+ may then trigger sleep induction and maintenance through the activation of CaMKII, one of the sleep-promoting kinases. CaMKII and its multisite phosphorylation status may provide a link between transient calcium dynamics typically observed in neurons and sleep-wake dynamics observed on the long-time scale.
Collapse
Affiliation(s)
- Yuyang Wang
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoichi Minami
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Koji L. Ode
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroki R. Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan,Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Japan,*Correspondence: Hiroki R. Ueda,
| |
Collapse
|
44
|
Ozawa M, Taguchi J, Katsuma K, Ishikawa-Yamauchi Y, Kikuchi M, Sakamoto R, Yamada Y, Ikawa M. Efficient simultaneous double DNA knock-in in murine embryonic stem cells by CRISPR/Cas9 ribonucleoprotein-mediated circular plasmid targeting for generating gene-manipulated mice. Sci Rep 2022; 12:21558. [PMID: 36513736 PMCID: PMC9748034 DOI: 10.1038/s41598-022-26107-z] [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: 03/18/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Gene targeting of embryonic stem (ES) cells followed by chimera production has been conventionally used for developing gene-manipulated mice. Although direct knock-in (KI) using murine zygote via CRISPR/Cas9-mediated genome editing has been reported, ES cell targeting still has merits, e.g., high throughput work can be performed in vitro. In this study, we first compared the KI efficiency of mouse ES cells with CRISPR/Cas9 expression vector and ribonucleoprotein (RNP), and confirmed that KI efficiency was significantly increased by using RNP. Using CRISPR/Cas9 RNP and circular plasmid with homologous arms as a targeting vector, knock-in within ES cell clones could be obtained efficiently without drug selection, thus potentially shortening the vector construction or cell culture period. Moreover, by incorporating a drug-resistant cassette into the targeting vectors, double DNA KI can be simultaneously achieved at high efficiency by a single electroporation. This technique will help to facilitate the production of genetically modified mouse models that are fundamental for exploring topics related to human and mammalian biology.
Collapse
Affiliation(s)
- Manabu Ozawa
- grid.26999.3d0000 0001 2151 536XLaboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Jumpei Taguchi
- grid.26999.3d0000 0001 2151 536XDivision of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Kento Katsuma
- grid.26999.3d0000 0001 2151 536XLaboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Yu Ishikawa-Yamauchi
- grid.26999.3d0000 0001 2151 536XLaboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Mio Kikuchi
- grid.26999.3d0000 0001 2151 536XDivision of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Reiko Sakamoto
- grid.26999.3d0000 0001 2151 536XDivision of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Yasuhiro Yamada
- grid.26999.3d0000 0001 2151 536XDivision of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Masahito Ikawa
- grid.26999.3d0000 0001 2151 536XLaboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan ,grid.136593.b0000 0004 0373 3971Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871 Japan
| |
Collapse
|
45
|
Savy V, Stein P, Shi M, Williams CJ. PMCA1 depletion in mouse eggs amplifies calcium signaling and impacts offspring growth†. Biol Reprod 2022; 107:1439-1451. [PMID: 36130203 PMCID: PMC10144700 DOI: 10.1093/biolre/ioac180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/19/2022] [Accepted: 09/19/2022] [Indexed: 11/12/2022] Open
Abstract
Egg activation in mammals is triggered by oscillations in egg intracellular calcium (Ca2+) level. Ca2+ oscillation patterns can be modified in vitro by changing the ionic composition of culture media or in vivo by conditions affecting mitochondrial function, such as obesity and inflammation. In mice, disruption of Ca2+ oscillations in vitro impacts embryo development and offspring growth. Here we tested the hypothesis that, even without in vitro manipulation, abnormal Ca2+ signaling following fertilization impacts offspring growth. Plasma membrane Ca2+ ATPases (PMCA) extrude cytosolic Ca2+ to restore Ca2+ homeostasis. To disrupt Ca2+ signaling in vivo, we conditionally deleted PMCA1 (cKO) in oocytes. As anticipated, in vitro fertilized cKO eggs had increased Ca2+ exposure relative to controls. To assess the impact on offspring growth, cKO females were mated to wild type males to generate pups that had high Ca2+ exposure at fertilization. Because these offspring would be heterozygous, we also tested the impact of global PMCA1 heterozygosity on offspring growth. Control heterozygous pups that had normal Ca2+ at fertilization were generated by mating wild type females to heterozygous males; these control offspring weighed significantly less than their wild type siblings. However, heterozygous offspring from cKO eggs (and high Ca2+ exposure) were larger than heterozygous controls at 12 week-of-age and males had altered body composition. Our results show that global PMCA1 haploinsufficiency impacts growth and support that abnormal Ca2+ signaling after fertilization in vivo has a long-term impact on offspring weight. These findings are relevant for environmental and medical conditions affecting Ca2+ handling and for design of culture conditions and procedures for domestic animal and human assisted reproduction.
Collapse
Affiliation(s)
- Virginia Savy
- Reproductive & Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Paula Stein
- Reproductive & Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Min Shi
- Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Carmen J Williams
- Reproductive & Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| |
Collapse
|
46
|
Yoshida K, Toyoizumi T. Information maximization explains state-dependent synaptic plasticity and memory reorganization during non-rapid eye movement sleep. PNAS NEXUS 2022; 2:pgac286. [PMID: 36712943 PMCID: PMC9833047 DOI: 10.1093/pnasnexus/pgac286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Slow waves during the non-rapid eye movement (NREM) sleep reflect the alternating up and down states of cortical neurons; global and local slow waves promote memory consolidation and forgetting, respectively. Furthermore, distinct spike-timing-dependent plasticity (STDP) operates in these up and down states. The contribution of different plasticity rules to neural information coding and memory reorganization remains unknown. Here, we show that optimal synaptic plasticity for information maximization in a cortical neuron model provides a unified explanation for these phenomena. The model indicates that the optimal synaptic plasticity is biased toward depression as the baseline firing rate increases. This property explains the distinct STDP observed in the up and down states. Furthermore, it explains how global and local slow waves predominantly potentiate and depress synapses, respectively, if the background firing rate of excitatory neurons declines with the spatial scale of waves as the model predicts. The model provides a unifying account of the role of NREM sleep, bridging neural information coding, synaptic plasticity, and memory reorganization.
Collapse
|
47
|
Haraguchi A, Saito K, Tahara Y, Shibata S. Polygalae Radix shortens the circadian period through activation of the CaMKII pathway. PHARMACEUTICAL BIOLOGY 2022; 60:689-698. [PMID: 35298359 PMCID: PMC8933028 DOI: 10.1080/13880209.2022.2048863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 06/14/2023]
Abstract
CONTEXT The mammalian circadian clock system regulates physiological function. Crude drugs, containing Polygalae Radix, and Kampō, combining multiple crude drugs, have been used to treat various diseases, but few studies have focussed on the circadian clock. OBJECTIVE We examine effective crude drugs, which cover at least one or two of Kampō, for the shortening effects on period length of clock gene expression rhythm, and reveal the mechanism of shortening effects. MATERIALS AND METHODS We prepared 40 crude drugs. In the in vitro experiments, we used mouse embryonic fibroblasts from PERIOD2::LUCIFERASE knock-in mice (background; C57BL/6J mice) to evaluate the effect of crude drugs on the period length of core clock gene, Per2, expression rhythm by chronic treatment (six days) with distilled water or crude drugs (100 μg/mL). In the in vivo experiments, we evaluated the free-running period length of C57BL/6J mice fed AIN-93M or AIN-93M supplemented with 1% crude drug (6 weeks) that shortened the period length of the PERIOD2::LUCIFERASE expression rhythm in the in vitro experiments. RESULTS We found that Polygalae Radix (ED50: 24.01 μg/mL) had the most shortened PERIOD2::LUCIFERASE rhythm period length in 40 crude drugs and that the CaMKII pathway was involved in this effect. Moreover, long-term feeding with AIN-93M+Polygalae Radix slightly shortened the free-running period of the mouse locomotor activity rhythm. DISCUSSION AND CONCLUSIONS Our results indicate that Polygalae Radix may be regarded as a new therapy for circadian rhythm disorder and that the CaMKII pathway may be regarded as a target pathway for circadian rhythm disorders.
Collapse
Affiliation(s)
- Atsushi Haraguchi
- School of Advanced Science and Engineering, Laboratory of Physiology and Pharmacology, Waseda University, Tokyo, Japan
| | - Keisuke Saito
- School of Advanced Science and Engineering, Laboratory of Physiology and Pharmacology, Waseda University, Tokyo, Japan
| | - Yu Tahara
- School of Advanced Science and Engineering, Laboratory of Physiology and Pharmacology, Waseda University, Tokyo, Japan
| | - Shigenobu Shibata
- School of Advanced Science and Engineering, Laboratory of Physiology and Pharmacology, Waseda University, Tokyo, Japan
| |
Collapse
|
48
|
Kim RE, Mabunga DF, Kim HJ, Han SH, Kim HY, Shin CY, Kwon KJ. Novel Therapeutics for Treating Sleep Disorders: New Perspectives on Maydis stigma. Int J Mol Sci 2022; 23:ijms232314612. [PMID: 36498940 PMCID: PMC9740493 DOI: 10.3390/ijms232314612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/26/2022] [Accepted: 11/05/2022] [Indexed: 11/24/2022] Open
Abstract
Sleep is a restorative period that plays a crucial role in the physiological functioning of the body, including that of the immune system, memory processing, and cognition. Sleep disturbances can be caused by various physical, mental, and social problems. Recently, there has been growing interest in sleep. Maydis stigma (MS, corn silk) is a female maize flower that is traditionally used as a medicinal plant to treat many diseases, including hypertension, edema, and diabetes. It is also used as a functional food in tea and other supplements. β-Sitosterol (BS) is a phytosterol and a natural micronutrient in higher plants, and it has a similar structure to cholesterol. It is a major component of MS and has anti-inflammatory, antidepressive, and sedative effects. However, the potential effects of MS on sleep regulation remain unclear. Here, we investigated the effects of MS on sleep in mice. The effects of MS on sleep induction were determined using pentobarbital-induced sleep and caffeine-induced sleep disruption mouse models. MS extracts decreased sleep latency and increased sleep duration in both the pentobarbital-induced sleep induction and caffeine-induced sleep disruption models compared to the positive control, valerian root extract. The butanol fraction of MS extracts decreased sleep latency time and increased sleep duration. In addition, β-sitosterol enhances sleep latency and sleep duration. Both MS extract and β-sitosterol increased alpha activity in the EEG analysis. We measured the mRNA expression of melatonin receptors 1 and 2 (MT1/2) using qRT-PCR. The mRNA expression of melatonin receptors 1 and 2 was increased by MS extract and β-sitosterol treatment in rat primary cultured neurons and the brain. In addition, MS extract increased the expression of clock genes including per1/2, cry1/2, and Bmal1 in the brain. MS extract and β-sitosterol increased the phosphorylation of ERK1/2 and αCaMKII. Our results demonstrate for the first time that MS has a sleep-promoting effect via melatonin receptor expression, which may provide new scientific evidence for its use as a potential therapeutic agent for the treatment and prevention of sleep disturbance.
Collapse
Affiliation(s)
- Ryeong-Eun Kim
- Department of Neuroscience, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Darine Froy Mabunga
- Department of Neuroscience, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hee Jin Kim
- Department of Pharmacy, Uimyung Research Institute for Neuroscience, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul 01795, Republic of Korea
| | - Seol-Heui Han
- Department of Neurology, Konkuk Hospital Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea
| | - Hahn Young Kim
- Department of Neurology, Konkuk Hospital Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea
| | - Chan Young Shin
- Department of Pharmacology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Kyoung Ja Kwon
- Department of Neuroscience, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
- Department of Neurology, Konkuk Hospital Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea
- Correspondence: ; Tel.: +82-2-454-5630; Fax: +82-2030-7899
| |
Collapse
|
49
|
Han G, Matsumoto S, Diaz J, Greene RW, Vogt KE. Dihydropyridine calcium blockers do not interfere with non-rapid eye movement sleep. Front Neurosci 2022; 16:969712. [PMID: 36340773 PMCID: PMC9626806 DOI: 10.3389/fnins.2022.969712] [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: 06/16/2022] [Accepted: 09/21/2022] [Indexed: 09/07/2024] Open
Abstract
Non-rapid eye movement (NREM) sleep is tightly homeostatically regulated and essential for survival. In the electroencephalogram (EEG), oscillations in the delta (0.5-4 Hz) range are prominent during NREM sleep. These delta oscillations are, to date, the best indicator for homeostatic sleep regulation; they are increased after prolonged waking and fade during NREM sleep. The precise mechanisms underlying sleep homeostasis and the generation of EEG delta oscillations are still being investigated. Activity-dependent neuronal calcium influx has been hypothesized to play an important role in generating delta oscillations and might be involved in downstream signaling that mediates sleep function. Dihydropyridine blockers of L-type voltage-gated calcium channels (VGCCs) are in wide clinical use to treat hypertension and other cardiovascular disorders and are readily blood-brain-barrier penetrant. We therefore, wanted to investigate their potential effects on EEG delta oscillation and homeostatic NREM sleep regulation in freely behaving mice. In vivo two-photon imaging of cortical neurons showed larger spontaneous calcium transients in NREM sleep compared to waking. Application of the dihydropyridine calcium blocker nicardipine significantly reduced cortical calcium transients without affecting the generation of delta oscillations. Nicardipine also did not affect EEG delta oscillations over 24 h following application. The time spent in NREM sleep and NREM episode duration was also not affected. Thus, acute block of calcium entry through L-type VGCCs does not interfere with EEG delta oscillations or their homeostatic regulation, despite prior evidence from calcium channel knockout mice.
Collapse
Affiliation(s)
- GoEun Han
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Sumire Matsumoto
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Javier Diaz
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Robert W. Greene
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Department of Psychiatry & Neuroscience, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, United States
| | - Kaspar E. Vogt
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
50
|
Tone D, Ode KL, Zhang Q, Fujishima H, Yamada RG, Nagashima Y, Matsumoto K, Wen Z, Yoshida SY, Mitani TT, Arisato Y, Ohno RI, Ukai-Tadenuma M, Yoshida Garçon J, Kaneko M, Shi S, Ukai H, Miyamichi K, Okada T, Sumiyama K, Kiyonari H, Ueda HR. Distinct phosphorylation states of mammalian CaMKIIβ control the induction and maintenance of sleep. PLoS Biol 2022; 20:e3001813. [PMID: 36194579 PMCID: PMC9531794 DOI: 10.1371/journal.pbio.3001813] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
The reduced sleep duration previously observed in Camk2b knockout mice revealed a role for Ca2+/calmodulin-dependent protein kinase II (CaMKII)β as a sleep-promoting kinase. However, the underlying mechanism by which CaMKIIβ supports sleep regulation is largely unknown. Here, we demonstrate that activation or inhibition of CaMKIIβ can increase or decrease sleep duration in mice by almost 2-fold, supporting the role of CaMKIIβ as a core sleep regulator in mammals. Importantly, we show that this sleep regulation depends on the kinase activity of CaMKIIβ. A CaMKIIβ mutant mimicking the constitutive-active (auto)phosphorylation state promotes the transition from awake state to sleep state, while mutants mimicking subsequent multisite (auto)phosphorylation states suppress the transition from sleep state to awake state. These results suggest that the phosphorylation states of CaMKIIβ differently control sleep induction and maintenance processes, leading us to propose a "phosphorylation hypothesis of sleep" for the molecular control of sleep in mammals.
Collapse
Affiliation(s)
- Daisuke Tone
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Koji L. Ode
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Qianhui Zhang
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Fujishima
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Rikuhiro G. Yamada
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Yoshiki Nagashima
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Thermo Fisher Scientific K.K., Yokohama, Kanagawa, Japan
| | - Katsuhiko Matsumoto
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Zhiqing Wen
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shota Y. Yoshida
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Graduate school of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tomoki T. Mitani
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Graduate school of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yuki Arisato
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Rei-ichiro Ohno
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Maki Ukai-Tadenuma
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Junko Yoshida Garçon
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Mari Kaneko
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe, Hyogo, Japan
| | - Shoi Shi
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hideki Ukai
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Kazunari Miyamichi
- Laboratory for Comparative Connections, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe, Hyogo, Japan
| | - Takashi Okada
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, the University of Tokyo, Minato-city, Tokyo, Japan
| | - Kenta Sumiyama
- Laboratory for Mouse Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe, Hyogo, Japan
| | - Hiroki R. Ueda
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|