1
|
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 2024:10.1038/s41586-024-08132-2. [PMID: 39506111 DOI: 10.1038/s41586-024-08132-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
2
|
Gay SM, Chartampila E, Lord JS, Grizzard S, Maisashvili T, Ye M, Barker NK, Mordant AL, Mills CA, Herring LE, Diering GH. Developing forebrain synapses are uniquely vulnerable to sleep loss. Proc Natl Acad Sci U S A 2024; 121:e2407533121. [PMID: 39441640 PMCID: PMC11536182 DOI: 10.1073/pnas.2407533121] [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: 04/15/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
Sleep is an essential behavior that supports lifelong brain health and cognition. Neuronal synapses are a major target for restorative sleep function and a locus of dysfunction in response to sleep deprivation (SD). Synapse density is highly dynamic during development, becoming stabilized with maturation to adulthood, suggesting sleep exerts distinct synaptic functions between development and adulthood. Importantly, problems with sleep are common in neurodevelopmental disorders including autism spectrum disorder (ASD). Moreover, early life sleep disruption in animal models causes long-lasting changes in adult behavior. Divergent plasticity engaged during sleep necessarily implies that developing and adult synapses will show differential vulnerability to SD. To investigate distinct sleep functions and mechanisms of vulnerability to SD across development, we systematically examined the behavioral and molecular responses to acute SD between juvenile (P21 to P28), adolescent (P42 to P49), and adult (P70 to P100) mice of both sexes. Compared to adults, juveniles lack robust adaptations to SD, precipitating cognitive deficits in the novel object recognition task. Subcellular fractionation, combined with proteome and phosphoproteome analysis revealed the developing synapse is profoundly vulnerable to SD, whereas adults exhibit comparative resilience. SD in juveniles, and not older mice, aberrantly drives induction of synapse potentiation, synaptogenesis, and expression of perineuronal nets. Our analysis further reveals the developing synapse as a putative node of convergence between vulnerability to SD and ASD genetic risk. Together, our systematic analysis supports a distinct developmental function of sleep and reveals how sleep disruption impacts key aspects of brain development, providing insights for ASD susceptibility.
Collapse
Affiliation(s)
- Sean M. Gay
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Elissavet Chartampila
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Julia S. Lord
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Sawyer Grizzard
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Tekla Maisashvili
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Michael Ye
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Natalie K. Barker
- University of North Carolina Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Angie L. Mordant
- University of North Carolina Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - C. Allie Mills
- University of North Carolina Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Laura E. Herring
- University of North Carolina Proteomics Core Facility, Department of Pharmacology, 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
|
3
|
Yang ZH, Cai X, Zhang CY, Zhang Q, Li M, Ding ZL, Guo Y, Ma G, Yang CH, Guo L, Chang H, Wang C, Li M, Xiao X. NEK4 modulates circadian fluctuations of emotional behaviors and synaptogenesis in male mice. Nat Commun 2024; 15:9180. [PMID: 39448584 PMCID: PMC11502819 DOI: 10.1038/s41467-024-53585-8] [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: 03/07/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
GWASs have linked the 3p21.1 locus, which is associated with the expression levels of NEK4, to bipolar disorder. Here, we use integrative analyses of GWAS statistics and eQTL annotations to establish that elevated NEK4 expression in the hippocampus is associated with an increased risk of bipolar disorder. To further study this association, we generate transgenic male mice that conditionally overexpress NEK4 in the pyramidal neurons of the adult forebrain, or use AAV to overexpress NEK4 in the dorsal hippocampus. Compared to the control mice, male mice of both strains exhibit a shift from a diurnal anxiety state to a nocturnal normal or anxiolytic-like state. Overexpression of NEK4 also affects the circadian fluctuations in dendritic spine morphology and synaptic structure. Furthermore, we show that treatment with lithium ameliorates the effects of NEK4 overexpression in male mice. We then perform phosphoproteomic analyses to demonstrate that the diurnal and nocturnal phosphoproteomic profiles of male control and NEK4 overexpressing mice are different. These results suggest that male mice with different NEK4 expression levels may recapitulate some of the core features observed in patients with bipolar disorder, indicating that interruption of the homeostatic dynamics of synapses may underlie the emotional swings in bipolar disorder.
Collapse
Affiliation(s)
- Zhi-Hui Yang
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xin Cai
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chu-Yi Zhang
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Qing Zhang
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Miao Li
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhong-Li Ding
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yingqi Guo
- Institutional Center for Shared Technologies and Facilities of Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Guolan Ma
- Institutional Center for Shared Technologies and Facilities of Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chao-Hao Yang
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Lei Guo
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Hong Chang
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chuang Wang
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Ming Li
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China.
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Xiao Xiao
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China.
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
4
|
Slabeva K, Baud MO. Timing Mechanisms for Circadian Seizures. Clocks Sleep 2024; 6:589-601. [PMID: 39449314 PMCID: PMC11503444 DOI: 10.3390/clockssleep6040040] [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: 07/21/2024] [Revised: 09/17/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024] Open
Abstract
For centuries, epileptic seizures have been noticed to recur with temporal regularity, suggesting that an underlying biological rhythm may play a crucial role in their timing. In this review, we propose to adopt the framework of chronobiology to study the circadian timing of seizures. We first review observations made on seizure timing in patients with epilepsy and animal models of the disorder. We then present the existing chronobiology paradigm to disentangle intertwined circadian and sleep-wake timing mechanisms. In the light of this framework, we review the existing evidence for specific timing mechanisms in specific epilepsy syndromes and highlight that current knowledge is far from sufficient. We propose that individual seizure chronotypes may result from an interplay between independent timing mechanisms. We conclude with a research agenda to help solve the urgency of ticking seizures.
Collapse
Affiliation(s)
- Kristina Slabeva
- Zentrum für Experimentelle Neurologie, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Maxime O. Baud
- Zentrum für Experimentelle Neurologie, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Schlaf-Wach Epilepsie Zentrum, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
5
|
Zhang Y, Chen Y, Li W, Tang L, Li J, Feng X. Targeting the circadian modulation: novel therapeutic approaches in the management of ASD. Front Psychiatry 2024; 15:1451242. [PMID: 39465045 PMCID: PMC11503653 DOI: 10.3389/fpsyt.2024.1451242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/09/2024] [Indexed: 10/29/2024] Open
Abstract
Circadian dysfunction is prevalent in neurodevelopmental disorders, particularly in autism spectrum disorder (ASD). A plethora of empirical studies demonstrate a strong correlation between ASD and circadian disruption, suggesting that modulation of circadian rhythms and the clocks could yield satisfactory advancements. Research indicates that circadian dysfunction associated with abnormal neurodevelopmental phenotypes in ASD individuals, potentially contribute to synapse plasticity disruption. Therefore, targeting circadian rhythms may emerge as a key therapeutic approach. In this study, we did a brief review of the mammalian circadian clock, and the correlation between the circadian mechanism and the pathology of ASD at multiple levels. In addition, we highlight that circadian is the target or modulator to participate in the therapeutic approaches in the management of ASD, such as phototherapy, melatonin, modulating circadian components, natural compounds, and chronotherapies. A deep understanding of the circadian clock's regulatory role in the neurodevelopmental phenotypes in ASD may inspire novel strategies for improving ASD treatment.
Collapse
Affiliation(s)
- Yuxing Zhang
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yinan Chen
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wu Li
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Liya Tang
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jiangshan Li
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiang Feng
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
6
|
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
|
7
|
Tsai YC, ElGrawani W, Muheim C, Spinnler A, Campbell BFN, Lasic D, Hleihil M, Brown SA, Tyagarajan SK. Modulation of sleep/wake patterns by gephyrin phosphorylation status. Eur J Neurosci 2024; 60:5431-5449. [PMID: 39032002 DOI: 10.1111/ejn.16464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/13/2024] [Accepted: 07/01/2024] [Indexed: 07/22/2024]
Abstract
Sleep/wake cycles intricately shape physiological activities including cognitive brain functions, yet the precise molecular orchestrators of sleep remain elusive. Notably, the clinical impact of benzodiazepine drugs underscores the pivotal role of GABAergic neurotransmission in sleep regulation. However, the specific contributions of distinct GABAA receptor subtypes and their principal scaffolding protein, gephyrin, in sleep dynamics remain unclear. The evolving role of synaptic phospho-proteome alterations at excitatory and inhibitory synapses suggests a potential avenue for modulating gephyrin and, consequently, GABAARs for sleep through on-demand kinase recruitment. Our study unveils the distinctive roles of two prevalent GABAA receptor subtypes, α1- and α2-GABAARs, in influencing sleep duration and electrical sleep activity. Notably, the absence of α1-GABAARs emerges as central in sleep regulation, manifesting significant alterations in both non-rapid eye movement (NREM) and rapid eye movement (REM) sleep during dark or active phases, accompanied by altered electroencephalogram (EEG) patterns across various frequencies. Gephyrin proteomics analysis reveals sleep/wake-dependent interactions with a repertoire of known and novel kinases. Crucially, we identify the regulation of gephyrin interaction with ERK1/2, and phosphorylations at serines 268 and 270 are dictated by sleep/wake cycles. Employing AAV-eGFP-gephyrin or its phospho-null variant (S268A/S270A), we disrupt sleep either globally or locally to demonstrate gephyrin phosphorylation as a sleep regulator. In summary, our findings support the local cortical sleep hypothesis and we unveil a molecular mechanism operating at GABAergic synapses, providing critical insights into the intricate regulation of sleep.
Collapse
Affiliation(s)
- Yuan-Chen Tsai
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Waleed ElGrawani
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Christine Muheim
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Andrea Spinnler
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Benjamin F N Campbell
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Denis Lasic
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Mohammad Hleihil
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Steven A Brown
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Shiva K Tyagarajan
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Centre Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
Sarrazin DH, Gardner W, Marchese C, Balzinger M, Ramanathan C, Schott M, Rozov S, Veleanu M, Vestring S, Normann C, Rantamäki T, Antoine B, Barrot M, Challet E, Bourgin P, Serchov T. Prefrontal cortex molecular clock modulates development of depression-like phenotype and rapid antidepressant response in mice. Nat Commun 2024; 15:7257. [PMID: 39179578 PMCID: PMC11344080 DOI: 10.1038/s41467-024-51716-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 08/13/2024] [Indexed: 08/26/2024] Open
Abstract
Depression is associated with dysregulated circadian rhythms, but the role of intrinsic clocks in mood-controlling brain regions remains poorly understood. We found increased circadian negative loop and decreased positive clock regulators expression in the medial prefrontal cortex (mPFC) of a mouse model of depression, and a subsequent clock countermodulation by the rapid antidepressant ketamine. Selective Bmal1KO in CaMK2a excitatory neurons revealed that the functional mPFC clock is an essential factor for the development of a depression-like phenotype and ketamine effects. Per2 silencing in mPFC produced antidepressant-like effects, while REV-ERB agonism enhanced the depression-like phenotype and suppressed ketamine action. Pharmacological potentiation of clock positive modulator ROR elicited antidepressant-like effects, upregulating plasticity protein Homer1a, synaptic AMPA receptors expression and plasticity-related slow wave activity specifically in the mPFC. Our data demonstrate a critical role for mPFC molecular clock in regulating depression-like behavior and the therapeutic potential of clock pharmacological manipulations influencing glutamatergic-dependent plasticity.
Collapse
Affiliation(s)
- David H Sarrazin
- Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, Institute of Cellular and Integrative Neurosciences (INCI) UPR 3212, Strasbourg, France
| | - Wilf Gardner
- Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, Institute of Cellular and Integrative Neurosciences (INCI) UPR 3212, Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), University of Strasbourg, Strasbourg, France
| | - Carole Marchese
- Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, Institute of Cellular and Integrative Neurosciences (INCI) UPR 3212, Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), University of Strasbourg, Strasbourg, France
| | - Martin Balzinger
- Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, Institute of Cellular and Integrative Neurosciences (INCI) UPR 3212, Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), University of Strasbourg, Strasbourg, France
| | | | - Marion Schott
- Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, Institute of Cellular and Integrative Neurosciences (INCI) UPR 3212, Strasbourg, France
| | - Stanislav Rozov
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Maxime Veleanu
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Vestring
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Berta-Ottenstein-Programme for Clinician Scientists, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Claus Normann
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Neuromodulation, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tomi Rantamäki
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Benedicte Antoine
- Sorbonne Université, INSERM, Centre de Recherches St-Antoine (CRSA), Paris, France
| | - Michel Barrot
- Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, Institute of Cellular and Integrative Neurosciences (INCI) UPR 3212, Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), University of Strasbourg, Strasbourg, France
| | - Etienne Challet
- Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, Institute of Cellular and Integrative Neurosciences (INCI) UPR 3212, Strasbourg, France
| | - Patrice Bourgin
- Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, Institute of Cellular and Integrative Neurosciences (INCI) UPR 3212, Strasbourg, France
- CIRCSom (International Research Center for ChronoSomnology) & Sleep Disorders Center, Strasbourg University Hospital, Strasbourg, France
| | - Tsvetan Serchov
- Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, Institute of Cellular and Integrative Neurosciences (INCI) UPR 3212, Strasbourg, France.
- University of Strasbourg Institute for Advanced Study (USIAS), University of Strasbourg, Strasbourg, France.
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
9
|
Liu J, Niethard N, Lun Y, Dimitrov S, Ehrlich I, Born J, Hallschmid M. Slow-wave sleep drives sleep-dependent renormalization of synaptic AMPA receptor levels in the hypothalamus. PLoS Biol 2024; 22:e3002768. [PMID: 39163472 PMCID: PMC11364421 DOI: 10.1371/journal.pbio.3002768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/30/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
According to the synaptic homeostasis hypothesis (SHY), sleep serves to renormalize synaptic connections that have been potentiated during the prior wake phase due to ongoing encoding of information. SHY focuses on glutamatergic synaptic strength and has been supported by numerous studies examining synaptic structure and function in neocortical and hippocampal networks. However, it is unknown whether synaptic down-regulation during sleep occurs in the hypothalamus, i.e., a pivotal center of homeostatic regulation of bodily functions including sleep itself. We show that sleep, in parallel with the synaptic down-regulation in neocortical networks, down-regulates the levels of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) in the hypothalamus of rats. Most robust decreases after sleep were observed at both sites for AMPARs containing the GluA1 subunit. Comparing the effects of selective rapid eye movement (REM) sleep and total sleep deprivation, we moreover provide experimental evidence that slow-wave sleep (SWS) is the driving force of the down-regulation of AMPARs in hypothalamus and neocortex, with no additional contributions of REM sleep or the circadian rhythm. SWS-dependent synaptic down-regulation was not linked to EEG slow-wave activity. However, spindle density during SWS predicted relatively increased GluA1 subunit levels in hypothalamic synapses, which is consistent with the role of spindles in the consolidation of memory. Our findings identify SWS as the main driver of the renormalization of synaptic strength during sleep and suggest that SWS-dependent synaptic renormalization is also implicated in homeostatic control processes in the hypothalamus.
Collapse
Affiliation(s)
- Jianfeng Liu
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
| | - Niels Niethard
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
| | - Yu Lun
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
| | - Stoyan Dimitrov
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
| | - Ingrid Ehrlich
- Department of Neurobiology, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany
| | - Jan Born
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
- Center for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University Tübingen (IDM), Tübingen, Germany
- German Center for Mental Health (DZPG), Tübingen, Germany
| | - Manfred Hallschmid
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University Tübingen (IDM), Tübingen, Germany
- German Center for Mental Health (DZPG), Tübingen, Germany
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Liu S, Zhuo K, Wang Y, Wang X, Zhao Y. Prolonged Sleep Deprivation Induces a Reprogramming of Circadian Rhythmicity with the Hepatic Metabolic Transcriptomic Profile. BIOLOGY 2024; 13:532. [PMID: 39056724 PMCID: PMC11274269 DOI: 10.3390/biology13070532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Sleep disturbances can disrupt the overall circadian rhythm. However, the impact of sleep deprivation on the circadian rhythm of the liver and its underlying mechanisms still requires further exploration. In this study, we subjected male mice to 5 days of sleep deprivation and performed liver transcriptome sequencing analysis at various time points within a 24-h period. Subsequently, we monitored the autonomic activity and food intake in these male mice for six days post-sleep deprivation. We observed alterations in sleep-wake and feeding rhythms in the first two days following sleep deprivation. Additionally, we also observed a decrease in 24-h serum-glucose levels. Liver transcriptome sequencing has shown that sleep deprivation induces the rhythmic transcription of a large number of genes, or alters the rhythmic properties of genes, which were then significantly enriched in the carbohydrate, lipid, and protein metabolism pathways. Our findings suggest that under conditions of prolonged sleep deprivation, the expression of metabolic-related genes in the liver was reset, leading to changes in the organism's metabolic state to ensure energy supply to sustain prolonged wakefulness.
Collapse
Affiliation(s)
| | | | | | | | - Yingying Zhao
- Department of Physiology, School of Basic Medical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China; (S.L.); (K.Z.); (Y.W.); (X.W.)
| |
Collapse
|
13
|
Squarcio F, Tononi G, Cirelli C. Effects of non-rapid eye movement sleep on the cortical synaptic expression of GluA1-containing AMPA receptors. Eur J Neurosci 2024; 60:3961-3972. [PMID: 38973508 DOI: 10.1111/ejn.16460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/11/2024] [Accepted: 06/19/2024] [Indexed: 07/09/2024]
Abstract
Converging electrophysiological, molecular and ultrastructural evidence supports the hypothesis that sleep promotes a net decrease in excitatory synaptic strength, counteracting the net synaptic potentiation caused by ongoing learning during waking. However, several outstanding questions about sleep-dependent synaptic weakening remain. Here, we address some of these questions by using two established molecular markers of synaptic strength, the levels of the AMPA (alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid) receptors containing the GluA1 subunit and the phosphorylation of GluA1 at serine 845 (p-GluA1(845)). We previously found that, in the rat cortex and hippocampus, these markers are lower after 6-8 h of sleep than after the same time spent awake. Here, we measure GluA1 and p-GluA1(845) levels in synaptosomes of mouse cortex after 5 h of either sleep, sleep deprivation, recovery sleep after sleep deprivation or selective REM sleep deprivation (32 C57BL/B6 adult mice, 16 females). We find that relative to after sleep deprivation, these synaptic markers are lower after sleep independent of whether the mice were allowed to enter REM sleep. Moreover, 5 h of recovery sleep following acute sleep deprivation is enough to renormalize their expression. Thus, the renormalization of GluA1 and p-GluA1(845) expression crucially relies on NREM sleep and can occur in a few hours of sleep after acute sleep deprivation.
Collapse
Affiliation(s)
- Fabio Squarcio
- Department of Psychiatry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Giulio Tononi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
14
|
Chen R, Nie P, Wang J, Wang GZ. Deciphering brain cellular and behavioral mechanisms: Insights from single-cell and spatial RNA sequencing. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1865. [PMID: 38972934 DOI: 10.1002/wrna.1865] [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: 01/31/2024] [Revised: 05/05/2024] [Accepted: 05/14/2024] [Indexed: 07/09/2024]
Abstract
The brain is a complex computing system composed of a multitude of interacting neurons. The computational outputs of this system determine the behavior and perception of every individual. Each brain cell expresses thousands of genes that dictate the cell's function and physiological properties. Therefore, deciphering the molecular expression of each cell is of great significance for understanding its characteristics and role in brain function. Additionally, the positional information of each cell can provide crucial insights into their involvement in local brain circuits. In this review, we briefly overview the principles of single-cell RNA sequencing and spatial transcriptomics, the potential issues and challenges in their data processing, and their applications in brain research. We further outline several promising directions in neuroscience that could be integrated with single-cell RNA sequencing, including neurodevelopment, the identification of novel brain microstructures, cognition and behavior, neuronal cell positioning, molecules and cells related to advanced brain functions, sleep-wake cycles/circadian rhythms, and computational modeling of brain function. We believe that the deep integration of these directions with single-cell and spatial RNA sequencing can contribute significantly to understanding the roles of individual cells or cell types in these specific functions, thereby making important contributions to addressing critical questions in those fields. This article is categorized under: RNA Evolution and Genomics > Computational Analyses of RNA RNA in Disease and Development > RNA in Development RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Renrui Chen
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Pengxing Nie
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jing Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guang-Zhong Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
15
|
Zhao H, Zhang T, Zhang H, Wang Y, Cheng L. Exercise-with-melatonin therapy improves sleep disorder and motor dysfunction in a rat model of ischemic stroke. Neural Regen Res 2024; 19:1336-1343. [PMID: 37905883 PMCID: PMC11467917 DOI: 10.4103/1673-5374.385844] [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: 02/19/2023] [Revised: 05/15/2023] [Accepted: 08/15/2023] [Indexed: 11/02/2023] Open
Abstract
Exercise-with-melatonin therapy has complementary and synergistic effects on spinal cord injury and Alzheimer’s disease, but its effect on stroke is still poorly understood. In this study, we established a rat model of ischemic stroke by occluding the middle cerebral artery for 60 minutes. We treated the rats with exercise and melatonin therapy for 7 consecutive days. Results showed that exercise-with-melatonin therapy significantly prolonged sleep duration in the model rats, increased delta power values, and regularized delta power rhythm. Additionally, exercise-with-melatonin therapy improved coordination, endurance, and grip strength, as well as learning and memory abilities. At the same time, it led to higher hippocampal CA1 neuron activity and postsynaptic density thickness and lower expression of glutamate receptor 2 than did exercise or melatonin therapy alone. These findings suggest that exercise-with-melatonin therapy can alleviate sleep disorder and motor dysfunction by increasing glutamate receptor 2 protein expression and regulating hippocampal CA1 synaptic plasticity.
Collapse
Affiliation(s)
- Haitao Zhao
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Neurological Rehabilitation, Beijing Bo’ai Hospital, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, University of Health and Rehabilitation Sciences, Qingdao, Shandong Province, China
- Laboratory of Brain Injury Repair and Rehabilitation, China Rehabilitation Science Institute, Beijing, China
| | - Tong Zhang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Neurological Rehabilitation, Beijing Bo’ai Hospital, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, University of Health and Rehabilitation Sciences, Qingdao, Shandong Province, China
- Laboratory of Brain Injury Repair and Rehabilitation, China Rehabilitation Science Institute, Beijing, China
| | - Haojie Zhang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Neurological Rehabilitation, Beijing Bo’ai Hospital, China Rehabilitation Research Center, Beijing, China
- Laboratory of Brain Injury Repair and Rehabilitation, China Rehabilitation Science Institute, Beijing, China
| | - Yunlei Wang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Neurological Rehabilitation, Beijing Bo’ai Hospital, China Rehabilitation Research Center, Beijing, China
- Laboratory of Brain Injury Repair and Rehabilitation, China Rehabilitation Science Institute, Beijing, China
| | - Lingna Cheng
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Neurological Rehabilitation, Beijing Bo’ai Hospital, China Rehabilitation Research Center, Beijing, China
- Laboratory of Brain Injury Repair and Rehabilitation, China Rehabilitation Science Institute, Beijing, China
| |
Collapse
|
16
|
Martin SC, Joyce KK, Lord JS, Harper KM, Nikolova VD, Cohen TJ, Moy SS, Diering GH. Sleep Disruption Precedes Forebrain Synaptic Tau Burden and Contributes to Cognitive Decline in a Sex-Dependent Manner in the P301S Tau Transgenic Mouse Model. eNeuro 2024; 11:ENEURO.0004-24.2024. [PMID: 38858068 PMCID: PMC11209651 DOI: 10.1523/eneuro.0004-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/02/2024] [Accepted: 05/14/2024] [Indexed: 06/12/2024] Open
Abstract
Sleep disruption and impaired synaptic processes are common features in neurodegenerative diseases, including Alzheimer's disease (AD). Hyperphosphorylated Tau is known to accumulate at neuronal synapses in AD, contributing to synapse dysfunction. However, it remains unclear how sleep disruption and synapse pathology interact to contribute to cognitive decline. Here, we examined sex-specific onset and consequences of sleep loss in AD/tauopathy model PS19 mice. Using a piezoelectric home-cage monitoring system, we showed PS19 mice exhibited early-onset and progressive hyperarousal, a selective dark-phase sleep disruption, apparent at 3 months in females and 6 months in males. Using the Morris water maze test, we report that chronic sleep disruption (CSD) accelerated the onset of decline of hippocampal spatial memory in PS19 males only. Hyperarousal occurs well in advance of robust forebrain synaptic Tau burden that becomes apparent at 6-9 months. To determine whether a causal link exists between sleep disruption and synaptic Tau hyperphosphorylation, we examined the correlation between sleep behavior and synaptic Tau, or exposed mice to acute or chronic sleep disruption at 6 months. While we confirm that sleep disruption is a driver of Tau hyperphosphorylation in neurons of the locus ceruleus, we were unable to show any causal link between sleep loss and Tau burden in forebrain synapses. Despite the finding that hyperarousal appears earlier in females, female cognition was resilient to the effects of sleep disruption. We conclude sleep disruption interacts with the synaptic Tau burden to accelerate the onset of cognitive decline with greater vulnerability in males.
Collapse
Affiliation(s)
- Shenée C Martin
- Departments of Cell Biology and Physiology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Kathryn K Joyce
- Departments of Cell Biology and Physiology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Julia S Lord
- Departments of Cell Biology and Physiology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Kathryn M Harper
- Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Viktoriya D Nikolova
- Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Todd J Cohen
- Neurology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Sheryl S Moy
- Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
- Carolina Institute for Developmental Disabilities, Carrboro, North Carolina 27510
| | - Graham H Diering
- Departments of Cell Biology and Physiology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
- Carolina Institute for Developmental Disabilities, Carrboro, North Carolina 27510
| |
Collapse
|
17
|
Carvalho Cabral P, Richard VR, Borchers CH, Olivier M, Cermakian N. Circadian Control of the Response of Macrophages to Plasmodium Spp.-Infected Red Blood Cells. Immunohorizons 2024; 8:442-456. [PMID: 38916585 PMCID: PMC11220744 DOI: 10.4049/immunohorizons.2400021] [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: 03/13/2024] [Accepted: 05/23/2024] [Indexed: 06/26/2024] Open
Abstract
Malaria is a serious vector-borne disease characterized by periodic episodes of high fever and strong immune responses that are coordinated with the daily synchronized parasite replication cycle inside RBCs. As immune cells harbor an autonomous circadian clock that controls various aspects of the immune response, we sought to determine whether the intensity of the immune response to Plasmodium spp., the parasite causing malaria, depends on time of infection. To do this, we developed a culture model in which mouse bone marrow-derived macrophages are stimulated with RBCs infected with Plasmodium berghei ANKA (iRBCs). Lysed iRBCs, but not intact iRBCs or uninfected RBCs, triggered an inflammatory immune response in bone marrow-derived macrophages. By stimulating at four different circadian time points (16, 22, 28, or 34 h postsynchronization of the cells' clock), 24-h rhythms in reactive oxygen species and cytokines/chemokines were found. Furthermore, the analysis of the macrophage proteome and phosphoproteome revealed global changes in response to iRBCs that varied according to circadian time. This included many proteins and signaling pathways known to be involved in the response to Plasmodium infection. In summary, our findings show that the circadian clock within macrophages determines the magnitude of the inflammatory response upon stimulation with ruptured iRBCs, along with changes of the cell proteome and phosphoproteome.
Collapse
Affiliation(s)
| | - Vincent R. Richard
- Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| | - Christoph H. Borchers
- Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| | - Martin Olivier
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Nicolas Cermakian
- Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
18
|
Coulson RL, Mourrain P, Wang GX. The intersection of sleep and synaptic translation in synaptic plasticity deficits in neurodevelopmental disorders. J Comp Physiol B 2024; 194:253-263. [PMID: 38396062 PMCID: PMC11233386 DOI: 10.1007/s00360-023-01531-3] [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: 08/28/2023] [Revised: 11/29/2023] [Accepted: 12/19/2023] [Indexed: 02/25/2024]
Abstract
Individuals with neurodevelopmental disorders experience persistent sleep deficits, and there is increasing evidence that sleep dysregulation is an underlying cause, rather than merely an effect, of the synaptic and behavioral defects observed in these disorders. At the molecular level, dysregulation of the synaptic proteome is a common feature of neurodevelopmental disorders, though the mechanism connecting these molecular and behavioral phenotypes is an ongoing area of investigation. A role for eIF2α in shifting the local proteome in response to changes in the conditions at the synapse has emerged. Here, we discuss recent progress in characterizing the intersection of local synaptic translation and sleep and propose a reciprocal mechanism of dysregulation in the development of synaptic plasticity defects in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Rochelle L Coulson
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Philippe Mourrain
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- INSERM 1024, Ecole Normale Supérieure, Paris, France
| | - Gordon X Wang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
19
|
Zhang Y, Wang Y, Wei R, Li X, Luo B, Zhang J, Zhang K, Fang S, Liu X, Chen G. Mitochondrial antioxidant elamipretide improves learning and memory impairment induced by chronic sleep deprivation in mice. Brain Behav 2024; 14:e3508. [PMID: 38688894 PMCID: PMC11061203 DOI: 10.1002/brb3.3508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/10/2024] [Accepted: 03/16/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND The inflammation and synaptic dysfunction induced by mitochondrial dysfunction play essential roles in the learning and memory impairment associated with sleep dysfunction. Elamipretide (SS-31), a novel mitochondrion-targeted antioxidant, was proven to improve mitochondrial dysfunction, the inflammatory response, synaptic dysfunction, and cognitive impairment in models of cerebral ischemia, sepsis, and type 2 diabetes. However, the potential for SS-31 to improve the cognitive impairment induced by chronic sleep deprivation (CSD) and its underlying mechanisms is unknown. METHODS Adult c57BL/6J mice were subjected to CSD for 21 days using an activity wheel accompanied by daily intraperitoneal injection of SS-31 (5 mg/kg). The novel object recognition and Morris water maze test were used to evaluate hippocampus-dependent cognitive function. Western blotting and reverse transcription-quantitative polymerase chain reaction assays were used to determine the effects of CSD and SS-31 on markers of mitochondria, inflammation response, and synaptic function. Enzyme-linked immunosorbent assays were used to examine the levels of proinflammatory cytokines. RESULTS SS-31 could improve the cognitive impairment induced by CSD. In particular, SS-31 treatment restored the CSD-induced decrease in sirtuin 1 (SIRT1) and peroxisome proliferator-activated receptor γ coactivator alpha levels and the increase in levels nuclear factor kappa-B and inflammatory cytokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor-alpha. Furthermore, SS-31 significantly increased the levels of brain-derived neurotrophic factor, postsynaptic density protein-95, and synaptophysin in CSD mice. CONCLUSION Taken together, these results suggest that SS-31 could improve CSD-induced mitochondrial biogenesis dysfunction, inflammatory response, synaptic dysfunction, and cognitive impairment by increasing SIRT1 expression levels.
Collapse
Affiliation(s)
- Yue‐Ming Zhang
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Ya‐Tao Wang
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Ru‐Meng Wei
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Xue‐Yan Li
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Bao‐Ling Luo
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Jing‐Ya Zhang
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Kai‐Xuan Zhang
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Shi‐Kun Fang
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Xue‐Chun Liu
- Department of NeurologyThe Second People's Hospital of Hefei and Affiliated Hefei Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Gui‐Hai Chen
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| |
Collapse
|
20
|
Chiu JC. Editorial: Rising stars in chronobiology 2022. Front Physiol 2024; 15:1412956. [PMID: 38725565 PMCID: PMC11079284 DOI: 10.3389/fphys.2024.1412956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 04/11/2024] [Indexed: 05/12/2024] Open
Affiliation(s)
- Joanna C. Chiu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California, Davis, CA, United States
| |
Collapse
|
21
|
Sun SED, Levenstein D, Li B, Mandelberg N, Chenouard N, Suutari BS, Sanchez S, Tian G, Rinzel J, Buzsáki G, Tsien RW. Synaptic homeostasis transiently leverages Hebbian mechanisms for a multiphasic response to inactivity. Cell Rep 2024; 43:113839. [PMID: 38507409 DOI: 10.1016/j.celrep.2024.113839] [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: 09/13/2022] [Revised: 12/19/2023] [Accepted: 02/05/2024] [Indexed: 03/22/2024] Open
Abstract
Homeostatic regulation of synapses is vital for nervous system function and key to understanding a range of neurological conditions. Synaptic homeostasis is proposed to operate over hours to counteract the destabilizing influence of long-term potentiation (LTP) and long-term depression (LTD). The prevailing view holds that synaptic scaling is a slow first-order process that regulates postsynaptic glutamate receptors and fundamentally differs from LTP or LTD. Surprisingly, we find that the dynamics of scaling induced by neuronal inactivity are not exponential or monotonic, and the mechanism requires calcineurin and CaMKII, molecules dominant in LTD and LTP. Our quantitative model of these enzymes reconstructs the unexpected dynamics of homeostatic scaling and reveals how synapses can efficiently safeguard future capacity for synaptic plasticity. This mechanism of synaptic adaptation supports a broader set of homeostatic changes, including action potential autoregulation, and invites further inquiry into how such a mechanism varies in health and disease.
Collapse
Affiliation(s)
- Simón E D Sun
- Center for Neural Science, New York University, New York, NY 10003, USA; Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA; Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Daniel Levenstein
- Center for Neural Science, New York University, New York, NY 10003, USA; Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA; Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, 3810 University Street, Montreal, QC, Canada
| | - Boxing Li
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA; Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510810, China
| | - Nataniel Mandelberg
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA
| | - Nicolas Chenouard
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA; Sorbonne Université, INSERM U1127, UMR CNRS 7225, Institut du Cerveau (ICM), 47 bld de l'hôpital, 75013 Paris, France
| | - Benjamin S Suutari
- Center for Neural Science, New York University, New York, NY 10003, USA; Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA
| | - Sandrine Sanchez
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA
| | - Guoling Tian
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA
| | - John Rinzel
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - György Buzsáki
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA
| | - Richard W Tsien
- Center for Neural Science, New York University, New York, NY 10003, USA; Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
22
|
Gay SM, Chartampila E, Lord JS, Grizzard S, Maisashvili T, Ye M, Barker NK, Mordant AL, Mills CA, Herring LE, Diering GH. Developing forebrain synapses are uniquely vulnerable to sleep loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.06.565853. [PMID: 37986967 PMCID: PMC10659326 DOI: 10.1101/2023.11.06.565853] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Sleep is an essential behavior that supports lifelong brain health and cognition. Neuronal synapses are a major target for restorative sleep function and a locus of dysfunction in response to sleep deprivation (SD). Synapse density is highly dynamic during development, becoming stabilized with maturation to adulthood, suggesting sleep exerts distinct synaptic functions between development and adulthood. Importantly, problems with sleep are common in neurodevelopmental disorders including autism spectrum disorder (ASD). Moreover, early life sleep disruption in animal models causes long lasting changes in adult behavior. Different plasticity engaged during sleep necessarily implies that developing and adult synapses will show differential vulnerability to SD. To investigate distinct sleep functions and mechanisms of vulnerability to SD across development, we systematically examined the behavioral and molecular responses to acute SD between juvenile (P21-28), adolescent (P42-49) and adult (P70-100) mice of both sexes. Compared to adults, juveniles lack robust adaptations to SD, precipitating cognitive deficits in the novel object recognition test. Subcellular fractionation, combined with proteome and phosphoproteome analysis revealed the developing synapse is profoundly vulnerable to SD, whereas adults exhibit comparative resilience. SD in juveniles, and not older mice, aberrantly drives induction of synapse potentiation, synaptogenesis, and expression of peri-neuronal nets. Our analysis further reveals the developing synapse as a convergent node between vulnerability to SD and ASD genetic risk. Together, our systematic analysis supports a distinct developmental function of sleep and reveals how sleep disruption impacts key aspects of brain development, providing mechanistic insights for ASD susceptibility.
Collapse
|
23
|
Chen Y, Zhang L, Yin H. Different emotion regulation strategies mediate the relations of corresponding connections within the default-mode network to sleep quality. Brain Imaging Behav 2024; 18:302-314. [PMID: 38057650 DOI: 10.1007/s11682-023-00828-9] [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] [Accepted: 11/19/2023] [Indexed: 12/08/2023]
Abstract
Despite a long history of interest in the relation of emotion regulation to sleep quality, how different strategies link with sleep quality at the neural level is still poorly understood. Thus, we utilized the process model of emotion regulation as an organizing framework for examining the neurological underpinning of the links between the two emotion regulation strategies and sleep quality. 183 young adults (51.7% females, Mage = 22.16) were guided to undergo the MRI scans and then complete the Pittsburgh Sleep Quality Index (PSQI) and the emotion regulation Questionnaire (ERQ) formed by two dimensions: cognitive reappraisal and expressive suppression. Results found that emotion regulation mediated the association between functional connectivity within the intrinsic default-mode network (DMN) and sleep quality. Specifically, rsFC analysis showed that cognitive reappraisal was positively correlated with rsFC within DMN, including left superior temporal gyrus (lSTG)-left lateral occipital cortex (lLOC), lSTG-left anterior cingulate gyrus (lACG), right lateral occipital cortex (rLOC)-left middle frontal gyrus (lMFG), and rLOC-lSTG. Further mediation analysis indicated a mediated role of cognitive reappraisal in the links between the four connectivity within the DMN and sleep quality. In addition, expressive suppression was positively correlated with rsFC within DMN, including left precuneus cortex (lPrcu)-right Temporal Pole (rTP) and lPrcu- lSTG. Further mediation analysis indicated a mediated role of expressive suppression in the links between the two connectivity within the DMN and sleep quality. Overall, this finding supports the process model of emotion regulation in that the effects of reappraisal and suppression have varying neural circuits that impact that strategy's effect on sleep quality.
Collapse
Affiliation(s)
- Yang Chen
- Department of Psychology, School of Education Science, Hunan Normal University, 36 Lushan Road, Changsha, Hunan, 410081, China
- Centre for Mind & Brain Science, Hunan Normal University, Changsha, China
| | - Li Zhang
- Department of Psychology, School of Education Science, Hunan Normal University, 36 Lushan Road, Changsha, Hunan, 410081, China
- Centre for Mind & Brain Science, Hunan Normal University, Changsha, China
| | - Huazhan Yin
- Department of Psychology, School of Education Science, Hunan Normal University, 36 Lushan Road, Changsha, Hunan, 410081, China.
- Centre for Mind & Brain Science, Hunan Normal University, Changsha, China.
| |
Collapse
|
24
|
Ling Z, Zhou S, Zhou Y, Zhong W, Su Z, Qin Z. Protective role of madecassoside from Centella asiatica against protein L-isoaspartyl methyltransferase deficiency-induced neurodegeneration. Neuropharmacology 2024; 246:109834. [PMID: 38181970 DOI: 10.1016/j.neuropharm.2023.109834] [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: 11/25/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/07/2024]
Abstract
Protein L-isoaspartyl methyltransferase (PIMT/PCMT1) could repair l-isoaspartate (L-isoAsp) residues formed by deamidation of asparaginyl (Asn) residues or isomerization of aspartyl (Asp) residues in peptides and proteins during aging. Aside from abnormal accumulation of L-isoAsp, PIMT knockout (KO) mice mirrors some neuropathological hallmarks such as anxiety-like behaviors, impaired spatial memory and aberrant synaptic plasticity in the hippocampus of neurodegenerative diseases (NDs), including Alzheimer's disease (AD) and related dementias, and Parkinson's disease (PD). While some reports indicate the neuroprotective effect of madecassoside (MA) as a triterpenoid saponin component of Centella asiatica, its role against NDs-related anxiety and cognitive impairment remains unclear. Therefore, we investigated the effect of MA against anxiety-related behaviors in PIMT deficiency-induced mouse model of NDs. Results obtained from the elevated plus maze (EPM) test revealed that MA treatment alleviated anxiety-like behaviors in PIMT knockout mice. Furthermore, Real-time PCR, electroencephalogram (EEG) recordings, transmission electron microscopy analysis and ELISA were carried out to evaluate the expression of clock genes, sleep and synaptic function, respectively. The PIMT knockout mice were characterized by abnormal clock patterns, sleep disturbance and synaptic dysfunction, which could be improved by MA administration. Collectively, these findings suggest that MA exhibits neuroprotective effects associated with improved circadian rhythms sleep-wake cycle and synaptic plasticity in PIMT deficient mice, which could be translated to ameliorate anxiety-related symptoms and cognitive impairments in NDs.
Collapse
Affiliation(s)
- Zicheng Ling
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sirui Zhou
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yancheng Zhou
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wanyu Zhong
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhonghao Su
- Department of Febrile Disease, School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zhenxia Qin
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
25
|
Li W, Tiedt S, Lawrence JH, Harrington ME, Musiek ES, Lo EH. Circadian Biology and the Neurovascular Unit. Circ Res 2024; 134:748-769. [PMID: 38484026 DOI: 10.1161/circresaha.124.323514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/14/2024] [Indexed: 03/19/2024]
Abstract
Mammalian physiology and cellular function are subject to significant oscillations over the course of every 24-hour day. It is likely that these daily rhythms will affect function as well as mechanisms of disease in the central nervous system. In this review, we attempt to survey and synthesize emerging studies that investigate how circadian biology may influence the neurovascular unit. We examine how circadian clocks may operate in neural, glial, and vascular compartments, review how circadian mechanisms regulate cell-cell signaling, assess interactions with aging and vascular comorbidities, and finally ask whether and how circadian effects and disruptions in rhythms may influence the risk and progression of pathophysiology in cerebrovascular disease. Overcoming identified challenges and leveraging opportunities for future research might support the development of novel circadian-based treatments for stroke.
Collapse
Affiliation(s)
- Wenlu Li
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (W.L., E.H.L.)
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
| | - Steffen Tiedt
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany (S.T.)
| | - Jennifer H Lawrence
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
- Department of Neurology, Washington University School of Medicine, St. Louis, MO (J.H.L., E.S.M.)
| | - Mary E Harrington
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
- Neuroscience Program, Smith College, Northampton, MA (M.E.H.)
| | - Erik S Musiek
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
- Department of Neurology, Washington University School of Medicine, St. Louis, MO (J.H.L., E.S.M.)
| | - Eng H Lo
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (W.L., E.H.L.)
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
| |
Collapse
|
26
|
Silverman D, Chen C, Chang S, Bui L, Zhang Y, Raghavan R, Jiang A, Darmohray D, Sima J, Ding X, Li B, Ma C, Dan Y. Activation of locus coeruleus noradrenergic neurons rapidly drives homeostatic sleep pressure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582852. [PMID: 38496507 PMCID: PMC10942400 DOI: 10.1101/2024.02.29.582852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Homeostatic sleep regulation is essential for optimizing the amount and timing of sleep, but the underlying mechanism remains unclear. Optogenetic activation of locus coeruleus noradrenergic neurons immediately increased sleep propensity following transient wakefulness. Fiber photometry showed that repeated optogenetic or sensory stimulation caused rapid declines of locus coeruleus calcium activity and noradrenaline release. This suggests that functional fatigue of noradrenergic neurons, which reduces their wake-promoting capacity, contributes to sleep pressure.
Collapse
|
27
|
Mohamad Sobri WBW, Naing NN, Wan-Arfah N, Abdullah S, Subramaniyan V, Wong LS, Selvaraj S. Prevalence and factors associated with excessive daytime sleepiness among Malaysian medical students. ELECTRONIC JOURNAL OF GENERAL MEDICINE 2024; 21:em571. [DOI: 10.29333/ejgm/14198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
The purpose of our study was to ascertain the frequency of excessive daytime sleepiness (EDS) amongst medical students as well as the contributing variables. In Kelantan, Malaysia, at the School of Medical Sciences of Universiti Sains Malaysia, a cross-sectional research study was carried out. The Epworth drowsiness scale was used to gauge the degree of daytime drowsiness, and the depression, anxiety, and stress scale was used to gauge the degree of depression, anxiety, and stress. The related factors of EDS were analyzed using ordinal logistic regression. 311 individuals, or 84.5% of the total, submitted the questionnaire out of 368. 42.7% of people had EDS (95% CI: 0.52, 0.70). The associated factors of EDS included year of study (adjusted odds ratio [AOR]=0.55 [95% confidence interval [CI]: 0.33, 0.91]), race (Chinese) (AOR=0.58 [95% CI: 0.32, 0.97]), anxiety level (mild) (AOR=2.68 [95% CI: 1.26, 5.68]), anxiety level (moderate) (AOR=3.70 [95% CI: 1.76, 7.75]), anxiety level (severe) (AOR=4.76 [95% CI: 1.06, 21.42]), stress level (mild) (AOR=3.37 [95% CI: 1.47, 7.30]) and stress level (moderate) (AOR=5.42 [95% CI: 2.05, 14.35]). As for conclusion, associated factors such as year of study, race, anxiety and stress level were significantly found to be associated with EDS among medical students.<b> </b>
Collapse
Affiliation(s)
| | - Nyi Nyi Naing
- Faculty of Medicine, Universiti Sultan Zainal Abidin, Kuala Terengganu, Terengganu, MALAYSIA
| | - Nadiah Wan-Arfah
- Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Kuala Terengganu, Terengganu, MALAYSIA
| | - Sarimah Abdullah
- Unit of Biostatistics and Research Methodology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, MALAYSIA
| | - Vetriselvan Subramaniyan
- Department of Pharmacology, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Subang Jaya, Selangor, MALAYSIA
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, INDIA
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai, Negeri Sembilan, MALAYSIA
| | | |
Collapse
|
28
|
Comas M, De Pietri Tonelli D, Berdondini L, Astiz M. Ontogeny of the circadian system: a multiscale process throughout development. Trends Neurosci 2024; 47:36-46. [PMID: 38071123 DOI: 10.1016/j.tins.2023.11.004] [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: 07/23/2023] [Revised: 10/02/2023] [Accepted: 11/12/2023] [Indexed: 01/12/2024]
Abstract
The 24 h (circadian) timing system develops in mammals during the perinatal period. It carries out the essential task of anticipating daily recurring environmental changes to identify the best time of day for each molecular, cellular, and systemic process. Although significant knowledge has been acquired about the organization and function of the adult circadian system, relatively little is known about its ontogeny. During the perinatal period, the circadian system progressively gains functionality under the influence of the early environment. This review explores current evidence on the development of the circadian clock in mammals, highlighting the multilevel complexity of the process and the importance of gaining a better understanding of its underlying biology.
Collapse
Affiliation(s)
- Maria Comas
- Circadian Physiology of Neurons and Glia Laboratory, Achucarro Basque Center for Neuroscience, 48940 Leioa, Basque Country, Spain
| | | | - Luca Berdondini
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy
| | - Mariana Astiz
- Circadian Physiology of Neurons and Glia Laboratory, Achucarro Basque Center for Neuroscience, 48940 Leioa, Basque Country, Spain; Ikerbasque - Basque Foundation for Science, Bilbao, Spain; Institute of Neurobiology, University of Lübeck, 23562 Lübeck, Germany.
| |
Collapse
|
29
|
Chen Z, Li Y, He K, Yang J, Deng Q, Chen Y, Fu Z. CircGPRC5A enhances colorectal cancer progress by stabilizing PPP1CA and inducing YAP dephosphorylation. J Exp Clin Cancer Res 2023; 42:334. [PMID: 38057879 PMCID: PMC10698990 DOI: 10.1186/s13046-023-02915-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/23/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND With the advancements in bioinformatic technology, an increasing number of circular RNAs (circRNAs) have been discovered and their crucial roles in the development and progression of various malignancies have been confirmed through multiple pathways. However, the specific mechanisms involving protein-binding circRNAs in colorectal cancer (CRC) remain largely unexplored. METHODS Differential circRNA expression was assessed using a human circRNA microarray in five CRC tissue and paired normal samples. CircGPRC5A expression was then confirmed in the CRC tissues and paired normal samples using qRT-PCR. The biological function of circGPRC5A in CRC were studied in vitro and in vivo. Western blotting, fluorescence in situ hybridization, immunofluorescence, RNA pulldown, mass spectrometry, immunoprecipitation, quantitative phosphoproteomics, and RNA-binding protein immunoprecipitation assays were used to study circGPRC5A. RESULTS Our analysis revealed that circGPRC5A expression was higher in CRC tissues compared to normal tissues and was associated with tumor size, tumor stage and lymph node status. CircGPRC5A promoted CRC cell proliferation, migration, and metastasis in vitro and in vivo. CircGPRC5A could stabilize PPP1CA protein by inhibiting the binding between UBA1 and PPP1CA, and increasing YAP dephosphorylation. CONCLUSIONS Our study revealed that circGPRC5A plays an essential function in CRC progression by stabilizing PPP1CA protein and enhancing YAP dephosphorylation. CircGPRC5A could act as a novel and potential target for CRC.
Collapse
Affiliation(s)
- Zhenzhou Chen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yidan Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kuan He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianguo Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qican Deng
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yajun Chen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhongxue Fu
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
30
|
Bernard C, Frauscher B, Gelinas J, Timofeev I. Sleep, oscillations, and epilepsy. Epilepsia 2023; 64 Suppl 3:S3-S12. [PMID: 37226640 PMCID: PMC10674035 DOI: 10.1111/epi.17664] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/27/2023] [Accepted: 05/23/2023] [Indexed: 05/26/2023]
Abstract
Sleep and wake are defined through physiological and behavioral criteria and can be typically separated into non-rapid eye movement (NREM) sleep stages N1, N2, and N3, rapid eye movement (REM) sleep, and wake. Sleep and wake states are not homogenous in time. Their properties vary during the night and day cycle. Given that brain activity changes as a function of NREM, REM, and wake during the night and day cycle, are seizures more likely to occur during NREM, REM, or wake at a specific time? More generally, what is the relationship between sleep-wake cycles and epilepsy? We will review specific examples from clinical data and results from experimental models, focusing on the diversity and heterogeneity of these relationships. We will use a top-down approach, starting with the general architecture of sleep, followed by oscillatory activities, and ending with ionic correlates selected for illustrative purposes, with respect to seizures and interictal spikes. The picture that emerges is that of complexity; sleep disruption and pathological epileptic activities emerge from reorganized circuits. That different circuit alterations can occur across patients and models may explain why sleep alterations and the timing of seizures during the sleep-wake cycle are patient-specific.
Collapse
Affiliation(s)
| | - Birgit Frauscher
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Jennifer Gelinas
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY, USA
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Igor Timofeev
- Faculté de Médecine, Département de Psychiatrie et de Neurosciences, Centre de Recherche CERVO, Université Laval, Québec, QC G1J2G3, Canada
| |
Collapse
|
31
|
Vaquer-Alicea A, Yu J, Liu H, Lucey BP. Plasma and cerebrospinal fluid proteomic signatures of acutely sleep-deprived humans: an exploratory study. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2023; 4:zpad047. [PMID: 38046221 PMCID: PMC10691441 DOI: 10.1093/sleepadvances/zpad047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/06/2023] [Indexed: 12/05/2023]
Abstract
Study Objectives Acute sleep deprivation affects both central and peripheral biological processes. Prior research has mainly focused on specific proteins or biological pathways that are dysregulated in the setting of sustained wakefulness. This exploratory study aimed to provide a comprehensive view of the biological processes and proteins impacted by acute sleep deprivation in both plasma and cerebrospinal fluid (CSF). Methods We collected plasma and CSF from human participants during one night of sleep deprivation and controlled normal sleep conditions. One thousand and three hundred proteins were measured at hour 0 and hour 24 using a high-scale aptamer-based proteomics platform (SOMAscan) and a systematic biological database tool (Metascape) was used to reveal altered biological pathways. Results Acute sleep deprivation decreased the number of upregulated and downregulated biological pathways and proteins in plasma but increased upregulated and downregulated biological pathways and proteins in CSF. Predominantly affected proteins and pathways were associated with immune response, inflammation, phosphorylation, membrane signaling, cell-cell adhesion, and extracellular matrix organization. Conclusions The identified modifications across biofluids add to evidence that acute sleep deprivation has important impacts on biological pathways and proteins that can negatively affect human health. As a hypothesis-driving study, these findings may help with the exploration of novel mechanisms that mediate sleep loss and associated conditions, drive the discovery of new sleep loss biomarkers, and ultimately aid in the identification of new targets for intervention to human diseases.
Collapse
Affiliation(s)
- Ana Vaquer-Alicea
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Jinsheng Yu
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Haiyan Liu
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Brendan P Lucey
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
32
|
Vanrobaeys Y, Peterson ZJ, Walsh EN, Chatterjee S, Lin LC, Lyons LC, Nickl-Jockschat T, Abel T. Spatial transcriptomics reveals unique gene expression changes in different brain regions after sleep deprivation. Nat Commun 2023; 14:7095. [PMID: 37925446 PMCID: PMC10625558 DOI: 10.1038/s41467-023-42751-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 10/20/2023] [Indexed: 11/06/2023] Open
Abstract
Sleep deprivation has far-reaching consequences on the brain and behavior, impacting memory, attention, and metabolism. Previous research has focused on gene expression changes in individual brain regions, such as the hippocampus or cortex. Therefore, it is unclear how uniformly or heterogeneously sleep loss affects the brain. Here, we use spatial transcriptomics to define the impact of a brief period of sleep deprivation across the brain in male mice. We find that sleep deprivation induced pronounced differences in gene expression across the brain, with the greatest changes in the hippocampus, neocortex, hypothalamus, and thalamus. Both the differentially expressed genes and the direction of regulation differed markedly across regions. Importantly, we developed bioinformatic tools to register tissue sections and gene expression data into a common anatomical space, allowing a brain-wide comparison of gene expression patterns between samples. Our results suggest that distinct molecular mechanisms acting in discrete brain regions underlie the biological effects of sleep deprivation.
Collapse
Affiliation(s)
- Yann Vanrobaeys
- Interdisciplinary Graduate Program in Genetics, University of Iowa, 357 Medical Research Center Iowa City, Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA, USA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 51 Newton Road, 2-417B Bowen Science Building, Iowa City, IA, USA
| | - Zeru J Peterson
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Emily N Walsh
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA, USA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 51 Newton Road, 2-417B Bowen Science Building, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, 356 Medical Research Center, Iowa City, IA, USA
| | - Snehajyoti Chatterjee
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA, USA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 51 Newton Road, 2-417B Bowen Science Building, Iowa City, IA, USA
| | - Li-Chun Lin
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA, USA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 51 Newton Road, 2-417B Bowen Science Building, Iowa City, IA, USA
- Department of Neurology, University of Iowa, Iowa City, IA, USA
| | - Lisa C Lyons
- Program in Neuroscience, Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Thomas Nickl-Jockschat
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA, USA.
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 51 Newton Road, 2-417B Bowen Science Building, Iowa City, IA, USA.
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA.
| | - Ted Abel
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA, USA.
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 51 Newton Road, 2-417B Bowen Science Building, Iowa City, IA, USA.
| |
Collapse
|
33
|
Pracucci E, Graham RT, Alberio L, Nardi G, Cozzolino O, Pillai V, Pasquini G, Saieva L, Walsh D, Landi S, Zhang J, Trevelyan AJ, Ratto GM. Daily rhythm in cortical chloride homeostasis underpins functional changes in visual cortex excitability. Nat Commun 2023; 14:7108. [PMID: 37925453 PMCID: PMC10625537 DOI: 10.1038/s41467-023-42711-7] [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: 11/10/2022] [Accepted: 10/19/2023] [Indexed: 11/06/2023] Open
Abstract
Cortical activity patterns are strongly modulated by fast synaptic inhibition mediated through ionotropic, chloride-conducting receptors. Consequently, chloride homeostasis is ideally placed to regulate activity. We therefore investigated the stability of baseline [Cl-]i in adult mouse neocortex, using in vivo two-photon imaging. We found a two-fold increase in baseline [Cl-]i in layer 2/3 pyramidal neurons, from day to night, with marked effects upon both physiological cortical processing and seizure susceptibility. Importantly, the night-time activity can be converted to the day-time pattern by local inhibition of NKCC1, while inhibition of KCC2 converts day-time [Cl-]i towards night-time levels. Changes in the surface expression and phosphorylation of the cation-chloride cotransporters, NKCC1 and KCC2, matched these pharmacological effects. When we extended the dark period by 4 h, mice remained active, but [Cl-]i was modulated as for animals in normal light cycles. Our data thus demonstrate a daily [Cl-]i modulation with complex effects on cortical excitability.
Collapse
Affiliation(s)
- Enrico Pracucci
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127, Pisa, Italy
| | - Robert T Graham
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Laura Alberio
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Gabriele Nardi
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127, Pisa, Italy
| | - Olga Cozzolino
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127, Pisa, Italy
| | - Vinoshene Pillai
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127, Pisa, Italy
| | - Giacomo Pasquini
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127, Pisa, Italy
| | - Luciano Saieva
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Darren Walsh
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Silvia Landi
- Institute of Neuroscience CNR, Pisa, Italy
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127, Pisa, Italy
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Institute of Health, University of Exeter, Hatherly Laboratories, Exeter, EX4 4PS, UK
- State Key Laboratory of Chemical Biology. Research Center of Chemical Kinomics, Shangai. Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Andrew J Trevelyan
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
| | - Gian-Michele Ratto
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127, Pisa, Italy.
- Institute of Neuroscience CNR, Pisa, Italy.
- Padova Neuroscience Center, Padova, Italy.
| |
Collapse
|
34
|
Kara N, Iweka CA, Blacher E. Chrono-Gerontology: Integrating Circadian Rhythms and Aging in Stroke Research. Adv Biol (Weinh) 2023; 7:e2300048. [PMID: 37409422 DOI: 10.1002/adbi.202300048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/14/2023] [Indexed: 07/07/2023]
Abstract
Stroke is a significant public health concern for elderly individuals. However, the majority of pre-clinical studies utilize young and healthy rodents, which may result in failure of candidate therapies in clinical trials. In this brief review/perspective, the complex link between circadian rhythms, aging, innate immunity, and the gut microbiome to ischemic injury onset, progression, and recovery is discussed. Short-chain fatty acids and nicotinamide adenine dinucleotide+ (NAD+ ) production by the gut microbiome are highlighted as key mechanisms with profound rhythmic behavior, and it is suggested to boost them as prophylactic/therapeutic approaches. Integrating aging, its associated comorbidities, and circadian regulation of physiological processes into stroke research may increase the translational value of pre-clinical studies and help to schedule the optimal time window for existing practices to improve stroke outcome and recovery.
Collapse
Affiliation(s)
- Nirit Kara
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus Givat-Ram, Jerusalem, 9190401, Israel
| | - Chinyere Agbaegbu Iweka
- Department of Neurology & Neurological Sciences, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Eran Blacher
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus Givat-Ram, Jerusalem, 9190401, Israel
| |
Collapse
|
35
|
Adamantidis AR, de Lecea L. Sleep and the hypothalamus. Science 2023; 382:405-412. [PMID: 37883555 DOI: 10.1126/science.adh8285] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/08/2023] [Indexed: 10/28/2023]
Abstract
Neural substrates of wakefulness, rapid eye movement sleep (REMS), and non-REMS (NREMS) in the mammalian hypothalamus overlap both anatomically and functionally with cellular networks that support physiological and behavioral homeostasis. Here, we review the roles of sleep neurons of the hypothalamus in the homeostatic control of thermoregulation or goal-oriented behaviors during wakefulness. We address how hypothalamic circuits involved in opposing behaviors such as core body temperature and sleep compute conflicting information and provide a coherent vigilance state. Finally, we highlight some of the key unresolved questions and challenges, and the promise of a more granular view of the cellular and molecular diversity underlying the integrative role of the hypothalamus in physiological and behavioral homeostasis.
Collapse
Affiliation(s)
- Antoine R Adamantidis
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Luis de Lecea
- Department of Psychiatry and Behavioural Sciences, Stanford, CA, USA
- Wu Tsai Neurosciences Institute Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
36
|
Noordally ZB, Hindle MM, Martin SF, Seaton DD, Simpson TI, Le Bihan T, Millar AJ. A phospho-dawn of protein modification anticipates light onset in the picoeukaryote Ostreococcus tauri. JOURNAL OF EXPERIMENTAL BOTANY 2023; 74:5514-5531. [PMID: 37481465 PMCID: PMC10540734 DOI: 10.1093/jxb/erad290] [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: 12/15/2022] [Accepted: 07/20/2023] [Indexed: 07/24/2023]
Abstract
Diel regulation of protein levels and protein modification had been less studied than transcript rhythms. Here, we compare transcriptome data under light-dark cycles with partial proteome and phosphoproteome data, assayed using shotgun MS, from the alga Ostreococcus tauri, the smallest free-living eukaryote. A total of 10% of quantified proteins but two-thirds of phosphoproteins were rhythmic. Mathematical modelling showed that light-stimulated protein synthesis can account for the observed clustering of protein peaks in the daytime. Prompted by night-peaking and apparently dark-stable proteins, we also tested cultures under prolonged darkness, where the proteome changed less than under the diel cycle. Among the dark-stable proteins were prasinophyte-specific sequences that were also reported to accumulate when O. tauri formed lipid droplets. In the phosphoproteome, 39% of rhythmic phospho-sites reached peak levels just before dawn. This anticipatory phosphorylation suggests that a clock-regulated phospho-dawn prepares green cells for daytime functions. Acid-directed and proline-directed protein phosphorylation sites were regulated in antiphase, implicating the clock-related casein kinases 1 and 2 in phase-specific regulation, alternating with the CMGC protein kinase family. Understanding the dynamic phosphoprotein network should be facilitated by the minimal kinome and proteome of O. tauri. The data are available from ProteomeXchange, with identifiers PXD001734, PXD001735, and PXD002909.
Collapse
Affiliation(s)
- Zeenat B Noordally
- SynthSys and School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Matthew M Hindle
- SynthSys and School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Sarah F Martin
- SynthSys and School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Daniel D Seaton
- SynthSys and School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - T Ian Simpson
- Institute for Adaptive and Neural Computation, School of Informatics, University of Edinburgh, Edinburgh EH8 9AB, UK
| | - Thierry Le Bihan
- SynthSys and School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Andrew J Millar
- SynthSys and School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| |
Collapse
|
37
|
Hu S, Jing Y, Li T, Wang YG, Liu Z, Gao J, Tian YC. Inferring circadian gene regulatory relationships from gene expression data with a hybrid framework. BMC Bioinformatics 2023; 24:362. [PMID: 37752445 PMCID: PMC10521455 DOI: 10.1186/s12859-023-05458-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 08/30/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND The central biological clock governs numerous facets of mammalian physiology, including sleep, metabolism, and immune system regulation. Understanding gene regulatory relationships is crucial for unravelling the mechanisms that underlie various cellular biological processes. While it is possible to infer circadian gene regulatory relationships from time-series gene expression data, relying solely on correlation-based inference may not provide sufficient information about causation. Moreover, gene expression data often have high dimensions but a limited number of observations, posing challenges in their analysis. METHODS In this paper, we introduce a new hybrid framework, referred to as Circadian Gene Regulatory Framework (CGRF), to infer circadian gene regulatory relationships from gene expression data of rats. The framework addresses the challenges of high-dimensional data by combining the fuzzy C-means clustering algorithm with dynamic time warping distance. Through this approach, we efficiently identify the clusters of genes related to the target gene. To determine the significance of genes within a specific cluster, we employ the Wilcoxon signed-rank test. Subsequently, we use a dynamic vector autoregressive method to analyze the selected significant gene expression profiles and reveal directed causal regulatory relationships based on partial correlation. CONCLUSION The proposed CGRF framework offers a comprehensive and efficient solution for understanding circadian gene regulation. Circadian gene regulatory relationships are inferred from the gene expression data of rats based on the Aanat target gene. The results show that genes Pde10a, Atp7b, Prok2, Per1, Rhobtb3 and Dclk1 stand out, which have been known to be essential for the regulation of circadian activity. The potential relationships between genes Tspan15, Eprs, Eml5 and Fsbp with a circadian rhythm need further experimental research.
Collapse
Affiliation(s)
- Shuwen Hu
- School of Computer Science, Queensland University of Technology, Brisbane, QLD, 4001, Australia
- Agriculture and Food, CSIRO, St Lucia, QLD, 4067, Australia
| | - Yi Jing
- Faculty of Science, The University of New South Wales, Sydney, 2052, Australia
| | - Tao Li
- School of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - You-Gan Wang
- Institute for Learning Sciences and Teacher Education, Australian Catholic University, Brisbane, QLD, 4000, Australia
| | - Zhenyu Liu
- School of Computer and Information Engineering, Inner Mongolia Agriculture University, Hohhot, 010018, China
| | - Jing Gao
- School of Computer and Information Engineering, Inner Mongolia Agriculture University, Hohhot, 010018, China.
| | - Yu-Chu Tian
- School of Computer Science, Queensland University of Technology, Brisbane, QLD, 4001, Australia.
| |
Collapse
|
38
|
Zhang BW, Dong H, Wu Z, Jiang X, Zou W. An Overview of the Mechanisms Involved in Neuralgia. J Inflamm Res 2023; 16:4087-4101. [PMID: 37745793 PMCID: PMC10516189 DOI: 10.2147/jir.s425966] [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: 06/14/2023] [Accepted: 08/26/2023] [Indexed: 09/26/2023] Open
Abstract
Neuralgia is a frequently occurring condition that causes chronic pain and burdens both patients and their families. Earlier research indicated that anti-inflammatory treatment, which was primarily utilized to address conditions like neuralgia, resulted in positive outcomes. However, recent years have witnessed the emergence of various novel mechanisms associated with pain-related disorders. This review provides a concise overview of the inflammatory mechanisms involved in neuralgia. It also examines recent advancements in research, exploring the influence of ion channels and synaptic proteins on neuralgia and its complications. Additionally, the interactions between these mechanisms are discussed with the aim of suggesting innovative therapeutic approaches and research directions for the management of neuralgia.
Collapse
Affiliation(s)
- Bai-Wen Zhang
- Heilongjiang University of Chinese Medicine, Harbin, 150040, People’s Republic of China
| | - Hao Dong
- Heilongjiang University of Chinese Medicine, Harbin, 150040, People’s Republic of China
| | - Zhe Wu
- Heilongjiang University of Chinese Medicine, Harbin, 150040, People’s Republic of China
| | - Xi Jiang
- Jinzhou Medical University, Jinzhou, 121001, People’s Republic of China
| | - Wei Zou
- The Third Department of Acupuncture and Moxibustion, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, People’s Republic of China
| |
Collapse
|
39
|
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] [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
|
40
|
Yuan Y, Chen Q, Brovkina M, Clowney EJ, Yadlapalli S. Clock-dependent chromatin accessibility rhythms regulate circadian transcription. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.15.553315. [PMID: 37645872 PMCID: PMC10462003 DOI: 10.1101/2023.08.15.553315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Chromatin organization plays a crucial role in gene regulation by controlling the accessibility of DNA to transcription machinery. While significant progress has been made in understanding the regulatory role of clock proteins in circadian rhythms, how chromatin organization affects circadian rhythms remains poorly understood. Here, we employed ATAC-seq (Assay for Transposase-Accessible Chromatin with Sequencing) on FAC-sorted Drosophila clock neurons to assess genome-wide chromatin accessibility over the circadian cycle. We observed significant circadian oscillations in chromatin accessibility at promoter and enhancer regions of hundreds of genes, with enhanced accessibility either at dusk or dawn, which correlated with their peak transcriptional activity. Notably, genes with enhanced accessibility at dusk were enriched with E-box motifs, while those more accessible at dawn were enriched with VRI/PDP1-box motifs, indicating that they are regulated by the core circadian feedback loops, PER/CLK and VRI/PDP1, respectively. Further, we observed a complete loss of chromatin accessibility rhythms in per01 null mutants, with chromatin consistently accessible throughout the circadian cycle, underscoring the critical role of Period protein in driving chromatin compaction during the repression phase. Together, this study demonstrates the significant role of chromatin organization in circadian regulation, revealing how the interplay between clock proteins and chromatin structure orchestrates the precise timing of biological processes throughout the day. This work further implies that variations in chromatin accessibility might play a central role in the generation of diverse circadian gene expression patterns in clock neurons.
Collapse
Affiliation(s)
- Ye Yuan
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Qianqian Chen
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Margarita Brovkina
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - E Josephine Clowney
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
- Michigan Neuroscience Institute Affiliate, University of Michigan, Ann Arbor, MI 48109, USA
| | - Swathi Yadlapalli
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
- Michigan Neuroscience Institute Affiliate, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
41
|
Lyons LC, Vanrobaeys Y, Abel T. Sleep and memory: The impact of sleep deprivation on transcription, translational control, and protein synthesis in the brain. J Neurochem 2023; 166:24-46. [PMID: 36802068 PMCID: PMC10919414 DOI: 10.1111/jnc.15787] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/20/2023]
Abstract
In countries around the world, sleep deprivation represents a widespread problem affecting school-age children, teenagers, and adults. Acute sleep deprivation and more chronic sleep restriction adversely affect individual health, impairing memory and cognitive performance as well as increasing the risk and progression of numerous diseases. In mammals, the hippocampus and hippocampus-dependent memory are vulnerable to the effects of acute sleep deprivation. Sleep deprivation induces changes in molecular signaling, gene expression and may cause changes in dendritic structure in neurons. Genome wide studies have shown that acute sleep deprivation alters gene transcription, although the pool of genes affected varies between brain regions. More recently, advances in research have drawn attention to differences in gene regulation between the level of the transcriptome compared with the pool of mRNA associated with ribosomes for protein translation following sleep deprivation. Thus, in addition to transcriptional changes, sleep deprivation also affects downstream processes to alter protein translation. In this review, we focus on the multiple levels through which acute sleep deprivation impacts gene regulation, highlighting potential post-transcriptional and translational processes that may be affected by sleep deprivation. Understanding the multiple levels of gene regulation impacted by sleep deprivation is essential for future development of therapeutics that may mitigate the effects of sleep loss.
Collapse
Affiliation(s)
- Lisa C Lyons
- Program in Neuroscience, Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Yann Vanrobaeys
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, Iowa City, Iowa, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, Iowa City, Iowa, USA
| |
Collapse
|
42
|
Martin SC, Joyce KK, Harper KM, Nikolova VD, Cohen TJ, Moy SS, Diering GH. Sleep disruption precedes forebrain synaptic Tau burden and contributes to cognitive decline in a sex-dependent manner in the P301S Tau transgenic mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544101. [PMID: 37333395 PMCID: PMC10274785 DOI: 10.1101/2023.06.07.544101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Background Sleep is an essential process that supports brain health and cognitive function in part through the modification of neuronal synapses. Sleep disruption, and impaired synaptic processes, are common features in neurodegenerative diseases, including Alzheimer's disease (AD). However, the casual role of sleep disruption in disease progression is not clear. Neurofibrillary tangles, made from hyperphosphorylated and aggregated Tau protein, form one of the major hallmark pathologies seen in AD and contribute to cognitive decline, synapse loss and neuronal death.Tau has been shown to aggregate in synapses which may impair restorative synapse processes occurring during sleep. However, it remains unclear how sleep disruption and synaptic Tau pathology interact to drive cognitive decline. It is also unclear whether the sexes show differential vulnerability to the effects of sleep loss in the context of neurodegeneration. Methods We used a piezoelectric home-cage monitoring system to measure sleep behavior in 3-11month-old transgenic hTau P301S Tauopathy model mice (PS19) and littermate controls of both sexes. Subcellular fractionation and Western blot was used to examine Tau pathology in mouse forebrain synapse fractions. To examine the role of sleep disruption in disease progression, mice were exposed to acute or chronic sleep disruption. The Morris water maze test was used to measure spatial learning and memory performance. Results PS19 mice exhibited a selective loss of sleep during the dark phase, referred to as hyperarousal, as an early symptom with an onset of 3months in females and 6months in males. At 6months, forebrain synaptic Tau burden did not correlate with sleep measures and was not affected by acute or chronic sleep disruption. Chronic sleep disruption accelerated the onset of decline of hippocampal spatial memory in PS19 males, but not females. Conclusions Dark phase hyperarousal is an early symptom in PS19 mice that precedes robust Tau aggregation. We find no evidence that sleep disruption is a direct driver of Tau pathology in the forebrain synapse. However, sleep disruption synergized with Tau pathology to accelerate the onset of cognitive decline in males. Despite the finding that hyperarousal appears earlier in females, female cognition was resilient to the effects of sleep disruption.
Collapse
|
43
|
Bárez-López S, Gadd GJ, Pauža AG, Murphy D, Greenwood MP. Isoflurane Rapidly Modifies Synaptic and Cytoskeletal Phosphoproteomes of the Supraoptic Nucleus of the Hypothalamus and the Cortex. Neuroendocrinology 2023; 113:1008-1023. [PMID: 37271138 DOI: 10.1159/000531352] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/24/2023] [Indexed: 06/06/2023]
Abstract
INTRODUCTION Despite the widespread use of general anaesthetics, the mechanisms mediating their effects are still not understood. Although suppressed in most parts of the brain, neuronal activity, as measured by FOS activation, is increased in the hypothalamic supraoptic nucleus (SON) by numerous general anaesthetics, and evidence points to this brain region being involved in the induction of general anaesthesia (GA) and natural sleep. Posttranslational modifications of proteins, including changes in phosphorylation, enable fast modulation of protein function which could be underlying the rapid effects of GA. In order to identify potential phosphorylation events in the brain-mediating GA effects, we have explored the phosphoproteome responses in the rat SON and compared these to cingulate cortex (CC) which displays no FOS activation in response to general anaesthetics. METHODS Adult Sprague-Dawley rats were treated with isoflurane for 15 min. Proteins from the CC and SON were extracted and processed for nano-LC mass spectrometry (LC-MS/MS). Phosphoproteomic determinations were performed by LC-MS/MS. RESULTS We found many changes in the phosphoproteomes of both the CC and SON in response to 15 min of isoflurane exposure. Pathway analysis indicated that proteins undergoing phosphorylation adaptations are involved in cytoskeleton remodelling and synaptic signalling events. Importantly, changes in protein phosphorylation appeared to be brain region specific suggesting that differential phosphorylation adaptations might underlie the different neuronal activity responses to GA between the CC and SON. CONCLUSION In summary, these data suggest that rapid posttranslational modifications in proteins involved in cytoskeleton remodelling and synaptic signalling events might mediate the central mechanisms mediating GA.
Collapse
Affiliation(s)
- Soledad Bárez-López
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - George J Gadd
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
| | - Audrys G Pauža
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
- Translational Cardio-Respiratory Research Group, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - David Murphy
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
| | - Michael P Greenwood
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
44
|
Liu W, Ma R, Sun C, Xu Y, Liu Y, Hu J, Ma Y, Wang D, Wen D, Yu Y. Implications from proteomic studies investigating circadian rhythm disorder-regulated neurodegenerative disease pathology. Sleep Med Rev 2023; 70:101789. [PMID: 37253318 DOI: 10.1016/j.smrv.2023.101789] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 06/01/2023]
Abstract
Neurodegenerative diseases (NDs) affect 15% of the world's population and are becoming an increasingly common cause of morbidity and mortality worldwide. Circadian rhythm disorders (CRDs) have been reported to be involved in the pathogenic regulation of various neurologic diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis and amyotrophic lateral sclerosis. Proteomic technology is helpful to explore treatment targets for CRDs in patients with NDs. Here, we review the key differentially expressed (DE) proteins identified in previous proteomic studies investigating NDs, CRDs and associated models and the related pathways identified by enrichment analysis. Furthermore, we summarize the advantages and disadvantages of the above studies and propose new proteomic technologies for the precise study of circadian disorder-mediated regulation of ND pathology. This review provides a theoretical and technical reference for the precise study of circadian disorder-mediated regulation of ND pathology.
Collapse
Affiliation(s)
- Weiwei Liu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China
| | - Ruze Ma
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; Department of Gerontology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Chen Sun
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Yingxi Xu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China
| | - Yang Liu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China
| | - Jiajin Hu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China
| | - Yanan Ma
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; Department of Epidemiology and Health Statistics, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Difei Wang
- Department of Gerontology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Deliang Wen
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China.
| | - Yang Yu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
45
|
Barrio-Alonso E, Lituma PJ, Notaras MJ, Albero R, Bouchekioua Y, Wayland N, Stankovic IN, Jain T, Gao S, Calderon DP, Castillo PE, Colak D. Circadian protein TIMELESS regulates synaptic function and memory by modulating cAMP signaling. Cell Rep 2023; 42:112375. [PMID: 37043347 PMCID: PMC10564971 DOI: 10.1016/j.celrep.2023.112375] [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: 07/21/2022] [Revised: 03/07/2023] [Accepted: 03/24/2023] [Indexed: 04/13/2023] Open
Abstract
The regulation of neurons by circadian clock genes is thought to contribute to the maintenance of neuronal functions that ultimately underlie animal behavior. However, the impact of specific circadian genes on cellular and molecular mechanisms controlling synaptic plasticity and cognitive function remains elusive. Here, we show that the expression of the circadian protein TIMELESS displays circadian rhythmicity in the mammalian hippocampus. We identify TIMELESS as a chromatin-bound protein that targets synaptic-plasticity-related genes such as phosphodiesterase 4B (Pde4b). By promoting Pde4b transcription, TIMELESS negatively regulates cAMP signaling to modulate AMPA receptor GluA1 function and influence synaptic plasticity. Conditional deletion of Timeless in the adult forebrain impairs working and contextual fear memory in mice. These cognitive phenotypes were accompanied by attenuation of hippocampal Schaffer-collateral synapse long-term potentiation. Together, these data establish a neuron-specific function of mammalian TIMELESS by defining a mechanism that regulates synaptic plasticity and cognitive function.
Collapse
Affiliation(s)
- Estibaliz Barrio-Alonso
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Pablo J Lituma
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Michael J Notaras
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Robert Albero
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Youcef Bouchekioua
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, USA
| | - Natalie Wayland
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Isidora N Stankovic
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Tanya Jain
- Program of Neurosciences, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Sijia Gao
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, USA
| | | | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Dilek Colak
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA; Gale & Ira Drukier Institute for Children's Health, Weill Cornell Medical College, Cornell University, New York, NY, USA.
| |
Collapse
|
46
|
Brodt S, Inostroza M, Niethard N, Born J. Sleep-A brain-state serving systems memory consolidation. Neuron 2023; 111:1050-1075. [PMID: 37023710 DOI: 10.1016/j.neuron.2023.03.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/23/2023] [Accepted: 03/06/2023] [Indexed: 04/08/2023]
Abstract
Although long-term memory consolidation is supported by sleep, it is unclear how it differs from that during wakefulness. Our review, focusing on recent advances in the field, identifies the repeated replay of neuronal firing patterns as a basic mechanism triggering consolidation during sleep and wakefulness. During sleep, memory replay occurs during slow-wave sleep (SWS) in hippocampal assemblies together with ripples, thalamic spindles, neocortical slow oscillations, and noradrenergic activity. Here, hippocampal replay likely favors the transformation of hippocampus-dependent episodic memory into schema-like neocortical memory. REM sleep following SWS might balance local synaptic rescaling accompanying memory transformation with a sleep-dependent homeostatic process of global synaptic renormalization. Sleep-dependent memory transformation is intensified during early development despite the immaturity of the hippocampus. Overall, beyond its greater efficacy, sleep consolidation differs from wake consolidation mainly in that it is supported, rather than impaired, by spontaneous hippocampal replay activity possibly gating memory formation in neocortex.
Collapse
Affiliation(s)
- Svenja Brodt
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; Max-Planck-Institute for Biological Cybernetics, Tübingen, Germany
| | - Marion Inostroza
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| | - Niels Niethard
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| | - Jan Born
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; Werner Reichert Center for Integrative Neuroscience, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
47
|
Schibler U, Dibner C, Ripperger J. Steve Brown. J Biol Rhythms 2023; 38:119-124. [PMID: 36762620 PMCID: PMC10037542 DOI: 10.1177/07487304231152275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Affiliation(s)
- Ueli Schibler
- Ueli Schibler, Department
of Molecular and Cellular Biology, University of Geneva, 30, Quai Ernest
Ansermet, Geneva, CH-1211, Switzerland; e-mail:
| | | | | |
Collapse
|
48
|
Liu S, Cheng L, Liu Y, Zhan S, Wu Z, Zhang X. Relationship between Dietary Polyphenols and Gut Microbiota: New Clues to Improve Cognitive Disorders, Mood Disorders and Circadian Rhythms. Foods 2023; 12:foods12061309. [PMID: 36981235 PMCID: PMC10048542 DOI: 10.3390/foods12061309] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/08/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Cognitive, mood and sleep disorders are common and intractable disorders of the central nervous system, causing great inconvenience to the lives of those affected. The gut-brain axis plays a vital role in studying neurological disorders such as neurodegenerative diseases by acting as a channel for a bidirectional information exchange between the gut microbiota and the nervous system. Dietary polyphenols have received widespread attention because of their excellent biological activity and their wide range of sources, structural diversity and low toxicity. Dietary intervention through the increased intake of dietary polyphenols is an emerging strategy for improving circadian rhythms and treating metabolic disorders. Dietary polyphenols have been shown to play an essential role in regulating intestinal flora, mainly by maintaining the balance of the intestinal flora and enhancing host immunity, thereby suppressing neurodegenerative pathologies. This paper reviewed the bidirectional interactions between the gut microbiota and the brain and their effects on the central nervous system, focusing on dietary polyphenols that regulate circadian rhythms and maintain the health of the central nervous system through the gut-brain axis.
Collapse
Affiliation(s)
- Siyu Liu
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Lu Cheng
- Department of Food Science, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Yanan Liu
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Shengnan Zhan
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Zufang Wu
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China
| |
Collapse
|
49
|
Sun C, Schuman E. A multi-omics view of neuronal subcellular protein synthesis. Curr Opin Neurobiol 2023; 80:102705. [PMID: 36913750 DOI: 10.1016/j.conb.2023.102705] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 03/13/2023]
Abstract
While it has long been known that protein synthesis is necessary for long-term memory in the brain, the logistics of neuronal protein synthesis is complicated by the extensive subcellular compartmentalization of the neuron. Local protein synthesis solves many of the logistic problems posed by the extreme complexity of dendritic and axonal arbors and the huge number of synapses. Here we review recent multi-omic and quantitative studies that elaborate a systems view of decentralized neuronal protein synthesis. We highlight recent insights from the transcriptomic, translatomic, and proteomic levels, discuss the nuanced logic of local protein synthesis for different protein features, and list the missing information needed to build a comprehensive logistic model for neuronal protein supply.
Collapse
Affiliation(s)
- Chao Sun
- Max Planck Institute for Brain Research, Frankfurt, Germany; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Denmark; Aarhus University, Department of Molecular Biology and Genetics, Universitetsbyen 81, 8000 Aarhus C, Denmark. https://twitter.com/LukeChaoSun
| | - Erin Schuman
- Max Planck Institute for Brain Research, Frankfurt, Germany.
| |
Collapse
|
50
|
Cerasuolo M, Papa M, Colangelo AM, Rizzo MR. Alzheimer’s Disease from the Amyloidogenic Theory to the Puzzling Crossroads between Vascular, Metabolic and Energetic Maladaptive Plasticity. Biomedicines 2023; 11:biomedicines11030861. [PMID: 36979840 PMCID: PMC10045635 DOI: 10.3390/biomedicines11030861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Alzheimer’s disease (AD) is a progressive and degenerative disease producing the most common type of dementia worldwide. The main pathogenetic hypothesis in recent decades has been the well-known amyloidogenic hypothesis based on the involvement of two proteins in AD pathogenesis: amyloid β (Aβ) and tau. Amyloid deposition reported in all AD patients is nowadays considered an independent risk factor for cognitive decline. Vascular damage and blood–brain barrier (BBB) failure in AD is considered a pivotal mechanism for brain injury, with increased deposition of both immunoglobulins and fibrin. Furthermore, BBB dysfunction could be an early sign of cognitive decline and the early stages of clinical AD. Vascular damage generates hypoperfusion and relative hypoxia in areas with high energy demand. Long-term hypoxia and the accumulation within the brain parenchyma of neurotoxic molecules could be seeds of a self-sustaining pathological progression. Cellular dysfunction comprises all the elements of the neurovascular unit (NVU) and neuronal loss, which could be the result of energy failure and mitochondrial impairment. Brain glucose metabolism is compromised, showing a specific region distribution. This energy deficit worsens throughout aging. Mild cognitive impairment has been reported to be associated with a glucose deficit in the entorhinal cortex and in the parietal lobes. The current aim is to understand the complex interactions between amyloid β (Aβ) and tau and elements of the BBB and NVU in the brain. This new approach aimed at the study of metabolic mechanisms and energy insufficiency due to mitochondrial impairment would allow us to define therapies aimed at predicting and slowing down the progression of AD.
Collapse
Affiliation(s)
- Michele Cerasuolo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Michele Papa
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- SYSBIO Centre of Systems Biology ISBE-IT, 20126 Milan, Italy
- Correspondence:
| | - Anna Maria Colangelo
- SYSBIO Centre of Systems Biology ISBE-IT, 20126 Milan, Italy
- Laboratory of Neuroscience “R. Levi-Montalcini”, Department of Biotechnology and Biosciences, NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126 Milano, Italy
| | - Maria Rosaria Rizzo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| |
Collapse
|