1
|
Wu T, Yu Q, Zhu X, Li Y, Zhang M, Deng J, Lu L. Embracing Internal States: A Review of Optimization of Repetitive Transcranial Magnetic Stimulation for Treating Depression. Neurosci Bull 2025; 41:866-880. [PMID: 39976854 PMCID: PMC12014982 DOI: 10.1007/s12264-024-01347-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/05/2024] [Indexed: 04/23/2025] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a rapid and effective therapy for major depressive disorder; however, there is significant variability in therapeutic outcomes both within and across individuals, with approximately 50% of patients showing no response to rTMS treatment. Many studies have personalized the stimulation parameters of rTMS (e.g., location and intensity of stimulation) according to the anatomical and functional structure of the brain. In addition to these parameters, the internal states of the individual, such as circadian rhythm, behavior/cognition, neural oscillation, and neuroplasticity, also contribute to the variation in rTMS effects. In this review, we summarize the current literature on the interaction between rTMS and internal states. We propose two possible methods, multimodal treatment, and adaptive closed-loop treatment, to integrate patients' internal states to achieve better rTMS treatment for depression.
Collapse
Affiliation(s)
- Tingting Wu
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No. 2018RU006), Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100080, China
| | - Qiuxuan Yu
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No. 2018RU006), Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100080, China
| | - Ximei Zhu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100080, China
| | - Yinjiao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100080, China
| | - Mingyue Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100080, China
| | - Jiahui Deng
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100080, China.
| | - Lin Lu
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No. 2018RU006), Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China.
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100080, China.
| |
Collapse
|
2
|
Sharma H, Robea MA, McGrory NH, Bazan DC, Burton EA, Burgess HA. Functional interrogation of neuronal connections by chemoptogenetic presynaptic ablation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647277. [PMID: 40236196 PMCID: PMC11996543 DOI: 10.1101/2025.04.04.647277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Most neurons are embedded in multiple circuits, with signaling to distinct postsynaptic partners playing functionally different roles. The function of specific connections can be interrogated using synaptically localized optogenetic effectors, however these tools are often experimentally difficult to validate or produce paradoxical outcomes. We have developed a system for photoablation of synaptic connections originating from genetically defined neurons, based on presynaptic localization of the fluorogen activating protein dL5** that acts as a photosensitizer when bound to a cell-permeable dye. Using the well mapped zebrafish escape circuit as a readout, we first show that cytoplasmically expressed dL5** enables efficient spatially targeted neuronal ablation using near infra-red light. We then demonstrate that spatially patterned illumination of presynaptically localized dL5** can effectively disconnect neurons from selected downstream partners, producing precise behavioral deficits. This technique should be applicable to almost any genetically tractable neuronal circuit, enabling precise manipulation of functional connectivity within the nervous system.
Collapse
|
3
|
Biswas T, Hassan H, Rohner N. Differentially expressed miRNAs offer new perspective into cave adaptation of Astyanax mexicanus. Ann N Y Acad Sci 2025; 1546:173-181. [PMID: 40082196 PMCID: PMC11998478 DOI: 10.1111/nyas.15300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Astyanax mexicanus, a species with both surface-dwelling and multiple cave-dwelling populations, offers a unique opportunity to study repeated adaptation to dark and resource-scarce environments. While previous work has identified large-scale gene expression changes between morphs under even identical laboratory conditions, the regulatory basis of these expression differences remains largely unexplored. In this study, we focus on microRNAs (miRNAs) as key regulators of gene expression. Our analysis identified 683 mature miRNAs, establishing the first comprehensive catalog of miRNAs for this species. We identified a unique subset of differentially expressed miRNAs common to all studied cave-dwelling populations, potentially orchestrating the nuanced gene expression patterns required for survival in the cave milieu. Furthermore, we performed in silico target prediction of these miRNAs, revealing possible roles in developmental and metabolic pathways pivotal for thriving in nutrient-limited cave conditions. Interestingly, we also observed that Molino, which is the "youngest" of the three cavefish analyzed in this study, exhibited the most abundant number of differentially expressed mature miRNAs among the cave morphs. The comprehensive miRNA catalog generated, along with the insight into their differential expression across different morphs, will guide future investigations into the intricate world of miRNA-mediated evolution of complex traits.
Collapse
Affiliation(s)
| | - Huzaifa Hassan
- Stowers Institute for Medical ResearchKansas CityMissouriUSA
| | - Nicolas Rohner
- Stowers Institute for Medical ResearchKansas CityMissouriUSA
- Institute for Integrative Cell Biology and PhysiologyUniversity of MünsterMünsterGermany
| |
Collapse
|
4
|
Aliev F, De Sa Nogueira D, Aston-Jones G, Dick DM. Genetic associations between orexin genes and phenotypes related to behavioral regulation in humans, including substance use. Mol Psychiatry 2025:10.1038/s41380-025-02895-4. [PMID: 39880903 DOI: 10.1038/s41380-025-02895-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/23/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025]
Abstract
The hypothalamic neuropeptide system of orexin (hypocretin) neurons provides projections throughout the neuraxis and has been linked to sleep regulation, feeding and motivation for salient rewards including drugs of abuse. However, relatively little has been done to examine genes associated with orexin signaling and specific behavioral phenotypes in humans. Here, we tested for association of twenty-seven genes involved in orexin signaling with behavioral phenotypes in humans. We tested the full gene set, functional subsets, and individual genes involved in orexin signaling. Our primary phenotype of interest was Externalizing, a composite factor comprised of behaviors and disorders associated with reward-seeking, motivation, and behavioral regulation. We also tested for association with additional phenotypes that have been related to orexin regulation in model organism studies, including alcohol consumption, problematic alcohol use, daytime sleepiness, insomnia, cigarettes per day, smoking initiation, and body mass index. The composite set of 27 genes corresponding to orexin function was highly associated with Externalizing, as well as with alcohol consumption, insomnia, cigarettes per day, smoking initiation and BMI. In addition, all gene subsets (except the OXR2/HCRTR2 subset) were associated with Externalizing. BMI was significantly associated with all gene subsets. The "validated factors for PPOX/HCRT" and "PPOX/HCRT upregulation" gene subsets also were associated with alcohol consumption. Individually, 8 genes showed a strong association with Externalizing, 12 with BMI, 7 with smoking initiation, 3 with alcohol consumption, and 2 with problematic alcohol use, after correction for multiple testing. This study indicates that orexin genes are associated with multiple behaviors and disorders related to self-regulation in humans. This is consistent with prior work in animals that implicated orexin signaling in motivational activation induced by salient stimuli, and supports the hypothesis that orexin signaling is an important potential therapeutic target for numerous behavioral disorders.
Collapse
Affiliation(s)
- Fazil Aliev
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - David De Sa Nogueira
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - Gary Aston-Jones
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - Danielle M Dick
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA.
| |
Collapse
|
5
|
Fu T, Zhang W, Guo R, He S, Yu S, Wang H, Zhang Y, Wu Y. Inclusion of hypocretin-1 improved performance of poor sleep quality prediction for elderly patients with acute ischemic stroke: a prospective cohort study. Front Aging Neurosci 2025; 16:1509846. [PMID: 39839304 PMCID: PMC11747788 DOI: 10.3389/fnagi.2024.1509846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
Background Hypocretin-1 is a vital neurotransmitter in regulating the sleep-wake cycle and provides neuroprotection against cerebral ischemia. We aims to develop a poor sleep quality predictive model for elderly population with acute ischemic stroke. Methods A total of 183 consecutively elderly patients were included in the prospective cohort study. Sleep quality was assessed by the Pittsburgh Sleep Quality Index (PSQI). Cerebrospinal fluid samples were taken within 24 h of acute ischemic stroke onset. After selecting optimal predictors via univariate analysis and bootstrapped stepwise logistic regression, the predictive model was developed. The models were internally validated and evaluated comprehensively via discrimination, calibration, and clinical utility. Results The prevalence of poor sleep (PSQI >7) was 64.5% among elderly individuals experiencing acute ischemic stroke. The study developed a predictive model using hypocretin-1, hypertension, stroke history, the National Institutes of Health Stroke score, and depression. Adding hypocretin-1 (as continuous variable) significantly improved the model performance greatly, as the area under the receiver operating characteristic curve increased from 0.799 to 0.845 (p < 0.001). The optimal cutoff value for hypocretin-1 was 74.94 pg/mL. Adding hypocretin-1 (as binary variable) significantly improved the model performance greatly, as the AUC increased from 0.799 to 0.857 (p < 0.001). Conclusion Reduced cerebrospinal fluid levels of hypocretin-1 at admission were an independent poor sleep quality predictor and the model demonstrated superior performance. The combination of hypocretin-1 could offer valuable prognostic information for post-stroke sleep quality in elderly patients with acute ischemic stroke.
Collapse
Affiliation(s)
- Ting Fu
- School of Nursing, Capital Medical University, Beijing, China
| | - Weiwei Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Rongrong Guo
- School of Nursing, Capital Medical University, Beijing, China
| | - Shuang He
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Saiying Yu
- School of Nursing, Capital Medical University, Beijing, China
| | - Huiying Wang
- School of Nursing, Capital Medical University, Beijing, China
| | - Yunfeng Zhang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Ying Wu
- School of Nursing, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Dyachuk V. The Role and Mechanisms of the Hypocretin System in Zebrafish ( Danio rerio). Int J Mol Sci 2024; 26:256. [PMID: 39796111 PMCID: PMC11719587 DOI: 10.3390/ijms26010256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/28/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
Sleep is the most important physiological function of all animals studied to date. Sleep disorders include narcolepsy, which is characterized by excessive daytime sleepiness, disruption of night sleep, and muscle weakness-cataplexy. Narcolepsy is known to be caused by the degeneration of orexin-synthesizing neurons (hypocretin (HCRT) neurons or orexin neurons) in the hypothalamus. In mammals, HCRT neurons primarily regulate the sleep/wake cycle, nutrition, reward seeking, and addiction development. The hypocretin system of the brain is involved in a number of neurological disorders. The distinctive pathologies associated with the disruption of HCRT neurons are narcolepsy and cataplexy, which are caused by the loss of hypocretin neurons that produce HCRT. In Danio, the hypocretin system is also involved in the regulation of sleep and wakefulness. It is represented by a single hcrt gene that encodes the peptides HCRT1 and HCRT2, as well as one HCRT receptor (HCRTR), which is structurally closest to the mammalian HCRTR2. The overexpression of the hcrt gene in Danio rerio larvae causes wakefulness, whereas the physical destruction of HCRT cells or a pharmacological blockade of the type 2 hypocretin receptor leads to fragmentation of sleep in fish larvae, which is also observed in patients with narcolepsy. These data confirm the evolutionary conservatism of the hypocretin system. Thus, Danio rerio is an ideal model for studying the functions of HCRT neural networks and their functions.
Collapse
Affiliation(s)
- Vyacheslav Dyachuk
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia
| |
Collapse
|
7
|
Sagi D, Tibi M, Admati I, Lerer-Goldshtein T, Hochgerner H, Zeisel A, Appelbaum L. Single-Cell Profiling Uncovers Evolutionary Divergence of Hypocretin/Orexin Neuronal Subpopulations. J Neurosci 2024; 44:e0095242024. [PMID: 39122556 PMCID: PMC11376333 DOI: 10.1523/jneurosci.0095-24.2024] [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: 01/15/2024] [Revised: 06/17/2024] [Accepted: 07/12/2024] [Indexed: 08/12/2024] Open
Abstract
Brain nuclei are traditionally defined by their anatomy, activity, and expression of specific markers. The hypothalamus contains discrete neuronal populations that coordinate fundamental behavioral functions, including sleep and wakefulness, in all vertebrates. Particularly, the diverse roles of hypocretin/orexin (Hcrt)-releasing neurons suggest functional heterogeneity among Hcrt neurons. Using single-cell RNA sequencing (scRNA-seq) and high-resolution imaging of the adult male and female zebrafish hypothalamic periventricular zone, we identified 21 glutamatergic and 28 GABAergic cell types. Integration of zebrafish and mouse scRNA-seq revealed evolutionary conserved and divergent hypothalamic cell types. The expression of specific genes, including npvf, which encodes a sleep-regulating neuropeptide, was enriched in subsets of glutamatergic Hcrt neurons in both larval and adult zebrafish. The genetic profile, activity, and neurite processing of the neuronal subpopulation that coexpresses both Hcrt and Npvf (Hcrt+Npvf+) differ from other Hcrt neurons. These interspecies findings provide a unified annotation of hypothalamic cell types and suggest that the heterogeneity of Hcrt neurons enables multifunctionality, such as consolidation of both wake and sleep by the Hcrt- and Npvf-releasing neuronal subpopulation.
Collapse
Affiliation(s)
- Dana Sagi
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 590002, Israel
| | - Muhammad Tibi
- The Faculty of Biotechnology and Food Engineering, Technion 3200, Israel
| | - Inbal Admati
- The Faculty of Biotechnology and Food Engineering, Technion 3200, Israel
| | - Tali Lerer-Goldshtein
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 590002, Israel
| | - Hannah Hochgerner
- The Faculty of Biotechnology and Food Engineering, Technion 3200, Israel
| | - Amit Zeisel
- The Faculty of Biotechnology and Food Engineering, Technion 3200, Israel
| | - Lior Appelbaum
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 590002, Israel
| |
Collapse
|
8
|
Cui M, Meng P, Wang S, Feng Q, Liu G, Zhao P. Differential effects of AKT1 and AKT2 on sleep-wake activity under basal conditions and in response to LPS challenge in mice. Sleep Biol Rhythms 2024; 22:411-421. [PMID: 38962788 PMCID: PMC11217225 DOI: 10.1007/s41105-024-00519-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 02/13/2024] [Indexed: 07/05/2024]
Abstract
Infectious challenge can trigger alterations in sleep-wake behavior. Accumulating evidence has shown that the serine/threonine kinases Akt1 and Akt2 are important targets in both physiological and infectious signaling processes. However, the involvement of Akt1 and Akt2 in sleep-wake activity under basal conditions and in response to inflammatory stimulation has not been established. In the present study, we assessed the precise role of Akt1 and Akt2 in sleep-wake behavior using electroencephalography (EEG)/electromyography (EMG) data from Akt1- and Akt2-deficient mice and wild-type (WT) mice. The results showed that both Akt1 and Akt2 deficiency affect sleep-wake activity, as indicated by reduced nonrapid eye movement (NREM) sleep and increased wakefulness in mutant mice compared to WT mice. Sleep amount and intensity (delta, theta and alpha activity) at night were also drastically attenuated in Akt1- and Akt2-deficient mice. Moreover, since Akt1 and Akt2 are involved in immune responses, we assessed their roles in the sleep response to the inflammatory stimulus lipopolysaccharide (LPS) throughout the following 24 h. We observed that the decrease in wakefulness and increase in NREM sleep induced by LPS were restored in Akt1 knockout mice but not in Akt2 knockout mice. Correspondingly, the decrease in the number of positive orexin-A neurons induced by LPS was abrogated in Akt1 knockout mice but not in Akt2 knockout mice. Our results revealed that both Akt1 and Akt2 deficiency affect the sleep response under basal conditions, but only Akt1 deficiency protects against the aberrant changes in sleep behavior induced by peripheral immune challenge. Supplementary Information The online version contains supplementary material available at 10.1007/s41105-024-00519-y.
Collapse
Affiliation(s)
- Meng Cui
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122 Jiangsu People’s Republic of China
| | - Pengfei Meng
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122 Jiangsu People’s Republic of China
| | - Shaohe Wang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122 Jiangsu People’s Republic of China
| | - Qingyuan Feng
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122 Jiangsu People’s Republic of China
| | - Guangming Liu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122 Jiangsu People’s Republic of China
| | - Peng Zhao
- Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214000 Jiangsu Province People’s Republic of China
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122 Jiangsu People’s Republic of China
| |
Collapse
|
9
|
Wang K, Ren S, Jia Y, Yan X. An Ultrasensitive Biomimetic Optic Afferent Nervous System with Circadian Learnability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309489. [PMID: 38468430 DOI: 10.1002/advs.202309489] [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: 12/06/2023] [Revised: 02/04/2024] [Indexed: 03/13/2024]
Abstract
The optic afferent nervous system (OANS) plays a significant role in generating vision and circadian behaviors based on light detection and signals from the endocrine system. However, the bionic simulation of this photochemically mediated behavior is still a challenge for neuromorphic devices. Herein, stimuli of neurotransmitters at ultralow concentrations and illumination are coupled to artificial synapses with the aid of biofunctionalized heterojunction and tunneling to successfully simulate a circadian neural response. Furthermore, the mechanisms underlying the photosensitive synaptic current in response to stimuli are described. Interestingly, this OANS is demonstrated to be capable of mimicking normal and abnormal circadian learnability by combining the measured synaptic current with a three-layer spike neural network. Strong theoretical and experimental evidence, as well as applications, are provided for the proposed biomimetic OANS to demonstrate that it can reproduce biological circadian behavior, thus establishing it as a promising candidate for future neuromorphic intelligent robots.
Collapse
Affiliation(s)
- Kaiyang Wang
- College of Electronic Information and Optical Engineering, Nankai University, Tianjin, 300071, P. R. China
| | - Shuhui Ren
- College of Electronic Information and Optical Engineering, Nankai University, Tianjin, 300071, P. R. China
| | - Yunfang Jia
- College of Electronic Information and Optical Engineering, Nankai University, Tianjin, 300071, P. R. China
| | - Xiaobing Yan
- Key Laboratory of Brain-Like Neuromorphic Devices and Systems of Hebei Province, College of Electron and Information Engineering, Hebei University, Baoding, 071002, P. R. China
| |
Collapse
|
10
|
Terzi A, Ngo KJ, Mourrain P. Phylogenetic conservation of the interdependent homeostatic relationship of sleep regulation and redox metabolism. J Comp Physiol B 2024; 194:241-252. [PMID: 38324048 PMCID: PMC11233307 DOI: 10.1007/s00360-023-01530-4] [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: 07/28/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 02/08/2024]
Abstract
Sleep is an essential and evolutionarily conserved process that affects many biological functions that are also strongly regulated by cellular metabolism. The interdependence between sleep homeostasis and redox metabolism, in particular, is such that sleep deprivation causes redox metabolic imbalances in the form of over-production of ROS. Likewise (and vice versa), accumulation of ROS leads to greater sleep pressure. Thus, it is theorized that one of the functions of sleep is to act as the brain's "antioxidant" at night by clearing oxidation built up from daily stress of the active day phase. In this review, we will highlight evidence linking sleep homeostasis and regulation to redox metabolism by discussing (1) the bipartite role that sleep-wake neuropeptides and hormones have in redox metabolism through comparing cross-species cellular and molecular mechanisms, (2) the evolutionarily metabolic changes that accompanied the development of sleep loss in cavefish, and finally, (3) some of the challenges of uncovering the cellular mechanism underpinning how ROS accumulation builds sleep pressure and cellularly, how this pressure is cleared.
Collapse
Affiliation(s)
- Aslihan Terzi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Keri J Ngo
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Department of Developmental Biology, 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.
| |
Collapse
|
11
|
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: 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: 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
|
12
|
Suppermpool A, Lyons DG, Broom E, Rihel J. Sleep pressure modulates single-neuron synapse number in zebrafish. Nature 2024; 629:639-645. [PMID: 38693264 PMCID: PMC11096099 DOI: 10.1038/s41586-024-07367-3] [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: 11/14/2022] [Accepted: 03/27/2024] [Indexed: 05/03/2024]
Abstract
Sleep is a nearly universal behaviour with unclear functions1. The synaptic homeostasis hypothesis proposes that sleep is required to renormalize the increases in synaptic number and strength that occur during wakefulness2. Some studies examining either large neuronal populations3 or small patches of dendrites4 have found evidence consistent with the synaptic homeostasis hypothesis, but whether sleep merely functions as a permissive state or actively promotes synaptic downregulation at the scale of whole neurons is unclear. Here, by repeatedly imaging all excitatory synapses on single neurons across sleep-wake states of zebrafish larvae, we show that synapses are gained during periods of wake (either spontaneous or forced) and lost during sleep in a neuron-subtype-dependent manner. However, synapse loss is greatest during sleep associated with high sleep pressure after prolonged wakefulness, and lowest in the latter half of an undisrupted night. Conversely, sleep induced pharmacologically during periods of low sleep pressure is insufficient to trigger synapse loss unless adenosine levels are boosted while noradrenergic tone is inhibited. We conclude that sleep-dependent synapse loss is regulated by sleep pressure at the level of the single neuron and that not all sleep periods are equally capable of fulfilling the functions of synaptic homeostasis.
Collapse
Affiliation(s)
- Anya Suppermpool
- Department of Cell and Developmental Biology, University College London, London, UK
- UCL Ear Institute, University College London, London, UK
| | - Declan G Lyons
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Elizabeth Broom
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College London, London, UK.
| |
Collapse
|
13
|
Cowen MH, Raizen DM, Hart MP. Structural neuroplasticity after sleep loss modifies behavior and requires neurexin and neuroligin. iScience 2024; 27:109477. [PMID: 38551003 PMCID: PMC10973677 DOI: 10.1016/j.isci.2024.109477] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/05/2023] [Accepted: 03/08/2024] [Indexed: 02/08/2025] Open
Abstract
Structural neuroplasticity (changes in the size, strength, number, and targets of synaptic connections) can be modified by sleep and sleep disruption. However, the causal relationships between genetic perturbations, sleep loss, neuroplasticity, and behavior remain unclear. The C. elegans GABAergic DVB neuron undergoes structural plasticity in adult males in response to adolescent stress, which rewires synaptic connections, alters behavior, and is dependent on conserved autism-associated genes NRXN1/nrx-1 and NLGN3/nlg-1. We find that four methods of sleep deprivation transiently induce DVB neurite extension in day 1 adults and increase the time to spicule protraction, which is the functional and behavioral output of the DVB neuron. Loss of nrx-1 and nlg-1 prevent DVB structural plasticity and behavioral changes at day 1 caused by adolescent sleep loss. Therefore, nrx-1 and nlg-1 mediate the morphologic and behavioral consequences of sleep loss, providing insight into the relationship between sleep, neuroplasticity, behavior, and neurologic disease.
Collapse
Affiliation(s)
- Mara H. Cowen
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David M. Raizen
- Department of Neurology and the Chronobiology and Sleep Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Michael P. Hart
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Weiss JT, Blundell MZ, Singh P, Donlea JM. Sleep deprivation drives brain-wide changes in cholinergic presynapse abundance in Drosophila melanogaster. Proc Natl Acad Sci U S A 2024; 121:e2312664121. [PMID: 38498719 PMCID: PMC10990117 DOI: 10.1073/pnas.2312664121] [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: 07/24/2023] [Accepted: 02/19/2024] [Indexed: 03/20/2024] Open
Abstract
Sleep is an evolutionarily conserved state that supports brain functions, including synaptic plasticity, in species across the animal kingdom. Here, we examine the neuroanatomical and cell-type distribution of presynaptic scaling in the fly brain after sleep loss. We previously found that sleep loss drives accumulation of the active zone scaffolding protein Bruchpilot (BRP) within cholinergic Kenyon cells of the Drosophila melanogaster mushroom body (MB), but not in other classes of MB neurons. To test whether similar cell type-specific trends in plasticity occur broadly across the brain, we used a flp-based genetic reporter to label presynaptic BRP in cholinergic, dopaminergic, GABAergic, or glutamatergic neurons. We then collected whole-brain confocal image stacks of BRP intensity to systematically quantify BRP, a marker of presynapse abundance, across 37 neuropil regions of the central fly brain. Our results indicate that sleep loss, either by overnight (12-h) mechanical stimulation or chronic sleep disruption in insomniac mutants, broadly elevates cholinergic synapse abundance across the brain, while synapse abundance in neurons that produce other neurotransmitters undergoes weaker, if any, changes. Extending sleep deprivation to 24 h drives brain-wide upscaling in glutamatergic, but not other, synapses. Finally, overnight male-male social pairings induce increased BRP in excitatory synapses despite male-female pairings eliciting more waking activity, suggesting experience-specific plasticity. Within neurotransmitter class and waking context, BRP changes are similar across the 37 neuropil domains, indicating that similar synaptic scaling rules may apply across the brain during acute sleep loss and that sleep need may broadly alter excitatory-inhibitory balance in the central brain.
Collapse
Affiliation(s)
- Jacqueline T. Weiss
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, CA90095
- Neuroscience Interdepartmental Program, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Mei Z. Blundell
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, CA90095
| | - Prabhjit Singh
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, CA90095
| | - Jeffrey M. Donlea
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, CA90095
| |
Collapse
|
15
|
Gottesman RF, Lutsey PL, Benveniste H, Brown DL, Full KM, Lee JM, Osorio RS, Pase MP, Redeker NS, Redline S, Spira AP. Impact of Sleep Disorders and Disturbed Sleep on Brain Health: A Scientific Statement From the American Heart Association. Stroke 2024; 55:e61-e76. [PMID: 38235581 DOI: 10.1161/str.0000000000000453] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Accumulating evidence supports a link between sleep disorders, disturbed sleep, and adverse brain health, ranging from stroke to subclinical cerebrovascular disease to cognitive outcomes, including the development of Alzheimer disease and Alzheimer disease-related dementias. Sleep disorders such as sleep-disordered breathing (eg, obstructive sleep apnea), and other sleep disturbances, as well, some of which are also considered sleep disorders (eg, insomnia, sleep fragmentation, circadian rhythm disorders, and extreme sleep duration), have been associated with adverse brain health. Understanding the causal role of sleep disorders and disturbances in the development of adverse brain health is complicated by the common development of sleep disorders among individuals with neurodegenerative disease. In addition to the role of sleep disorders in stroke and cerebrovascular injury, mechanistic hypotheses linking sleep with brain health and biomarker data (blood-based, cerebrospinal fluid-based, and imaging) suggest direct links to Alzheimer disease-specific pathology. These potential mechanisms and the increasing understanding of the "glymphatic system," and the recognition of the importance of sleep in poststroke recovery, as well, support a biological basis for the indirect (through the worsening of vascular disease) and direct (through specific effects on neuropathology) connections between sleep disorders and brain health. Given promising evidence for the benefits of treatment and prevention, sleep disorders and disturbances represent potential targets for early treatment that may improve brain health more broadly. In this scientific statement, we discuss the evidence supporting an association between sleep disorders and disturbances and poor brain health ranging from stroke to dementia and opportunities for prevention and early treatment.
Collapse
|
16
|
Pan D, Wang Z, Chen Y, Cao J. Melanopsin-mediated optical entrainment regulates circadian rhythms in vertebrates. Commun Biol 2023; 6:1054. [PMID: 37853054 PMCID: PMC10584931 DOI: 10.1038/s42003-023-05432-7] [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: 07/08/2023] [Accepted: 10/09/2023] [Indexed: 10/20/2023] Open
Abstract
Melanopsin (OPN4) is a light-sensitive protein that plays a vital role in the regulation of circadian rhythms and other nonvisual functions. Current research on OPN4 has focused on mammals; more evidence is needed from non-mammalian vertebrates to fully assess the significance of the non-visual photosensitization of OPN4 for circadian rhythm regulation. There are species differences in the regulatory mechanisms of OPN4 for vertebrate circadian rhythms, which may be due to the differences in the cutting variants, tissue localization, and photosensitive activation pathway of OPN4. We here summarize the distribution of OPN4 in mammals, birds, and teleost fish, and the classical excitation mode for the non-visual photosensitive function of OPN4 in mammals is discussed. In addition, the role of OPN4-expressing cells in regulating circadian rhythm in different vertebrates is highlighted, and the potential rhythmic regulatory effects of various neuropeptides or neurotransmitters expressed in mammalian OPN4-expressing ganglion cells are summarized among them.
Collapse
Affiliation(s)
- Deng Pan
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Zixu Wang
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Yaoxing Chen
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China
| | - Jing Cao
- Laboratory of Anatomy of Domestic Animals, National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Haidian, 100193, Beijing, China.
| |
Collapse
|
17
|
Roh SE, Xiao M, Delgado A, Kwak C, Savonenko A, Bakker A, Kwon HB, Worley P. Sleep and circadian rhythm disruption by NPTX2 loss of function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.26.559408. [PMID: 37808783 PMCID: PMC10557648 DOI: 10.1101/2023.09.26.559408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Sleep and circadian rhythm disruption (SCRD) is commonly observed in aging, especially in individuals who experience progressive cognitive decline to mild cognitive impairment (MCI) and Alzheimer's disease (AD). However, precise molecular mechanisms underlying the association between SCRD and aging are not fully understood. Orexin A is a well-characterized "sleep neuropeptide" that is expressed in hypothalamic neurons and evokes wake behavior. The importance of Orexin is exemplified in narcolepsy where it is profoundly down-regulated. Interestingly, the synaptic immediate early gene NPTX2 is co-expressed in Orexin neurons and is similarly reduced in narcolepsy. NPTX2 is also down-regulated in CSF of some cognitively normal older individuals and predicts the time of transition from normal cognition to MCI. The association between Orexin and NPTX2 is further evinced here where we observe that Orexin A and NPTX2 are highly correlated in CSF of cognitively normal aged individuals and raises the question of whether SCRD that are typically attributed to Orexin A loss of function may be modified by concomitant NPTX2 down-regulation. Is NPTX2 an effector of sleep or simply a reporter of orexin-dependent SCRD? To address this question, we examined NPTX2 KO mice and found they retain Orexin expression in the brain and so provide an opportunity to examine the specific contribution of NPTX2 to SCRD. Our results reveal that NPTX2 KO mice exhibit a disrupted circadian onset time, coupled with increased activity during the sleep phase, suggesting difficulties in maintaining states. Sleep EEG indicates distinct temporal allocation shifts across vigilance states, characterized by reduced wake and increased NREM time. Evident sleep fragmentation manifests through alterations of event occurrences during Wake and NREM, notably during light transition periods, in conjunction with an increased frequency of sleep transitions in NPTX2 KO mice, particularly between Wake and NREM. EEG spectral analysis indicated significant shifts in power across various frequency bands in the wake, NREM, and REM states, suggestive of disrupted neuronal synchronicity. An intriguing observation is the diminished occurrence of sleep spindles, one of the earliest measures of human sleep disruption, in NPTX2 KO mice. These findings highlight the effector role of NPTX2 loss of function as an instigator of SCRD and a potential mediator of sleep disruption in aging.
Collapse
Affiliation(s)
- Seung-Eon Roh
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Meifang Xiao
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ana Delgado
- Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chuljung Kwak
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alena Savonenko
- Department of Neuroanatomy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Arnold Bakker
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hyung-Bae Kwon
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Paul Worley
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
18
|
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: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
19
|
Du XF, Li FN, Peng XL, Xu B, Zhang Y, Li G, Liu T, Li Y, Wang H, Yan J, Du JL. Circadian regulation of developmental synaptogenesis via the hypocretinergic system. Nat Commun 2023; 14:3195. [PMID: 37268623 DOI: 10.1038/s41467-023-38973-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/24/2023] [Indexed: 06/04/2023] Open
Abstract
The circadian clock orchestrates a wide variety of physiological and behavioral processes, enabling animals to adapt to daily environmental changes, particularly the day-night cycle. However, the circadian clock's role in the developmental processes remains unclear. Here, we employ the in vivo long-term time-lapse imaging of retinotectal synapses in the optic tectum of larval zebrafish and reveal that synaptogenesis, a fundamental developmental process for neural circuit formation, exhibits circadian rhythm. This rhythmicity arises primarily from the synapse formation rather than elimination and requires the hypocretinergic neural system. Disruption of this synaptogenic rhythm, by impairing either the circadian clock or the hypocretinergic system, affects the arrangement of the retinotectal synapses on axon arbors and the refinement of the postsynaptic tectal neuron's receptive field. Thus, our findings demonstrate that the developmental synaptogenesis is under hypocretin-dependent circadian regulation, suggesting an important role of the circadian clock in neural development.
Collapse
Affiliation(s)
- Xu-Fei Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China.
- University of Chinese Academy of Sciences, 19A Yu-Quan Road, 100049, Beijing, China.
| | - Fu-Ning Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 19A Yu-Quan Road, 100049, Beijing, China
| | - Xiao-Lan Peng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Bing Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Yu Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Guang Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Taole Liu
- Center for Circadian Clocks, Soochow University, 215123, Suzhou, Jiangsu, China
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Ying Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, 215123, Suzhou, Jiangsu, China
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Jun Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 19A Yu-Quan Road, 100049, Beijing, China
| | - Jiu-Lin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China.
- University of Chinese Academy of Sciences, 19A Yu-Quan Road, 100049, Beijing, China.
- School of Life Science and Technology, ShanghaiTech University, 319 Yue-Yang Road, 200031, Shanghai, China.
| |
Collapse
|
20
|
Salehinejad MA, Azarkolah A, Ghanavati E, Nitsche MA. Circadian disturbances, sleep difficulties and the COVID-19 pandemic. Sleep Med 2022; 91:246-252. [PMID: 34334305 PMCID: PMC8277544 DOI: 10.1016/j.sleep.2021.07.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/12/2021] [Accepted: 07/07/2021] [Indexed: 12/24/2022]
Abstract
The COVID-19 pandemic has imposed extraordinary and unpredictable changes on our lifestyle for an unknown duration. Consequently, core aspects of wellbeing including behavior, emotion, cognition, and social interactions are negatively affected. Sleep and circadian rhythms, with an extensive impact on physiology, behavior, emotion, and cognition are affected too. We provided an updated overview of the impact of the COVID-19 pandemic on circadian rhythms and sleep based on the results of published studies (n = 48) in three sections. First, we focus on circadian misalignment due to the pandemic in the general population (including shift workers, health staff, students) and COVID-19 patients and summarize the most critically contributing factors to circadian misalignment. Next, we address sleep difficulties and poor sleep quality during the pandemic, their contributing factors, rate and prevalence, and their effects on both the general population and COVID-19 patients. Finally, we summarize the currently applied/recommended interventions for aligning circadian rhythms and improving sleep quality in both, the general population, and COVID-19 patients during the pandemic situation. Briefly, circadian misalignment and sleep difficulties are common consequences of the pandemic in the general population (with elderly, students, children, health and night-work shifters as risk groups) and COVID-19 patients. Home confinement and its physiological, circadian, and psychological derivates are central to these difficulties. Symptoms severity, treatment progress, recovery duration, and even diagnosis of COVID-19 patients are considerably affected by circadian and sleep difficulties. Behavioral interventions for normalizing the factors that contribute to circadian and sleep difficulties are helpful.
Collapse
Affiliation(s)
- Mohammad Ali Salehinejad
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany.
| | - Anita Azarkolah
- Department of Psychiatry, Fatemi Hospital, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Ghanavati
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Michael A Nitsche
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany; Department of Neurology, University Medical Hospital Bergmannsheil, Bochum, Germany
| |
Collapse
|
21
|
Lee DA, Oikonomou G, Prober DA. Large-scale Analysis of Sleep in Zebrafish. Bio Protoc 2022; 12:e4313. [PMID: 35284597 PMCID: PMC8855086 DOI: 10.21769/bioprotoc.4313] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/18/2021] [Accepted: 12/01/2021] [Indexed: 03/20/2025] Open
Abstract
Over the past decade, zebrafish have emerged as a powerful model for the study of vertebrate sleep and wake behaviors. Experimental evidence has demonstrated behavioral, anatomical, genetic, and pharmacological conservation of sleep between zebrafish and mammals, suggesting that discoveries in zebrafish can inform our understanding of mammalian sleep. Here, we describe a protocol for performing sleep behavioral experiments in larval zebrafish, using a high-throughput video tracking system. We explain how to set up a sleep behavioral experiment and provide guidelines on how to analyze the data. Using this protocol, a typical experiment can be completed in less than five days, and this method provides a scalable platform to perform genetic and pharmacological screens in a simple and cost-effective vertebrate model. By combining high-throughput behavioral assays with several advantageous features of zebrafish, this model system provides new opportunities to make discoveries that clarify the genetic and neurological mechanisms that regulate sleep.
Collapse
Affiliation(s)
- Daniel A. Lee
- Division of Biology and Biological Engineering, Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of Technology, Pasadena, California, United States of America
| | - Grigorios Oikonomou
- Division of Biology and Biological Engineering, Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of Technology, Pasadena, California, United States of America
| | - David A. Prober
- Division of Biology and Biological Engineering, Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of Technology, Pasadena, California, United States of America
| |
Collapse
|
22
|
Jaggard JB, Wang GX, Mourrain P. Non-REM and REM/paradoxical sleep dynamics across phylogeny. Curr Opin Neurobiol 2021; 71:44-51. [PMID: 34583217 PMCID: PMC8719594 DOI: 10.1016/j.conb.2021.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022]
Abstract
All animals carefully studied sleep, suggesting that sleep as a behavioral state exists in all animal life. Such evolutionary maintenance of an otherwise vulnerable period of environmental detachment suggests that sleep must be integral in fundamental biological needs. Despite over a century of research, the knowledge of what sleep does at the tissue, cellular or molecular levels remain cursory. Currently, sleep is defined based on behavioral criteria and physiological measures rather than at the cellular or molecular level. Physiologically, sleep has been described as two main states, non-rapid eye moment (NREM) and REM/paradoxical sleep (PS), which are defined in the neocortex by synchronous oscillations and paradoxical wake-like activity, respectively. For decades, these two sleep states were believed to be defining characteristics of only mammalian and avian sleep. Recent work has revealed slow oscillation, silencing, and paradoxical/REM-like activities in reptiles, fish, flies, worms, and cephalopods suggesting that these sleep dynamics and associated physiological states may have emerged early in animal evolution. Here, we discuss these recent developments supporting the conservation of neural dynamics (silencing, oscillation, paradoxical activity) of sleep states across phylogeny.
Collapse
Affiliation(s)
- James B Jaggard
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Gordon X Wang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Wu Tsai Neuroscience Institute, 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.
| |
Collapse
|
23
|
Mack KL, Jaggard JB, Persons JL, Roback EY, Passow CN, Stanhope BA, Ferrufino E, Tsuchiya D, Smith SE, Slaughter BD, Kowalko J, Rohner N, Keene AC, McGaugh SE. Repeated evolution of circadian clock dysregulation in cavefish populations. PLoS Genet 2021; 17:e1009642. [PMID: 34252077 PMCID: PMC8297936 DOI: 10.1371/journal.pgen.1009642] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 07/22/2021] [Accepted: 06/03/2021] [Indexed: 11/18/2022] Open
Abstract
Circadian rhythms are nearly ubiquitous throughout nature, suggesting they are critical for survival in diverse environments. Organisms inhabiting largely arrhythmic environments, such as caves, offer a unique opportunity to study the evolution of circadian rhythms in response to changing ecological pressures. Populations of the Mexican tetra, Astyanax mexicanus, have repeatedly invaded caves from surface rivers, where individuals must contend with perpetual darkness, reduced food availability, and limited fluctuations in daily environmental cues. To investigate the molecular basis for evolved changes in circadian rhythms, we investigated rhythmic transcription across multiple independently-evolved cavefish populations. Our findings reveal that evolution in a cave environment has led to the repeated disruption of the endogenous biological clock, and its entrainment by light. The circadian transcriptome shows widespread reductions and losses of rhythmic transcription and changes to the timing of the activation/repression of core-transcriptional clock. In addition to dysregulation of the core clock, we find that rhythmic transcription of the melatonin regulator aanat2 and melatonin rhythms are disrupted in cavefish under darkness. Mutants of aanat2 and core clock gene rorca disrupt diurnal regulation of sleep in A. mexicanus, phenocopying circadian modulation of sleep and activity phenotypes of cave populations. Together, these findings reveal multiple independent mechanisms for loss of circadian rhythms in cavefish populations and provide a platform for studying how evolved changes in the biological clock can contribute to variation in sleep and circadian behavior.
Collapse
Affiliation(s)
- Katya L. Mack
- Biology, Stanford University, Stanford, California, United States of America
| | - James B. Jaggard
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida, United States of America
- Center for Sleep Sciences and Medicine, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California, United States of America
| | - Jenna L. Persons
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Emma Y. Roback
- Ecology, Evolution, and Behavior, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Courtney N. Passow
- Ecology, Evolution, and Behavior, University of Minnesota, Saint Paul, Minnesota, United States of America
| | - Bethany A. Stanhope
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida, United States of America
| | - Estephany Ferrufino
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida, United States of America
- Wilkes Honors College, Florida Atlantic University, Jupiter, Florida, United States of America
| | - Dai Tsuchiya
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Sarah E. Smith
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Brian D. Slaughter
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Johanna Kowalko
- Wilkes Honors College, Florida Atlantic University, Jupiter, Florida, United States of America
| | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Alex C. Keene
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida, United States of America
| | - Suzanne E. McGaugh
- Ecology, Evolution, and Behavior, University of Minnesota, Saint Paul, Minnesota, United States of America
| |
Collapse
|
24
|
Chen W, Xie L, Yu F, Li Y, Chen C, Xie W, Huang T, Zhang Y, Zhang S, Li P. Zebrafish as a Model for In-Depth Mechanistic Study for Stroke. Transl Stroke Res 2021; 12:695-710. [PMID: 34050491 DOI: 10.1007/s12975-021-00907-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/22/2022]
Abstract
Stroke is one of the world's leading causes of death and disability, posing enormous burden to the society. However, the pathogenesis and mechanisms that underlie brain injury and brain repair remain largely unknown. There's an unmet need of in-depth mechanistic research in this field. Zebrafish (Danio rerio) is a powerful tool in brain science research mainly due to its small size and transparent body, high genome synteny with human, and similar nervous system structures. It can be used to establish both hemorrhagic and ischemic stroke models easily and effectively through different ways. After the establishment of stroke model, research methods including behavioral test, in vivo imaging, and drug screening are available to explore mechanisms that underlie the brain injury and brain repair after stroke. This review focuses on the advantages and the feasibility of zebrafish stroke model, and will also introduce the key methods available for stroke studies in zebrafish, which may drive future mechanistic studies in the pursuit of discovering novel therapeutic targets for stroke patients.
Collapse
Affiliation(s)
- Weijie Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Lv Xie
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Fang Yu
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Yan Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Chen Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Wanqing Xie
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Tingting Huang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Yueman Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Song Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China.
| | - Peiying Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China.
| |
Collapse
|
25
|
Yan YL, Titus T, Desvignes T, BreMiller R, Batzel P, Sydes J, Farnsworth D, Dillon D, Wegner J, Phillips JB, Peirce J, Dowd J, Undiagnosed Diseases Network, Buck CL, Miller A, Westerfield M, Postlethwait JH. A fish with no sex: gonadal and adrenal functions partition between zebrafish NR5A1 co-orthologs. Genetics 2021; 217:iyaa030. [PMID: 33724412 PMCID: PMC8045690 DOI: 10.1093/genetics/iyaa030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
People with NR5A1 mutations experience testicular dysgenesis, ovotestes, or adrenal insufficiency, but we do not completely understand the origin of this phenotypic diversity. NR5A1 is expressed in gonadal soma precursor cells before expression of the sex-determining gene SRY. Many fish have two co-orthologs of NR5A1 that likely partitioned ancestral gene subfunctions between them. To explore ancestral roles of NR5A1, we knocked out nr5a1a and nr5a1b in zebrafish. Single-cell RNA-seq identified nr5a1a-expressing cells that co-expressed genes for steroid biosynthesis and the chemokine receptor Cxcl12a in 1-day postfertilization (dpf) embryos, as does the mammalian adrenal-gonadal (interrenal-gonadal) primordium. In 2dpf embryos, nr5a1a was expressed stronger in the interrenal-gonadal primordium than in the early hypothalamus but nr5a1b showed the reverse. Adult Leydig cells expressed both ohnologs and granulosa cells expressed nr5a1a stronger than nr5a1b. Mutants for nr5a1a lacked the interrenal, formed incompletely differentiated testes, had no Leydig cells, and grew far larger than normal fish. Mutants for nr5a1b formed a disorganized interrenal and their gonads completely disappeared. All homozygous mutant genotypes lacked secondary sex characteristics, including male breeding tubercles and female sex papillae, and had exceedingly low levels of estradiol, 11-ketotestosterone, and cortisol. RNA-seq showed that at 21dpf, some animals were developing as females and others were not, independent of nr5a1 genotype. By 35dpf, all mutant genotypes greatly under-expressed ovary-biased genes. Because adult nr5a1a mutants form gonads but lack an interrenal and conversely, adult nr5a1b mutants lack a gonad but have an interrenal, the adrenal, and gonadal functions of the ancestral nr5a1 gene partitioned between ohnologs after the teleost genome duplication, likely owing to reciprocal loss of ancestral tissue-specific regulatory elements. Identifying such elements could provide hints to otherwise unexplained cases of Differences in Sex Development.
Collapse
Affiliation(s)
- Yi-Lin Yan
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | - Tom Titus
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | - Thomas Desvignes
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | - Ruth BreMiller
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | - Peter Batzel
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | - Jason Sydes
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | - Dylan Farnsworth
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | - Danielle Dillon
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Jeremy Wegner
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | | | - Judy Peirce
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | - John Dowd
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | | | - Charles Loren Buck
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Adam Miller
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | - Monte Westerfield
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | | |
Collapse
|
26
|
Sagi D, de Lecea L, Appelbaum L. Heterogeneity of Hypocretin/Orexin Neurons. FRONTIERS OF NEUROLOGY AND NEUROSCIENCE 2021; 45:61-74. [PMID: 34052814 PMCID: PMC8961008 DOI: 10.1159/000514964] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 02/02/2021] [Indexed: 01/21/2023]
Abstract
The multifunctional, hypothalamic hypocretin/orexin (HCRT)-producing neurons regulate an array of physiological and behavioral states including arousal, sleep, feeding, emotions, stress, and reward. How a presumably uniform HCRT neuron population regulates such a diverse set of functions is not clear. The role of the HCRT neuropeptides may vary depending on the timing and localization of secretion and neuronal activity. Moreover, HCRT neuropeptides may not mediate all functions ascribed to HCRT neurons. Some could be orchestrated by additional neurotransmitters and neuropeptides that are expressed in HCRT neurons. We hypothesize that HCRT neurons are segregated into genetically, anatomically and functionally distinct subpopulations. We discuss accumulating data that suggest the existence of such HCRT neuron subpopulations that may effectuate the diverse functions of these neurons in mammals and fish.
Collapse
Affiliation(s)
- Dana Sagi
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Luis de Lecea
- Dept of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Lior Appelbaum
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel.,Corresponding author: Lior Appelbaum, Bar-Ilan University, Ramat-Gan 5290002, Israel. Telephone: +972-3-7384536,
| |
Collapse
|
27
|
Hartsock MJ, Spencer RL. Memory and the circadian system: Identifying candidate mechanisms by which local clocks in the brain may regulate synaptic plasticity. Neurosci Biobehav Rev 2020; 118:134-162. [PMID: 32712278 DOI: 10.1016/j.neubiorev.2020.07.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 07/14/2020] [Accepted: 07/18/2020] [Indexed: 12/11/2022]
Abstract
The circadian system is an endogenous biological network responsible for coordinating near-24-h cycles in behavior and physiology with daily timing cues from the external environment. In this review, we explore how the circadian system regulates memory formation, retention, and recall. Circadian rhythms in these memory processes may arise through several endogenous pathways, and recent work highlights the importance of genetic timekeepers found locally within tissues, called local clocks. We evaluate the circadian memory literature for evidence of local clock involvement in memory, identifying potential nodes for direct interactions between local clock components and mechanisms of synaptic plasticity. Our discussion illustrates how local clocks may pervasively modulate neuronal plastic capacity, a phenomenon that we designate here as circadian metaplasticity. We suggest that this function of local clocks supports the temporal optimization of memory processes, illuminating the potential for circadian therapeutic strategies in the prevention and treatment of memory impairment.
Collapse
Affiliation(s)
- Matthew J Hartsock
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado 80309, United States.
| | - Robert L Spencer
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado 80309, United States.
| |
Collapse
|
28
|
Abstract
Sleep-dependent synaptic plasticity is crucial for optimal cognition. However, establishing the direction of synaptic plasticity during sleep has been particularly challenging since data in support of both synaptic potentiation and depotentiation have been reported. This review focuses on structural synaptic plasticity across sleep and wake and summarizes recent developments in the use of 3-dimensional electron microscopy as applied to this field.
Collapse
Affiliation(s)
- Michele Bellesi
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, BS8 1TD Bristol, UK
| | - Luisa de Vivo
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, BS8 1TD Bristol, UK
| |
Collapse
|
29
|
Coupling Neuropeptide Levels to Structural Plasticity in Drosophila Clock Neurons. Curr Biol 2020; 30:3154-3166.e4. [PMID: 32619484 DOI: 10.1016/j.cub.2020.06.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/05/2020] [Accepted: 06/02/2020] [Indexed: 12/24/2022]
Abstract
We have previously reported that pigment dispersing factor (PDF) neurons, which are essential in the control of rest-activity cycles in Drosophila, undergo circadian remodeling of their axonal projections, a phenomenon called circadian structural plasticity. Axonal arborizations display higher complexity during the day and become simpler at night, and this remodeling involves changes in the degree of connectivity. This phenomenon depends on the clock present within the ventrolateral neurons (LNvs) as well as in glia. In this work, we characterize in detail the contribution of the PDF neuropeptide to structural plasticity at different times across the day. Using diverse genetic strategies to temporally restrict its downregulation, we demonstrate that even subtle alterations to PDF cycling at the dorsal protocerebrum correlate with impaired remodeling, underscoring its relevance for the characteristic morning spread; PDF released from the small LNvs (sLNvs) and the large LNvs (lLNvs) contribute to the process. Moreover, forced depolarization recruits activity-dependent mechanisms to mediate growth only at night, overcoming the restriction imposed by the clock on membrane excitability. Interestingly, the active process of terminal remodeling requires PDF receptor (PDFR) signaling acting locally through the cyclic-nucleotide-gated channel ion channel subunit A (CNGA). Thus, clock-dependent PDF signaling shapes the connectivity of these essential clock neurons on daily basis.
Collapse
|
30
|
Wang H, Yang Z, Li X, Huang D, Yu S, He J, Li Y, Yan J. Single-cell in vivo imaging of cellular circadian oscillators in zebrafish. PLoS Biol 2020; 18:e3000435. [PMID: 32168317 PMCID: PMC7069618 DOI: 10.1371/journal.pbio.3000435] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 02/10/2020] [Indexed: 12/22/2022] Open
Abstract
The circadian clock is a cell-autonomous time-keeping mechanism established gradually during embryonic development. Here, we generated a transgenic zebrafish line carrying a destabilized fluorescent protein driven by the promoter of a core clock gene, nr1d1, to report in vivo circadian rhythm at the single-cell level. By time-lapse imaging of this fish line and 3D reconstruction, we observed the sequential initiation of the reporter expression starting at photoreceptors in the pineal gland, then spreading to the cells in other brain regions at the single-cell level. Even within the pineal gland, we found heterogeneous onset of nr1d1 expression, in which each cell undergoes circadian oscillation superimposed over a cell type–specific developmental trajectory. Furthermore, we found that single-cell expression of nr1d1 showed synchronous circadian oscillation under a light–dark (LD) cycle. Remarkably, single-cell oscillations were dramatically dampened rather than desynchronized in animals raised under constant darkness, while the developmental trend still persists. It suggests that light exposure in early zebrafish embryos has significant effect on cellular circadian oscillations. A transgenic zebrafish line, nr1d1-VNP, enables the monitoring of single-cell circadian rhythms in live zebrafish; using this fish line, the authors find that light exposure in early development initializes rather than synchronizes single-cell oscillators.
Collapse
Affiliation(s)
- Haifang Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Zeyong Yang
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
| | - Xingxing Li
- Department of Anesthesiology, First Affiliated Hospital of AnHui Medical University, Hefei, Anhui, China
| | - Dengfeng Huang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Shuguang Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Jie He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
- * E-mail: (JY); (YL); (JH)
| | - Yuanhai Li
- Department of Anesthesiology, First Affiliated Hospital of AnHui Medical University, Hefei, Anhui, China
- * E-mail: (JY); (YL); (JH)
| | - Jun Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, China
- * E-mail: (JY); (YL); (JH)
| |
Collapse
|
31
|
Abstract
The scientific community has searched for years for ways of examining neuronal tissue to track neural activity with reliable anatomical markers for stimulated neuronal activity. Existing studies that focused on hypothalamic systems offer a few options but do not always compare approaches or validate them for dependence on cell firing, leaving the reader uncertain of the benefits and limitations of each method. Thus, in this article, potential markers will be presented and, where possible, placed into perspective in terms of when and how these methods pertain to hypothalamic function. An example of each approach is included. In reviewing the approaches, one is guided through how neurons work, the consequences of their stimulation, and then the potential markers that could be applied to hypothalamic systems are discussed. Approaches will use features of neuronal glucose utilization, water/oxygen movement, changes in neuron-glial interactions, receptor translocation, cytoskeletal changes, stimulus-synthesis coupling that includes expression of the heteronuclear or mature mRNA for transmitters or the enzymes that make them, and changes in transcription factors (immediate early gene products, precursor buildup, use of promoter-driven surrogate proteins, and induced expression of added transmitters. This article includes discussion of methodological limitations and the power of combining approaches to understand neuronal function. © 2020 American Physiological Society. Compr Physiol 10:549-575, 2020.
Collapse
Affiliation(s)
- Gloria E. Hoffman
- Department of Biology, Morgan State University, Baltimore, Maryland, USA
| |
Collapse
|
32
|
Wang F, Ren D, Liang X, Ke S, Zhang B, Hu B, Song X, Wang X. A long noncoding RNA cluster-based genomic locus maintains proper development and visual function. Nucleic Acids Res 2020; 47:6315-6329. [PMID: 31127312 PMCID: PMC6614851 DOI: 10.1093/nar/gkz444] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 04/29/2019] [Accepted: 05/10/2019] [Indexed: 01/07/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) represent a group of regulatory RNAs that play critical roles in numerous cellular events, but their functional importance in development remains largely unexplored. Here, we discovered a series of previously unidentified gene clusters harboring conserved lncRNAs at the nonimprinting regions in brain (CNIBs). Among the seven identified CNIBs, human CNIB1 locus is located at Chr 9q33.3 and conserved from Danio rerio to Homo sapiens. Chr 9q33.3-9q34.11 microdeletion has previously been linked to human nail-patella syndrome (NPS) which is frequently accompanied by developmental and visual deficiencies. By generating CNIB1 deletion alleles in zebrafish, we demonstrated the requirement of CNIB1 for proper growth and development, and visual activities. Furthermore, we found that the role of CNIB1 on visual activity is mediated through a regulator of ocular development-lmx1bb. Collectively, our study shows that CNIB1 lncRNAs are important for zebrafish development and provides an lncRNA cluster-mediated pathophysiological mechanism for human Chr 9q33.3-9q34.11 microdeletion syndrome.
Collapse
Affiliation(s)
- Fei Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Dalong Ren
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xiaolin Liang
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Shengwei Ke
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Bowen Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Bing Hu
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xiaoyuan Song
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xiangting Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
33
|
Madaro A, Kristiansen TS, Pavlidis MA. How Fish Cope with Stress? Anim Welf 2020. [DOI: 10.1007/978-3-030-41675-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
34
|
Lombardo MV, Auyeung B, Pramparo T, Quartier A, Courraud J, Holt RJ, Waldman J, Ruigrok ANV, Mooney N, Bethlehem RAI, Lai MC, Kundu P, Bullmore ET, Mandel JL, Piton A, Baron-Cohen S. Sex-specific impact of prenatal androgens on social brain default mode subsystems. Mol Psychiatry 2020; 25:2175-2188. [PMID: 30104728 PMCID: PMC7473837 DOI: 10.1038/s41380-018-0198-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/14/2018] [Accepted: 07/09/2018] [Indexed: 01/04/2023]
Abstract
Early-onset neurodevelopmental conditions (e.g., autism) affect males more frequently than females. Androgens may play a role in this male-bias by sex-differentially impacting early prenatal brain development, particularly neural circuits that later develop specialized roles in social cognition. Here, we find that increasing prenatal testosterone in humans is associated with later reduction of functional connectivity between social brain default mode (DMN) subsystems in adolescent males, but has no effect in females. Since testosterone can work directly via the androgen receptor (AR) or indirectly via the estrogen receptor through aromatase conversion to estradiol, we further examined how a potent non-aromatizable androgen, dihydrotestosterone (DHT), acts via the AR to influence gene expression in human neural stem cells (hNSC)-particularly for genes of high-relevance for DMN circuitry. DHT dysregulates a number of genes enriched for syndromic causes of autism and intellectual disability and for genes that in later development are expressed in anatomical patterns that highly correspond to the cortical midline DMN subsystem. DMN-related and DHT-affected genes (e.g., MEF2C) are involved in a number of synaptic processes, many of which impact excitation-inhibition balance. Androgens have male-specific prenatal influence over social brain circuitry in humans and may be relevant towards explaining some component of male-bias in early-onset neurodevelopmental conditions.
Collapse
Affiliation(s)
- Michael V. Lombardo
- grid.6603.30000000121167908Center for Applied Neuroscience, Department of Psychology, University of Cyprus, Nicosia, Cyprus ,grid.5335.00000000121885934Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Bonnie Auyeung
- grid.5335.00000000121885934Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom ,grid.4305.20000 0004 1936 7988Department of Psychology, School of Philosophy, Psychology, and Language Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Tiziano Pramparo
- grid.266100.30000 0001 2107 4242Department of Neurosciences, University of California, San Diego, CA USA
| | - Angélique Quartier
- grid.420255.40000 0004 0638 2716Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France ,grid.4444.00000 0001 2112 9282Centre National de la Recherche Scientifique, UMR7104 Illkirch, France ,Institut National de la Santé et de la Recherche Médicale, U964 Illkirch, France ,grid.420255.40000 0004 0638 2716Université de Strasbourg, Illkirch, France
| | - Jérémie Courraud
- grid.420255.40000 0004 0638 2716Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France ,grid.4444.00000 0001 2112 9282Centre National de la Recherche Scientifique, UMR7104 Illkirch, France ,Institut National de la Santé et de la Recherche Médicale, U964 Illkirch, France ,grid.420255.40000 0004 0638 2716Université de Strasbourg, Illkirch, France
| | - Rosemary J. Holt
- grid.5335.00000000121885934Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Jack Waldman
- grid.5335.00000000121885934Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Amber N. V. Ruigrok
- grid.5335.00000000121885934Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Natasha Mooney
- grid.5335.00000000121885934Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Richard A. I. Bethlehem
- grid.5335.00000000121885934Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Meng-Chuan Lai
- grid.5335.00000000121885934Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom ,grid.17063.330000 0001 2157 2938Child and Youth Mental Health Collaborative, Centre for Addiction and Mental Health and the Hospital for Sick Children, Department of Psychiatry, University of Toronto, Toronto, ON Canada ,grid.412094.a0000 0004 0572 7815Department of Psychiatry, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Prantik Kundu
- grid.59734.3c0000 0001 0670 2351Section on Advanced Functional Neuroimaging, Departments of Radiology & Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Edward T. Bullmore
- grid.5335.00000000121885934Brain Mapping Unit, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom ,Cambridgeshire and Peterborough National Health Service Foundation Trust, Cambridge, United Kingdom ,grid.418236.a0000 0001 2162 0389ImmunoPsychiatry, GlaxoSmithKline Research and Development, Stevenage, United Kingdom
| | - Jean-Louis Mandel
- grid.420255.40000 0004 0638 2716Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France ,grid.4444.00000 0001 2112 9282Centre National de la Recherche Scientifique, UMR7104 Illkirch, France ,Institut National de la Santé et de la Recherche Médicale, U964 Illkirch, France ,grid.420255.40000 0004 0638 2716Université de Strasbourg, Illkirch, France ,grid.410533.00000 0001 2179 2236Chair of Human Genetics, Collège de France, Paris, France
| | - Amélie Piton
- grid.420255.40000 0004 0638 2716Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France ,grid.4444.00000 0001 2112 9282Centre National de la Recherche Scientifique, UMR7104 Illkirch, France ,Institut National de la Santé et de la Recherche Médicale, U964 Illkirch, France ,grid.420255.40000 0004 0638 2716Université de Strasbourg, Illkirch, France
| | - Simon Baron-Cohen
- grid.5335.00000000121885934Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom ,Cambridgeshire and Peterborough National Health Service Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
35
|
Abstract
Sleep durations vary greatly across animals from 2 to 20 hours with no clear explanation. A small Mexican cavefish reveals how the brain can adapt to increase its wake-stabilizing hypocretin circuit and dramatically reduce sleep, likely to allow adaptive foraging.
Collapse
Affiliation(s)
- Louis C Leung
- Department of Psychiatry and Behavioral Sciences, Stanford Center for Sleep Sciences and Medicine, Stanford, CA 94305, USA
| | - Philippe Mourrain
- Department of Psychiatry and Behavioral Sciences, Stanford Center for Sleep Sciences and Medicine, Stanford, CA 94305, USA; INSERM 1024, Ecole Normale Supérieure Paris, 75005, France.
| |
Collapse
|
36
|
Zhong Y, Ye Q, Chen C, Wang M, Wang H. Ezh2 promotes clock function and hematopoiesis independent of histone methyltransferase activity in zebrafish. Nucleic Acids Res 2019; 46:3382-3399. [PMID: 29447387 PMCID: PMC5909462 DOI: 10.1093/nar/gky101] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 02/06/2018] [Indexed: 12/13/2022] Open
Abstract
EZH2 is a subunit of polycomb repressive complex 2 (PRC2) that silences gene transcription via H3K27me3 and was shown to be essential for mammalian liver circadian regulation and hematopoiesis through gene silencing. Much less, however, is known about how Ezh2 acts in live zebrafish. Here, we show that zebrafish ezh2 is regulated directly by the circadian clock via both E-box and RORE motif, while core circadian clock genes per1a, per1b, cry1aa and cry1ab are down-regulated in ezh2 null mutant and ezh2 morphant zebrafish, and either knockdown or overexpression of ezh2 alters locomotor rhythms, indicating that Ezh2 is required for zebrafish circadian regulation. In contrast to its canonical silencing function, zebrafish Ezh2 up-regulates these key circadian clock genes independent of histone methyltransferase activity by directly binding to key circadian clock proteins. Similarly, Ezh2 contributes to hematopoiesis by enhancing expression of hematopoietic genes such as cmyb and lck. Together, our findings demonstrate for the first time that Ezh2 acts in both circadian regulation and hematopoiesis independent of silencing PRC2.
Collapse
Affiliation(s)
- Yingbin Zhong
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, PR China.,School of Biology & Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu, PR China
| | - Qiang Ye
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, PR China.,School of Biology & Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu, PR China
| | - Chengyan Chen
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, PR China.,School of Biology & Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu, PR China
| | - Mingyong Wang
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, PR China.,School of Biology & Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu, PR China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, PR China.,School of Biology & Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu, PR China
| |
Collapse
|
37
|
Abstract
Slow-wave sleep and rapid eye movement (or paradoxical) sleep have been found in mammals, birds and lizards, but it is unclear whether these neuronal signatures are found in non-amniotic vertebrates. Here we develop non-invasive fluorescence-based polysomnography for zebrafish, and show-using unbiased, brain-wide activity recording coupled with assessment of eye movement, muscle dynamics and heart rate-that there are at least two major sleep signatures in zebrafish. These signatures, which we term slow bursting sleep and propagating wave sleep, share commonalities with those of slow-wave sleep and paradoxical or rapid eye movement sleep, respectively. Further, we find that melanin-concentrating hormone signalling (which is involved in mammalian sleep) also regulates propagating wave sleep signatures and the overall amount of sleep in zebrafish, probably via activation of ependymal cells. These observations suggest that common neural signatures of sleep may have emerged in the vertebrate brain over 450 million years ago.
Collapse
|
38
|
Alzheimer's Disease and Sleep-Wake Disturbances: Amyloid, Astrocytes, and Animal Models. J Neurosci 2019; 38:2901-2910. [PMID: 29563238 DOI: 10.1523/jneurosci.1135-17.2017] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/21/2017] [Accepted: 10/18/2017] [Indexed: 01/24/2023] Open
Abstract
Sleep-wake abnormalities are common in patients with Alzheimer's disease, and can be a major reason for institutionalization. However, an emerging concept is that these sleep-wake disturbances are part of the causal pathway accelerating the neurodegenerative process. Recently, new findings have provided intriguing evidence for a positive feedback loop between sleep-wake dysfunction and β-amyloid (Aβ) aggregation. Studies in both humans and animal models have shown that extended periods of wakefulness increase Aβ levels and aggregation, and accumulation of Aβ causes fragmentation of sleep. This perspective is aimed at presenting evidence supporting causal links between sleep-wake dysfunction and aggregation of Aβ peptide in Alzheimer's disease, and explores the role of astrocytes, a specialized type of glial cell, in this context underlying Alzheimer's disease pathology. The utility of current animal models and the unexplored potential of alternative animal models for testing mechanisms involved in the reciprocal relationship between sleep disruption and Aβ are also discussed.Dual Perspectives Companion Paper: Microglia-Mediated Synapse Loss in Alzheimer's Disease by Lawrence Rajendran and Rosa Paolicelli.
Collapse
|
39
|
|
40
|
Komagata N, Latifi B, Rusterholz T, Bassetti CLA, Adamantidis A, Schmidt MH. Dynamic REM Sleep Modulation by Ambient Temperature and the Critical Role of the Melanin-Concentrating Hormone System. Curr Biol 2019; 29:1976-1987.e4. [PMID: 31155350 DOI: 10.1016/j.cub.2019.05.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/02/2019] [Accepted: 05/01/2019] [Indexed: 02/06/2023]
Abstract
Ambient temperature (Ta) warming toward the high end of the thermoneutral zone (TNZ) preferentially increases rapid eye movement (REM) sleep over non-REM (NREM) sleep across species. The control and function of this temperature-induced REM sleep expression have remained unknown. Melanin-concentrating hormone (MCH) neurons play an important role in REM sleep control. We hypothesize that the MCH system may modulate REM sleep as a function of Ta. Here, we show that wild-type (WT) mice dynamically increased REM sleep durations specifically during warm Ta pulsing within the TNZ, compared to both the TNZ cool and baseline constant Ta conditions, without significantly affecting either wake or NREM sleep durations. However, genetically engineered MCH receptor-1 knockout (MCHR1-KO) mice showed no significant changes in REM sleep as a function of Ta, even with increased sleep pressure following a 4-h sleep deprivation. Using MCH-cre mice transduced with channelrhodopsin, we then optogenetically activated MCH neurons time locked with Ta warming, showing an increase in REM sleep expression beyond what Ta warming in yellow fluorescent protein (YFP) control mice achieved. Finally, in mice transduced with archaerhodopsin-T, semi-chronic optogenetic MCH neuronal silencing during Ta warming completely blocked the increase in REM sleep seen in YFP controls. These data demonstrate a previously unknown role for the MCH system in the dynamic output expression of REM sleep during Ta manipulation. These findings are consistent with the energy allocation hypothesis of sleep function, suggesting that endotherms have evolved neural circuits to opportunistically express REM sleep when the need for thermoregulatory defense is minimized.
Collapse
Affiliation(s)
- Noëmie Komagata
- Bern University Hospital (Inselspital), University of Bern, 3010 Bern, Switzerland
| | - Blerina Latifi
- Bern University Hospital (Inselspital), University of Bern, 3010 Bern, Switzerland
| | - Thomas Rusterholz
- Center for Experimental Neurology, Department of Neurology, Bern University Hospital (Inselspital), University of Bern, 3010 Bern, Switzerland
| | - Claudio L A Bassetti
- Department of Neurology, Bern University Hospital (Inselspital), University of Bern, Freiburgstrasse 18, 3010 Bern, Switzerland
| | - Antoine Adamantidis
- Center for Experimental Neurology, Department of Neurology, Bern University Hospital (Inselspital), University of Bern, 3010 Bern, Switzerland; Department of Biomedical Research (DBMR), Bern University Hospital (Inselspital), University of Bern, 3010 Bern, Switzerland
| | - Markus H Schmidt
- Center for Experimental Neurology, Department of Neurology, Bern University Hospital (Inselspital), University of Bern, 3010 Bern, Switzerland; Department of Neurology, Bern University Hospital (Inselspital), University of Bern, Freiburgstrasse 18, 3010 Bern, Switzerland; Ohio Sleep Medicine Institute, 4975 Bradenton Avenue, Dublin, OH 43017, USA.
| |
Collapse
|
41
|
Zada D, Bronshtein I, Lerer-Goldshtein T, Garini Y, Appelbaum L. Sleep increases chromosome dynamics to enable reduction of accumulating DNA damage in single neurons. Nat Commun 2019; 10:895. [PMID: 30837464 PMCID: PMC6401120 DOI: 10.1038/s41467-019-08806-w] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 01/30/2019] [Indexed: 11/09/2022] Open
Abstract
Sleep is essential to all animals with a nervous system. Nevertheless, the core cellular function of sleep is unknown, and there is no conserved molecular marker to define sleep across phylogeny. Time-lapse imaging of chromosomal markers in single cells of live zebrafish revealed that sleep increases chromosome dynamics in individual neurons but not in two other cell types. Manipulation of sleep, chromosome dynamics, neuronal activity, and DNA double-strand breaks (DSBs) showed that chromosome dynamics are low and the number of DSBs accumulates during wakefulness. In turn, sleep increases chromosome dynamics, which are necessary to reduce the amount of DSBs. These results establish chromosome dynamics as a potential marker to define single sleeping cells, and propose that the restorative function of sleep is nuclear maintenance.
Collapse
Affiliation(s)
- D Zada
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - I Bronshtein
- Department of Physics and the Institute for Nanotechnology, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - T Lerer-Goldshtein
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Y Garini
- Department of Physics and the Institute for Nanotechnology, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - L Appelbaum
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, 5290002, Israel.
| |
Collapse
|
42
|
Mansilla A, Jordán-Álvarez S, Santana E, Jarabo P, Casas-Tintó S, Ferrús A. Molecular mechanisms that change synapse number. J Neurogenet 2018; 32:155-170. [DOI: 10.1080/01677063.2018.1506781] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
43
|
Azeez IA, Del Gallo F, Cristino L, Bentivoglio M. Daily Fluctuation of Orexin Neuron Activity and Wiring: The Challenge of "Chronoconnectivity". Front Pharmacol 2018; 9:1061. [PMID: 30319410 PMCID: PMC6167434 DOI: 10.3389/fphar.2018.01061] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022] Open
Abstract
In the heterogeneous hub represented by the lateral hypothalamus, neurons containing the orexin/hypocretin peptides play a key role in vigilance state transitions and wakefulness stability, energy homeostasis, and other functions relevant for motivated behaviors. Orexin neurons, which project widely to the neuraxis, are innervated by multiple extra- and intra-hypothalamic sources. A key property of the adaptive capacity of orexin neurons is represented by daily variations of activity, which is highest in the period of the animal’s activity and wakefulness. These sets of data are here reviewed. They concern the discharge profile during the sleep/wake cycle, spontaneous Fos induction, peptide synthesis and release reflected by immunostaining intensity and peptide levels in the cerebrospinal fluid as well as postsynaptic effects. At the synaptic level, adaptive capacity of orexin neurons subserved by remodeling of excitatory and inhibitory inputs has been shown in response to changes in the nutritional status and prolonged wakefulness. The present review wishes to highlight that synaptic plasticity in the wiring of orexin neurons also occurs in unperturbed conditions and could account for diurnal variations of orexin neuron activity. Data in zebrafish larvae have shown rhythmic changes in the density of inhibitory innervation of orexin dendrites in relation to vigilance states. Recent findings in mice have indicated a diurnal reorganization of the excitatory/inhibitory balance in the perisomatic innervation of orexin neurons. Taken together these sets of data point to “chronoconnectivity,” i.e., a synaptic rearrangement of inputs to orexin neurons over the course of the day in relation to sleep and wake states. This opens questions on the underlying circadian and homeostatic regulation and on the involved players at synaptic level, which could implicate dual transmitters, cytoskeletal rearrangements, hormonal regulation, as well as surrounding glial cells and extracellular matrix. Furthermore, the question arises of a “chronoconnectivity” in the wiring of other neuronal cell groups of the sleep-wake-regulatory network, many of which are characterized by variations of their firing rate during vigilance states.
Collapse
Affiliation(s)
- Idris A Azeez
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Federico Del Gallo
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | | | - Marina Bentivoglio
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.,National Institute of Neuroscience, Verona Unit, Verona, Italy
| |
Collapse
|
44
|
Swanson A, Wolf T, Sitzmann A, Willette AA. Neuroinflammation in Alzheimer's disease: Pleiotropic roles for cytokines and neuronal pentraxins. Behav Brain Res 2018; 347:49-56. [PMID: 29462653 PMCID: PMC5988985 DOI: 10.1016/j.bbr.2018.02.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/30/2018] [Accepted: 02/13/2018] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is a potential factor speculated to underlie Alzheimer's disease (AD) etiopathogenesis and progression. The overwhelming focus in this area of research to date has been on the chronic upregulation of pro-inflammatory cytokines to understand how neuroinflammatory mechanisms contribute to neurodegeneration. Yet, it is important to understand the pleiotropic roles of these cytokines in modulating neuroinflammation in which they cannot be labeled as a strictly "good" or "bad" biomarker phenotype. As such, biomarkers with more precise functions are needed to better understand how neuroinflammation impacts the brain in AD. Neuronal pentraxins are a concentration- dependent group of pro- or anti- inflammatory cytokines. There is contradictory evidence of these pentraxins as being both neuroprotective and potentially detrimental in AD. Potential neuroprotective examples include their ability to predict AD-related outcomes such as cognition, memory function and synaptic refinement. This review will briefly outline the basis of AD and subsequently summarize findings for neuropathological mechanisms of neuroinflammation, roles for traditional pro-and anti-inflammatory cytokines, and data found thus far on the neuronal pentraxins.
Collapse
Affiliation(s)
- Ashley Swanson
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States.
| | - Tovah Wolf
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States.
| | - Alli Sitzmann
- Department of Psychology, Iowa State University, W112 Lagomarcino Hall, 901 Stange Road, Ames, IA 50011, United States.
| | - Auriel A Willette
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States; Department of Psychology, Iowa State University, W112 Lagomarcino Hall, 901 Stange Road, Ames, IA 50011, United States; Department of Biomedical Sciences, Iowa State University, 2008 Veterinary Medicine, Ames, IA 50011, United States; Department of Neurology, University of Iowa, 2007 Roy Carver Pavilion, 200 Hawkins Drive, Iowa City, IA 52242, United States.
| |
Collapse
|
45
|
Tyree SM, Borniger JC, de Lecea L. Hypocretin as a Hub for Arousal and Motivation. Front Neurol 2018; 9:413. [PMID: 29928253 PMCID: PMC5997825 DOI: 10.3389/fneur.2018.00413] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/18/2018] [Indexed: 01/01/2023] Open
Abstract
The lateral hypothalamus is comprised of a heterogeneous mix of neurons that serve to integrate and regulate sleep, feeding, stress, energy balance, reward, and motivated behavior. Within these populations, the hypocretin/orexin neurons are among the most well studied. Here, we provide an overview on how these neurons act as a central hub integrating sensory and physiological information to tune arousal and motivated behavior accordingly. We give special attention to their role in sleep-wake states and conditions of hyper-arousal, as is the case with stress-induced anxiety. We further discuss their roles in feeding, drug-seeking, and sexual behavior, which are all dependent on the motivational state of the animal. We further emphasize the application of powerful techniques, such as optogenetics, chemogenetics, and fiber photometry, to delineate the role these neurons play in lateral hypothalamic functions.
Collapse
Affiliation(s)
- Susan M Tyree
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Jeremy C Borniger
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| |
Collapse
|
46
|
Krzeptowski W, Hess G, Pyza E. Circadian Plasticity in the Brain of Insects and Rodents. Front Neural Circuits 2018; 12:32. [PMID: 29770112 PMCID: PMC5942159 DOI: 10.3389/fncir.2018.00032] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 04/09/2018] [Indexed: 12/22/2022] Open
Abstract
In both vertebrate and invertebrate brains, neurons, glial cells and synapses are plastic, which means that the physiology and structure of these components are modified in response to internal and external stimuli during development and in mature brains. The term plasticity has been introduced in the last century to describe experience-dependent changes in synapse strength and number. These changes result from local functional and morphological synapse modifications; however, these modifications also occur more commonly in pre- and postsynaptic neurons. As a result, neuron morphology and neuronal networks are constantly modified during the life of animals and humans in response to different stimuli. Nevertheless, it has been discovered in flies and mammals that the number of synapses and size and shape of neurons also oscillate during the day. In most cases, these rhythms are circadian since they are generated by endogenous circadian clocks; however, some rhythmic changes in neuron morphology and synapse number and structure are controlled directly by environmental cues or by both external cues and circadian clocks. When the circadian clock is involved in generating cyclic changes in the nervous system, this type of plasticity is called circadian plasticity. It seems to be important in processing sensory information, in learning and in memory. Disruption of the clock may affect major brain functions.
Collapse
Affiliation(s)
- Wojciech Krzeptowski
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Grzegorz Hess
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland.,Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Elżbieta Pyza
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| |
Collapse
|
47
|
Holland PR. Biology of Neuropeptides: Orexinergic Involvement in Primary Headache Disorders. Headache 2018; 57 Suppl 2:76-88. [PMID: 28485849 DOI: 10.1111/head.13078] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 03/08/2017] [Indexed: 01/01/2023]
Abstract
Migraine is a very common, severe disabling condition that can last for days and strike multiple times per month. Attacks, often characterized by severe unilateral throbbing pain that is exacerbated by activity, are commonly preceded by several diverse symptoms including fatigue, irritability, and yawning. This premonitory (prodromal) phase represents the earliest identifiable feature of an attack that is a reliable predictor of ensuing headache. The diversity of these symptoms underlines the complex nature of migraine and focuses considerable attention on the hypothalamus due to its prominent role in homeostatic regulation allowing state dependent behavioral modifications. While multiple neurotransmitter and neuropeptide systems have been proposed to play a role in migraine, the current review will focus on the emerging role of the hypothalamic orexinergic system in primary headache disorders. Specifically the potential role of altered orexinergic signalling in premonitory symptomatology and the future potential of targeted orexinergic therapies that could with other approaches act during the premonitory phase to prevent the occurrence of the headache or reduce an individual's susceptibility to attacks by altering the brain's response to external and internal triggers.
Collapse
Affiliation(s)
- Philip R Holland
- Headache Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
48
|
Akladious A, Azzam S, Hu Y, Feng P. Bmal1 knockdown suppresses wake and increases immobility without altering orexin A, corticotrophin-releasing hormone, or glutamate decarboxylase. CNS Neurosci Ther 2018; 24:549-563. [PMID: 29446232 DOI: 10.1111/cns.12815] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 01/02/2018] [Accepted: 01/10/2018] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE To determine the effect of Bmal1 knockdown (KD) on sleep, activity, immobility, hypothalamic levels of orexin, corticotrophin-releasing hormone (CRH), and GABAergic glutamate decarboxylase (GAD). METHODS We used Bmal1 siRNA, or control siRNA intracerebroventricular (ICV) injection to knock down Bmal1 in C57BL/6 mice. Sleep polysomnography, wheel-running activity, and tail suspension test were performed. Polysomnographic (PSG) recordings in both groups were preceded by ICV injection made during both the light phase and the dark phase. We also measured brain orexin A and CRH using an ELISA and measured GAD using immunoblotting. RESULTS Compared with control group, Bmal1 KD group had reduced wheel activity and increased immobility. Compared with control, the Bmal1 KD group had reduced wheel activity and increased immobility. During the first 24 hours after treatment, we observed that control siRNA induced a much greater increase in sleep during the dark phase, which was associated with lower orexin levels. However, beginning 24 hours after treatment, we observed an increase in sleep and a decrease in time spent awake during the dark phase in the Bmal1 KD group. These changes were not associated with changes in brain levels of orexin A, CRH, or GAD. CONCLUSION Bmal1 KD led to reduced activity, increased immobility, and dramatic reduction in time spent awake as well as an increase in sleep during the dark phase. Early after injection, there was a slight change in sleep but brain levels of orexin, CRH, and GAD remain unchanged. Control siRNA also affected sleep associated with changes in orexin levels.
Collapse
Affiliation(s)
- Afaf Akladious
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Sausan Azzam
- Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Yufen Hu
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Pingfu Feng
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
49
|
Circadian Regulation of Hippocampal-Dependent Memory: Circuits, Synapses, and Molecular Mechanisms. Neural Plast 2018; 2018:7292540. [PMID: 29593785 PMCID: PMC5822921 DOI: 10.1155/2018/7292540] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/18/2017] [Indexed: 01/01/2023] Open
Abstract
Circadian modulation of learning and memory efficiency is an evolutionarily conserved phenomenon, occurring in organisms ranging from invertebrates to higher mammalian species, including humans. While the suprachiasmatic nucleus (SCN) of the hypothalamus functions as the master mammalian pacemaker, recent evidence suggests that forebrain regions, including the hippocampus, exhibit oscillatory capacity. This finding, as well as work on the cellular signaling events that underlie learning and memory, has opened promising new avenues of investigation into the precise cellular, molecular, and circuit-based mechanisms by which clock timing impacts plasticity and cognition. In this review, we examine the complex molecular relationship between clock timing and memory, with a focus on hippocampal-dependent tasks. We evaluate how the dysregulation of circadian timing, both at the level of the SCN and at the level of ancillary forebrain clocks, affects learning and memory. Further, we discuss experimentally validated intracellular signaling pathways (e.g., ERK/MAPK and GSK3β) and potential cellular signaling mechanisms by which the clock affects learning and memory formation. Finally, we examine how long-term potentiation (LTP), a synaptic process critical to the establishment of several forms of memory, is regulated by clock-gated processes.
Collapse
|
50
|
Shams S, Rihel J, Ortiz JG, Gerlai R. The zebrafish as a promising tool for modeling human brain disorders: A review based upon an IBNS Symposium. Neurosci Biobehav Rev 2018; 85:176-190. [DOI: 10.1016/j.neubiorev.2017.09.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 08/28/2017] [Accepted: 09/02/2017] [Indexed: 12/12/2022]
|