1
|
Tort ABL, Laplagne DA, Draguhn A, Gonzalez J. Global coordination of brain activity by the breathing cycle. Nat Rev Neurosci 2025; 26:333-353. [PMID: 40204908 DOI: 10.1038/s41583-025-00920-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
Neuronal activities that synchronize with the breathing rhythm have been found in humans and a host of mammalian species, not only in brain areas closely related to respiratory control or olfactory coding but also in areas linked to emotional and higher cognitive functions. In parallel, evidence is mounting for modulations of perception and action by the breathing cycle. In this Review, we discuss the extent to which brain activity locks to breathing across areas, levels of organization and brain states, and the physiological origins of this global synchrony. We describe how waves of sensory activity evoked by nasal airflow spread through brain circuits, synchronizing neuronal populations to the breathing cycle and modulating faster oscillations, cell assembly formation and cross-area communication, thereby providing a mechanistic link from breathing to neural coding, emotion and cognition. We argue that, through evolution, the breathing rhythm has come to shape network functions across species.
Collapse
Affiliation(s)
- Adriano B L Tort
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil.
| | - Diego A Laplagne
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil.
| | - Andreas Draguhn
- Institute for Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Joaquin Gonzalez
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Neuroscience Institute and Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
2
|
Imamura T, Wasilczuk AZ, Reitz SL, Lian J, Imamura M, Keenan BT, Shimizu N, Pack AI, Kelz MB. Parafacial GABAergic neurone ablation induces behavioural resistance to volatile anaesthetic-induced hypnosis without reducing sleep. Br J Anaesth 2025; 134:1696-1708. [PMID: 40240218 DOI: 10.1016/j.bja.2025.02.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND It is hypothesised that general anaesthetics co-opt the neural circuits regulating endogenous sleep and wakefulness to produce hypnosis. To further probe this association, we focused on the GABAergic neurones of the parafacial zone (PZGABA), a brainstem site capable of promoting non-rapid eye movement sleep. METHODS To determine whether PZ neurones are activated by a hypnotic dose of anaesthetics, c-Fos immunohistochemistry was performed. The behavioural and physiological contributions of PZGABA neurones to anaesthetic sensitivity were assessed in mice transfected with an adeno-associated virus (AAV)-driving expression of an mCherry fluorescent control or a caspase that irreversibly eliminates PZGABA neurones. EEG-defined sleep was measured in PZGABA-ablated and mCherry control mice, as was the homeostatic drive to sleep after sleep deprivation. RESULTS Consistent with anaesthetic-induced depolarisation, hypnotic doses of isoflurane significantly increased c-Fos expression three-fold in PZGABA neurones compared with oxygen-exposed mice. PZGABA-ablated mice developed significant and durable behavioural resistance to both isoflurane- and sevoflurane-induced hypnosis, with roughly 50% higher likelihood of intact righting than controls. PZGABA-ablated mice emerged from isoflurane significantly faster than mCherry controls with purposeful movements. The degree of anaesthetic resistance was inversely correlated with the number of surviving PZGABA neurones. Despite confirming that PZGABA ablation reduced the potency of two distinct volatile anaesthetics behaviourally, ablation did not alter the amount of endogenous sleep or wakefulness, nor did it affect the homeostatic sleep drive after sleep deprivation, and it did not produce EEG signatures of anaesthetic resistance during isoflurane exposure. CONCLUSIONS There was an unexpected dissociation in which destruction of up to 70-80% of PZGABA neurones was sufficient to alter anaesthetic susceptibility behaviourally without causing insomnia or altering sleep pressure. These findings suggest that PZGABA neurones are more critical to drug-induced hypnosis than to the regulation of natural sleep and arousal.
Collapse
Affiliation(s)
- Toshihiro Imamura
- Chronobiology and Sleep Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Division of Pulmonary and Sleep Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Sleep Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Andrzej Z Wasilczuk
- Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah L Reitz
- Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jie Lian
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Miyoko Imamura
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Brendan T Keenan
- Chronobiology and Sleep Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Division of Sleep Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Naoki Shimizu
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Allan I Pack
- Chronobiology and Sleep Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Division of Sleep Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Max B Kelz
- Chronobiology and Sleep Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Takeda K, Watanabe K, Iijima S, Nagahiro T, Suzuki H, Izumo K, Ikegaya Y, Matsumoto N. Ramelteon coordinates theta and gamma oscillations in the hippocampus for novel object recognition memory in mice. J Pharmacol Sci 2025; 158:121-130. [PMID: 40288822 DOI: 10.1016/j.jphs.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/14/2025] [Accepted: 03/29/2025] [Indexed: 04/29/2025] Open
Abstract
Object recognition memory is an animal's ability to discriminate between novel and familiar items and is supported by neural activities in not only the perirhinal cortex but also the hippocampus and prefrontal cortex. Since we previously demonstrated that ramelteon enhanced object recognition memory in mice, we sought neural correlates of the memory improvement. We recorded neural activity in the hippocampus and prefrontal cortex of mice while they performed a novel object recognition task. We found that theta oscillations in the hippocampus were enhanced when ramelteon-treated mice explored both novel and familiar objects. Moreover, we showed high coherence in phases at low gamma frequencies between the hippocampus and prefrontal cortex. We assume that theta enhancement is indicative of increased cholinergic activity by melatonin receptor activation. High coherence of low gamma oscillations between the hippocampal and prefrontal network in ramelteon-treated mice sampling novel objects suggests better cognitive operations for discrimination between novelty and familiarity. The current study sheds light upon physiological consequences of melatonin receptor activation, further contributing improved cognitive functions.
Collapse
Affiliation(s)
- Kinjiro Takeda
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Kisa Watanabe
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Sena Iijima
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Takeshi Nagahiro
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Haruka Suzuki
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Kano Izumo
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo, 113-0033, Japan; Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City, Osaka, 565-0871, Japan
| | - Nobuyoshi Matsumoto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
4
|
Chen T, Jia J, Gao C, Zhong Q, Tang L, Sui X, Li S, Chen C, Zhang Z. Integrative metabolomics and transcriptomics analysis of hippocampus reveals taurine metabolism and sphingolipid metabolism dysregulation associated with sleep deprivation-induced memory impairment. Brain Res Bull 2025:111397. [PMID: 40409601 DOI: 10.1016/j.brainresbull.2025.111397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 05/06/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
Sleep plays a crucial role in restoring and repairing the body, consolidating memory, regulating emotions, maintaining metabolic and so on. Sleep deprivation is known to impair cognitive functions. In this study, we investigated the mechanisms underlying memory impairment induced by sleep deprivation through a combined metabolomic and transcriptomic analysis of hippocampus. Eight-week-old mice were selected as the study subjects and the sleep deprivation chamber was used to establish a sleep deprivation (SD) model. Novel object recognition tests (NOR), and Y-maze tests were used to assess the behavioral outcomes in mice. The hippocampus were extracted and studied using the untargeted metabolomics or transcriptomics high-throughput sequencing method. An integrative analysis was conducted to elucidate the metabolic and genetic changes. Behavioral tests showed that SD group exhibited memory impairment. Metabolomic analysis identified 84 differentially expressed metabolites (DEMs), including 12 under the positive ion mode and 72 under the negative ion mode. The analysis revealed that sleep deprivation caused abnormalities in several metabolic pathways, with particularly pronounced effects observed in glycerophospholipid metabolism, linoieic acid metabolism, alanine, aspartate, glutamate metabolism, taurine and hypotaurine metabolism, and purine metabolism. While transcriptomic analysis releaved 97 differentially expressed genes (DEGs) (51 were down-regulated and 46 were up-regulated DEGs). Integrative analysis of the metabolomic and transcriptomic identified profiles showed that sleep deprivation may regulate taurine and hypotaurine metabolism and sphingolipid metabolism, there by influencing memory. Our results prompt severe metabolic disturbances occur in the hippocampus with SD in mice, which can provide a basis for the mechanism research.
Collapse
Affiliation(s)
- Ting Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China.
| | - Junke Jia
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Chenyi Gao
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China; Department of Anesthesiology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315040, China
| | - Qi Zhong
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Lijuan Tang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Xiaokai Sui
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Shuang Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China; Sleep medicine center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Chang Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China.
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China.
| |
Collapse
|
5
|
Ono A, Iwatani Y, Yoshizaki A, Nishimura T, Mohri I, Kagitani-Shimono K, Taniike M. Exploring Lifestyles and Sensory Processing Patterns of Toddlers in Relation to Sleep Patterns Using Body Movement Analysis. Clocks Sleep 2025; 7:25. [PMID: 40407631 PMCID: PMC12101242 DOI: 10.3390/clockssleep7020025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/27/2025] [Accepted: 05/07/2025] [Indexed: 05/26/2025] Open
Abstract
This study explored the impact of lifestyle habits and sensory processing patterns on sleep quality by analyzing body movements (BMs) during the first and last 3 h of sleep in toddlers. We collected cross-sectional data about sleep-related habits from 58 toddlers using a mobile application. Actigraphy measured BMs during nighttime sleep and 1 h before bedtime, as well as sleep latency, over 8 consecutive days. The Infant/Toddler Sensory Profile was used to assess the toddlers' sensory processing patterns. The participants had a mean age of 22.0 ± 2.0 months. BMs were significantly lower during the first 3 h of sleep. Longer sleep latency was significantly associated with media use and higher activity levels before bedtime. Ending a nap earlier and consuming a substantial breakfast were correlated with lower BMs during the first 3 h of sleep. Auditory and oral sensory scores were positively correlated with BMs during the first 3 h of sleep. However, no specific factors related to lifestyle habits or sensory processing patterns were found to correlate with BMs during the last 3 h of sleep. Lifestyle habits and sensory processing patterns have a significant impact on toddlers' sleep quality, emphasizing the importance of appropriate routines and environments.
Collapse
Affiliation(s)
- Azusa Ono
- Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan; (A.O.); (Y.I.); (T.N.); (I.M.); (K.K.-S.)
| | - Yoshiko Iwatani
- Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan; (A.O.); (Y.I.); (T.N.); (I.M.); (K.K.-S.)
| | - Arika Yoshizaki
- Molecular Research Center for Children’s Mental Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan;
| | - Tomoko Nishimura
- Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan; (A.O.); (Y.I.); (T.N.); (I.M.); (K.K.-S.)
| | - Ikuko Mohri
- Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan; (A.O.); (Y.I.); (T.N.); (I.M.); (K.K.-S.)
| | - Kuriko Kagitani-Shimono
- Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan; (A.O.); (Y.I.); (T.N.); (I.M.); (K.K.-S.)
| | - Masako Taniike
- Molecular Research Center for Children’s Mental Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan;
| |
Collapse
|
6
|
Khandayataray P, Murthy MK. Exploring the nexus: Sleep disorders, circadian dysregulation, and Alzheimer's disease. Neuroscience 2025; 574:21-41. [PMID: 40189132 DOI: 10.1016/j.neuroscience.2025.03.066] [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: 01/02/2025] [Revised: 03/10/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
We reviewed the connections among Alzheimer's disease (AD), sleep deprivation, and circadian rhythm disorders. Evidence is mounting that disrupted sleep and abnormal circadian rhythms are not merely symptoms of AD, but are also involved in accelerating the disease. Amyloid-beta (Aβ) accumulates, a feature of AD, and worsens with sleep deprivation because glymphatic withdrawal is required to clear toxic proteins from the brain. In addition, disturbances in circadian rhythm can contribute to the induction of neuroinflammation and oxidative stress, thereby accelerating neurodegenerative processes. While these interactions are bidirectional, Alzheimer's pathology further disrupts sleep and circadian function in a vicious cycle that worsens cognitive decline, which is emphasized in the review. The evidence that targeting sleep and circadian mechanisms may serve as therapeutic strategies for AD was strengthened by this study through the analysis of the molecular and physiological pathways. Further work on this nexus could help unravel the neurobiological mechanisms common to the onset of Alzheimer's and disrupted sleep and circadian regulation, which could result in earlier intervention to slow or prevent the onset of the disease.
Collapse
Affiliation(s)
- Pratima Khandayataray
- Department of Biotechnology, Academy of Management and Information Technology, Utkal University, Bhubaneswar, Odisha 752057, India
| | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Punjab 140401, India.
| |
Collapse
|
7
|
Sharon O, Zhelezniakov V, Gat Y, Falach R, Narbayev D, Shiner T, Walker MP, Tauman R, Bregman N, Nir Y. Slow wave synchrony during NREM sleep tracks cognitive impairment in prodromal Alzheimer's disease. Alzheimers Dement 2025; 21:e70247. [PMID: 40399753 PMCID: PMC12094885 DOI: 10.1002/alz.70247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 05/23/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD) disrupts human sleep architecture more severely than normal aging. However, it remains unclear how AD changes oscillatory neural activity during sleep, and whether such changes foreshadow cognitive decline in AD. METHODS We used high-density electroencephalography sleep recordings in 55 participants: (1) 21 healthy older adults, (2) 28 patients with amnestic mild cognitive impairment (aMCI)-a prodromal AD stage, and (3) 6 AD patients. RESULTS Cognitive performance robustly decreases with the slow wave (SW) trough amplitude and its synchronization across broad frontocentral cortical areas. Thus, across the AD spectrum, slow wave synchrony declines with cognition, as in normal aging, but at an accelerated pace. Moreover, delayed rapid eye movement (REM) sleep onset in aMCI and AD patients was associated with deficient SW activity, suggesting insufficiently restorative non-REM sleep. DISCUSSION These findings suggest that impaired slow waves are closely linked to cognitive impairment and mark disrupted neural activity in AD progression. HIGHLIGHTS Detailed analysis of high-density sleep electroencephalography was performed in amnestic mild cognitive impairment and Alzheimer's disease (AD) patients. Cognitive status robustly correlates with slow wave trough and its cortical spread. Delayed rapid eye movement sleep onset associated with AD correlates with diminished slow wave troughs. Impaired slow waves mark progressively disrupted neural activity in prodromal AD.
Collapse
Affiliation(s)
- Omer Sharon
- Center for Human Sleep Science, Department of PsychologyUniversity of California, BerkeleyBerkeleyCaliforniaUSA
- Helen Wills Neuroscience InstituteUniversity of California, BerkeleyBerkeleyCaliforniaUSA
- Department of Physiology & Pharmacology, Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Vladislav Zhelezniakov
- Department of Physiology & Pharmacology, Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Yael Gat
- Department of Physiology & Pharmacology, Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| | - Rotem Falach
- Department of Physiology & Pharmacology, Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| | - Darya Narbayev
- Department of Physiology & Pharmacology, Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Tamara Shiner
- Cognitive Neurology UnitTel Aviv Sourasky Medical CenterTel AvivIsrael
- Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Matthew P. Walker
- Center for Human Sleep Science, Department of PsychologyUniversity of California, BerkeleyBerkeleyCaliforniaUSA
- Helen Wills Neuroscience InstituteUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Riva Tauman
- Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
- The Sieratzki‐Sagol Center for Sleep MedicineTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Noa Bregman
- Cognitive Neurology UnitTel Aviv Sourasky Medical CenterTel AvivIsrael
- Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Yuval Nir
- Department of Physiology & Pharmacology, Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- The Sieratzki‐Sagol Center for Sleep MedicineTel Aviv Sourasky Medical CenterTel AvivIsrael
- Department of Biomedical Engineering, Faculty of EngineeringTel Aviv UniversityTel AvivIsrael
- Sagol Brain InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| |
Collapse
|
8
|
Xie L, Wu Q, Huang H, Wang K, Ying K, Liu Z, Li S. Neuroregulation of histamine of circadian rhythm disorder induced by chronic intermittent hypoxia. Eur J Pharmacol 2025; 999:177662. [PMID: 40311833 DOI: 10.1016/j.ejphar.2025.177662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 04/20/2025] [Accepted: 04/22/2025] [Indexed: 05/03/2025]
Abstract
Obstructive sleep apnea (OSA) is characterized by intermittent hypoxemia, sleep fragmentation, and excessive daytime sleepiness. OSA patients are at an elevated risk for circadian rhythm disturbances. Histamine is known to regulate the sleep-wake cycle predominantly via histamine H1 receptors. We utilized a C57BL/6 mouse model exposed to chronic intermittent hypoxia (CIH) for three weeks to assess alterations in circadian rhythmicity. Sleep architecture and voluntary wheel-running activity were evaluated. Additionally, c-fos expression and mPer2 levels in the frontal cortex (FC) and the suprachiasmatic nucleus (SCN) were examined. BV-2 microglial cells were subjected to intermittent hypoxia (IH) for 12 h to explore the underlying signaling pathways. CIH exposure led to a significant prolongation of the wake phase and a reduction in the Non-rapid eye movement (NREM) phase, accompanied by increased sleep fragmentation and disruption of circadian rhythms. Treatment with mepyramine, an H1 receptor antagonist, mitigated these effects by reducing arousal duration, extending NREM phase, and decreasing sleep fragmentation. CIH also resulted in increased c-fos expression and elevated mPer2 levels in the FC and SCN, both of which were reversed following mepyramine administration. In vitro studies on BV-2 cells demonstrated that histamine exerts its modulatory effects through the activation of the PLC and PKA signaling pathways, influencing mPer2 expression via the regulation of K+, Na + -Ca2+, and Ca2+ ion channels. In conclusion, CIH disrupts circadian rhythms through histamine-mediated mechanisms, and mepyramine effectively ameliorates these disruptions. These findings highlight histamine as a promising therapeutic target for addressing circadian rhythm disorders associated with OSA.
Collapse
Affiliation(s)
- Liang Xie
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, China; Clinical Centre for Sleep Breathing Disorders and Snoring, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qinhan Wu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, China; Clinical Centre for Sleep Breathing Disorders and Snoring, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huai Huang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, China; Clinical Centre for Sleep Breathing Disorders and Snoring, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kexin Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, China; Clinical Centre for Sleep Breathing Disorders and Snoring, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kelu Ying
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, China; Clinical Centre for Sleep Breathing Disorders and Snoring, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zilong Liu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, China; Clinical Centre for Sleep Breathing Disorders and Snoring, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Shanqun Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, China; Clinical Centre for Sleep Breathing Disorders and Snoring, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Zhang P, An X, Yang R, Qi M, Gao Z, Zhang X, Wu Z, Zheng Z, Dong X, Wang W, Wang X, Zha D. Echoes in the night: How sleep quality influences auditory health. Neuroscience 2025; 577:200-216. [PMID: 40294844 DOI: 10.1016/j.neuroscience.2025.04.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/27/2025] [Accepted: 04/23/2025] [Indexed: 04/30/2025]
Abstract
The intricate relationship between sleep disorders and hearing loss emerges as a burgeoning field of scholarly inquiry. Numerous studies have illuminated a potential correlation between the two, affecting the quality of life and overall health of individuals. Hearing loss, or auditory impairment, serves as a critical indicator of physiological dysfunction, casting a pall over the daily existence and professional endeavors of those affected, potentially leading to irreversible deafness if left untreated. Sleep disorders may cause physical and psychological changes that further affect hearing, while auditory dysfunction may detrimentally impact sleep experienced by individuals. Although certain studies have failed to find a direct link between sleep duration and hearing loss, it is evident that sleep-related issues do increase the risk of hearing loss. Thus, understanding the relationship between sleep disorders and hearing loss, alongside the underlying mechanisms, will help establish interventions aimed at enhancing sleep quality and safeguarding auditory health. This systematic review endeavors to elucidate the correlation between sleep disorders and hearing loss, offering valuable insights and guidance for future basic research and clinical applications.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Otolaryngology, Xijing Hospital, Air Force Medical University, Xi'an 710032 Shaanxi Province, China
| | - Xiaogang An
- Department of Otolaryngology, Xijing Hospital, Air Force Medical University, Xi'an 710032 Shaanxi Province, China
| | - Runqin Yang
- Department of Otolaryngology, Xijing Hospital, Air Force Medical University, Xi'an 710032 Shaanxi Province, China
| | - Meihao Qi
- Department of Otolaryngology, Xijing Hospital, Air Force Medical University, Xi'an 710032 Shaanxi Province, China
| | - Zejun Gao
- Department of Otolaryngology, Xijing Hospital, Air Force Medical University, Xi'an 710032 Shaanxi Province, China
| | - Xinyu Zhang
- Department of Otolaryngology, Xijing Hospital, Air Force Medical University, Xi'an 710032 Shaanxi Province, China
| | - Ziqi Wu
- Department of Otolaryngology, Xijing Hospital, Air Force Medical University, Xi'an 710032 Shaanxi Province, China
| | - Zeyu Zheng
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of Education, Air Force Medical University, Xi'an 710032 Shaanxi Province, China
| | - Xinyu Dong
- Department of Otolaryngology, Xijing Hospital, Air Force Medical University, Xi'an 710032 Shaanxi Province, China
| | - Wenyue Wang
- Department of Otolaryngology, Xijing Hospital, Air Force Medical University, Xi'an 710032 Shaanxi Province, China
| | - Xiaocheng Wang
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of Education, Air Force Medical University, Xi'an 710032 Shaanxi Province, China.
| | - Dingjun Zha
- Department of Otolaryngology, Xijing Hospital, Air Force Medical University, Xi'an 710032 Shaanxi Province, China.
| |
Collapse
|
10
|
Topchiy I, Kocsis B. CB-1 receptor agonist drastically changes oscillatory activity, defining active sleep. Proc Natl Acad Sci U S A 2025; 122:e2411063122. [PMID: 40249784 PMCID: PMC12037043 DOI: 10.1073/pnas.2411063122] [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: 06/03/2024] [Accepted: 03/01/2025] [Indexed: 04/20/2025] Open
Abstract
Brain oscillations in different behavioral states are essential for cognition, and oscillopathies contribute to cognitive dysfunction in neuropsychiatric diseases. Cannabis-1 receptor (CB1-R) activation was reported to suppress theta and fast gamma activities in rats during waking exploration, and here, we show that cannabis fundamentally alters network activity during sleep as well. Prominent theta rhythm is present in rapid eye movement sleep (REMS), whereas fast oscillations appear as regular sequences of sleep spindles during intermediate sleep (IS)-both implicated in dreaming and memory consolidation. The CB1-R agonist disrupted these mechanisms, restructuring IS-REMS episodes; IS lengthened sixfold and intruded REMS, where ongoing theta was drastically reduced. The spindle architecture was also affected; its amplitude increased, and its peak frequency downshifted into the theta range. Cannabis is known to induce psychotic-like conditions and cognitive deficits; thus, our results may help in understanding the dual effect of cannabis on cognitive states and the role of network oscillations in psychiatric pathology.
Collapse
Affiliation(s)
- Irina Topchiy
- Department Psychiatry, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Department Psychiatry, Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Boston, MA02478
| | - Bernat Kocsis
- Department Psychiatry, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| |
Collapse
|
11
|
van Hasselt SJ, Martinez-Gonzalez D, Mekenkamp GJ, Vyssotski AL, Verhulst S, Beckers GJL, Rattenborg NC, Meerlo P. Sleep pressure causes birds to trade asymmetric sleep for symmetric sleep. Curr Biol 2025; 35:1918-1926.e3. [PMID: 40168984 DOI: 10.1016/j.cub.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/14/2025] [Accepted: 03/06/2025] [Indexed: 04/03/2025]
Abstract
Sleep is a dangerous part of an animal's life.1,2,3 Nonetheless, following sleep loss, mammals and birds sleep longer and deeper, as reflected by increased electroencephalogram (EEG) slow-wave activity (SWA; ≈1-5 Hz spectral power) during non-rapid eye movement (NREM) sleep.4,5 Stimulating a brain region during wakefulness also causes that region to sleep deeper afterwards,6,7,8,9 indicating that NREM sleep is a local, homeostatically regulated process.10,11 Birds and some marine mammals can keep one eye open during NREM sleep,12,13 a behavior associated with lighter sleep or wakefulness in the hemisphere opposite the open eye-states called asymmetric and unihemispheric NREM sleep, respectively.13,14,15,16,17,18,19,20,21,22,23 Closure of both eyes is associated with symmetric NREM or REM sleep. Birds rely on asymmetric and unihemispheric sleep to stay safe.17,24,25 However, as sleeping deeply with only one hemisphere at a time increases the time required for both hemispheres to fulfill their need for NREM sleep, increased sleep pressure might cause birds to engage in symmetric sleep at the expense of asymmetric sleep.26,27 Using high-density EEG recordings of European jackdaws (Coloeus monedula), we investigated intra- and inter-hemispheric asymmetries during normal sleep and following sleep deprivation (SD). The proportion of asymmetric sleep was lower early in the sleep period and following SD-periods of increased sleep pressure. Our findings demonstrate a trade-off between the benefits of sleep and vigilance and indicate that a bird's utilization of asymmetric sleep is constrained by temporal dynamics in their need for sleep.
Collapse
Affiliation(s)
- Sjoerd J van Hasselt
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 79747 Groningen, the Netherlands
| | - Dolores Martinez-Gonzalez
- Max Planck Institute for Biological Intelligence, Avian Sleep, Eberhard-Gwinner-Straβe 5, 82319 Seewiesen, Germany
| | - Gert-Jan Mekenkamp
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 79747 Groningen, the Netherlands
| | - Alexei L Vyssotski
- Institute of Neuroinformatics, University of Zürich, Swiss Federal Institute of Technology (ETH) Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Simon Verhulst
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 79747 Groningen, the Netherlands
| | - Gabriël J L Beckers
- Experimental Psychology and Helmholtz Institute, Utrecht University, Yalelaan 2, 3584 CM Utrecht, the Netherlands
| | - Niels C Rattenborg
- Max Planck Institute for Biological Intelligence, Avian Sleep, Eberhard-Gwinner-Straβe 5, 82319 Seewiesen, Germany.
| | - Peter Meerlo
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 79747 Groningen, the Netherlands.
| |
Collapse
|
12
|
Martínez-Moreno A, Terán-Pérez G, Arana-Lechuga Y, Velázquez-Moctezuma J, Sánchez-Escandón O, Guarneros-Roniger D, Mercadillo RE. Enhancing Mental Health and Cognitive Functioning in Victims of Violence: Cognitive Behavioral Therapy for Sleep Disorders Among Journalists, Human Rights Defenders, and Relatives of Disappeared Persons in Mexico City. Behav Sci (Basel) 2025; 15:530. [PMID: 40282151 PMCID: PMC12024299 DOI: 10.3390/bs15040530] [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: 11/13/2024] [Revised: 02/27/2025] [Accepted: 03/10/2025] [Indexed: 04/29/2025] Open
Abstract
In Mexico, pervasive violence, forced disappearances, and homicides have deeply impacted certain groups, particularly journalists, activists, and human rights defenders, who are at high risk of victimization. While these groups receive state support for physical and legal safety, mental health and sleep-focused interventions remain insufficient. Collaborating with a Mexico City-based institution supporting human rights defenders and journalists, we conducted a psychometric assessment of 47 individuals affected by violence. Results showed that 80% exhibited symptoms of post-traumatic stress disorder, 25.5% showed depression, and 57.4% displayed anxiety; 95.7% reported poor sleep quality based on the Pittsburgh Sleep Quality Index. In a second phase, neuropsychological tests and polysomnographic recordings identified cognitive impairments in attention, memory, and decision-making in some participants, along with sleep disorders such as insomnia, primary snoring, obstructive sleep apnea, and bruxism. A third phase introduced Cognitive Behavioral Therapy for insomnia, nightmares, and circadian rhythm issues. Results showed improvements in sleep quality, total sleep time, and a reduction in depression, anxiety, and post-traumatic stress disorder symptoms. This approach suggests that treating sleep issues in high-risk populations can improve mental health.
Collapse
Affiliation(s)
- Araceli Martínez-Moreno
- Neurological and Sleep Center, Benito Juárez, Mexico City 03020, Mexico; (A.M.-M.); (G.T.-P.); (Y.A.-L.); (J.V.-M.); (O.S.-E.)
| | - Guadalupe Terán-Pérez
- Neurological and Sleep Center, Benito Juárez, Mexico City 03020, Mexico; (A.M.-M.); (G.T.-P.); (Y.A.-L.); (J.V.-M.); (O.S.-E.)
| | - Yoaly Arana-Lechuga
- Neurological and Sleep Center, Benito Juárez, Mexico City 03020, Mexico; (A.M.-M.); (G.T.-P.); (Y.A.-L.); (J.V.-M.); (O.S.-E.)
| | - Javier Velázquez-Moctezuma
- Neurological and Sleep Center, Benito Juárez, Mexico City 03020, Mexico; (A.M.-M.); (G.T.-P.); (Y.A.-L.); (J.V.-M.); (O.S.-E.)
- Area of Neurosciences, Biology of Reproduction Department, Universidad Autónoma Metropolitana, Iztapalapa, Mexico City 09340, Mexico
- Sleep Disorders Clinic, Universidad Autónoma Metropolitana, Iztapalapa, Mexico City 09340, Mexico
| | - Oscar Sánchez-Escandón
- Neurological and Sleep Center, Benito Juárez, Mexico City 03020, Mexico; (A.M.-M.); (G.T.-P.); (Y.A.-L.); (J.V.-M.); (O.S.-E.)
- Neurophysiology Service, ABC Hospital, Mexico City 05330, Mexico
| | | | - Roberto E. Mercadillo
- Consejo Nacional de Humanidades, Ciencias y Tecnologías, CONAHCYT, Benito Juárez, Mexico City 03940, Mexico
| |
Collapse
|
13
|
Deng Z, Fei X, Zhang S, Xu M. A time window for memory consolidation during NREM sleep revealed by cAMP oscillation. Neuron 2025:S0896-6273(25)00220-X. [PMID: 40233747 DOI: 10.1016/j.neuron.2025.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 01/29/2025] [Accepted: 03/14/2025] [Indexed: 04/17/2025]
Abstract
Memory formation requires specific neural activity in coordination with intracellular signaling mediated by second messengers such as cyclic adenosine monophosphate (cAMP). However, the real-time dynamics of cAMP remain largely unknown. Here, using a genetically encoded cAMP sensor with high temporal resolution, we found neural-activity-dependent rapid cAMP elevation during learning. Interestingly, in slow-wave sleep, during which memory consolidation occurs, the cAMP level in mice was anti-correlated with neural activity and exhibited norepinephrine β1 receptor-dependent infra-slow oscillations that were synchronized across the hippocampus and cortex. Furthermore, the hippocampal-cortical interactions increased during the narrow time-window of the peak cAMP level; suppressing hippocampal activity specifically during this window impaired spatial memory consolidation. Thus, hippocampal-dependent memory consolidation occurs within a specific time window of high cAMP activity during slow-wave sleep.
Collapse
Affiliation(s)
- Ziru Deng
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiang Fei
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Siyu Zhang
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Min Xu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
14
|
Lucey BP. Sleep Alterations and Cognitive Decline. Semin Neurol 2025. [PMID: 40081821 DOI: 10.1055/a-2557-8422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Sleep disturbances and cognitive decline are intricately connected, and both are prevalent in aging populations and individuals with neurodegenerative disorders such as Alzheimer's disease (AD) and other dementias. Sleep is vital for cognitive functions including memory consolidation, executive function, and attention. Disruption in these processes is associated with cognitive decline, although causal evidence is mixed. This review delves into the bidirectional relationship between alterations in sleep and cognitive impairment, exploring key mechanisms such as amyloid-β accumulation, tau pathology, synaptic homeostasis, neurotransmitter dysregulation, oxidative stress, and vascular contributions. Evidence from both experimental research and population-based studies underscores the necessity of early interventions targeting sleep to mitigate risks of neurodegenerative diseases. A deeper understanding of the interplay between sleep and cognitive health may pave the way for innovative strategies to prevent or reduce cognitive decline through improved sleep management.
Collapse
Affiliation(s)
- Brendan P Lucey
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri
- Center On Biological Rhythms and Sleep, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
15
|
Turi GF, Teng S, Chen X, Lim ECY, Dias C, Hu R, Wang R, Zhen F, Peng Y. Serotonin modulates infraslow oscillation in the dentate gyrus during non-REM sleep. eLife 2025; 13:RP100196. [PMID: 40178074 PMCID: PMC11968106 DOI: 10.7554/elife.100196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Synchronous neuronal activity is organized into neuronal oscillations with various frequency and time domains across different brain areas and brain states. For example, hippocampal theta, gamma, and sharp wave oscillations are critical for memory formation and communication between hippocampal subareas and the cortex. In this study, we investigated the neuronal activity of the dentate gyrus (DG) with optical imaging tools during sleep-wake cycles in mice. We found that the activity of major glutamatergic cell populations in the DG is organized into infraslow oscillations (0.01-0.03 Hz) during NREM sleep. Although the DG is considered a sparsely active network during wakefulness, we found that 50% of granule cells and about 25% of mossy cells exhibit increased activity during NREM sleep, compared to that during wakefulness. Further experiments revealed that the infraslow oscillation in the DG was correlated with rhythmic serotonin release during sleep, which oscillates at the same frequency but in an opposite phase. Genetic manipulation of 5-HT receptors revealed that this neuromodulatory regulation is mediated by Htr1a receptors and the knockdown of these receptors leads to memory impairment. Together, our results provide novel mechanistic insights into how the 5-HT system can influence hippocampal activity patterns during sleep.
Collapse
Affiliation(s)
- Gergely F Turi
- New York State Psychiatric Institute, Division of Systems Neuroscience New YorkNew YorkUnited States
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
| | - Sasa Teng
- Institute for Genomic Medicine, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
| | - Xinyue Chen
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
- Department of Neuroscience, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
| | - Emily CY Lim
- Columbia College, Columbia UniversityNew YorkUnited States
| | - Carla Dias
- New York State Psychiatric Institute, Division of Systems Neuroscience New YorkNew YorkUnited States
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
| | - Ruining Hu
- Institute for Genomic Medicine, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
| | - Ruizhi Wang
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
| | - Fenghua Zhen
- Institute for Genomic Medicine, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
| | - Yueqing Peng
- Institute for Genomic Medicine, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia UniversityNew YorkUnited States
| |
Collapse
|
16
|
Chen H, Wang B, Zhan Y, Liu J, Yang S, Tan X, Zhang W, Zhang J, Yang Y, Liu Y, Wang M, Zhang H, Li X, Yao Z, Pema D, Li H, Chen H, Hu B. Dynamics of hippocampal reactivation for temporal association memory in mice. Prog Neurobiol 2025; 247:102729. [PMID: 40023311 DOI: 10.1016/j.pneurobio.2025.102729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 03/04/2025]
Abstract
Reactivation refers to the re-emergence of activity in neuronal ensembles that were active during information encoding. Hippocampal CA1 neuronal ensembles generate firing activities that encode the temporal association among time-separated events. However, whether and how temporal association memory-related CA1 neuronal ensembles reactivate during sleep and their role in temporal association memory consolidation remain unclear. We utilized multiple unit recordings to monitor CA1 neuronal activity in mice learning a trace eyeblink conditioning (tEBC) task, in which presentation of the conditioned stimulus (CS, a light flash) was paired with presentation of the unconditioned stimulus (US, corneal puff) by a time-separated interval. We found that the CS-US paired training mice exhibited few conditioned eyeblink responses (CRs) at the initial-learning stage (ILS) and an asymptotic level of CRs at the well-learning stage (WLS). More than one third of CA1 pyramidal cells (PYR) in the CS-US paired training mice manifested a CS-evoked firing activity that was sustained from the CS to time-separated interval. The CS-evoked PYR firing activity was required for the tEBC acquisition and was greater when the CRs occurred. Intriguingly, the CS-evoked firing PYR ensembles reactivated, which coincided with increased hippocampal ripples during post-training sleep. The reactivation of CS-evoked firing PYR ensembles diminished across learning stages, with greater strength in the ILS. Disrupting the ripple-associated PYR activity impaired both the reactivation of CS-evoked firing PYR ensembles and tEBC consolidation. Our findings highlight the features of hippocampal CA1 neuronal ensemble reactivation during sleep, which support the consolidation of temporal association memory.
Collapse
Affiliation(s)
- Hui Chen
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China; Department of Radiology, 7T Magnetic Resonance Translational Medicine Research Center, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Bin Wang
- Department of Histology and Embryology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yue Zhan
- Department of Histology and Embryology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Junqi Liu
- Department of Histology and Embryology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Sicheng Yang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Xuan Tan
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Weiwei Zhang
- Department of Physiology, School of Basic Medicine, Sichuan College of Traditional Chinese Medicine, Mianyang 621000, China
| | - Jie Zhang
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Ye Yang
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yanji Liu
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Meilin Wang
- Department of Histology and Embryology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Haibo Zhang
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Xuan Li
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Zhongxiang Yao
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Drolma Pema
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Hongli Li
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China.
| | - Hao Chen
- Department of Histology and Embryology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China; Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China.
| | - Bo Hu
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
17
|
Saito Y, Osako Y, Odagawa M, Oisi Y, Matsubara C, Kato S, Kobayashi K, Morita M, Johansen JP, Murayama M. Amygdalo-cortical dialogue underlies memory enhancement by emotional association. Neuron 2025; 113:931-948.e7. [PMID: 39884277 DOI: 10.1016/j.neuron.2025.01.001] [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: 01/19/2024] [Revised: 11/15/2024] [Accepted: 01/03/2025] [Indexed: 02/01/2025]
Abstract
Emotional arousal plays a critical role in determining what is remembered from experiences. It is hypothesized that activation of the amygdala by emotional stimuli enhances memory consolidation in its downstream brain regions. However, the physiological basis of the inter-regional interaction and its functions remain unclear. Here, by adding emotional information to a perceptual recognition task that relied on a frontal-sensory cortical circuit in mice, we demonstrated that the amygdala not only associates emotional information with perceptual information but also enhances perceptual memory retention via amygdalo-frontal cortical projections. Furthermore, emotional association increased reactivation of coordinated activity across the amygdalo-cortical circuit during non-rapid eye movement (NREM) sleep but not during rapid eye movement (REM) sleep. Notably, this increased reactivation was associated with amygdala high-frequency oscillations. Silencing of amygdalo-cortical inputs during NREM sleep selectively disrupted perceptual memory enhancement. Our findings indicate that inter-regional reactivation triggered by the amygdala during NREM sleep underlies emotion-induced perceptual memory enhancement.
Collapse
Affiliation(s)
- Yoshihito Saito
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Wako-shi 351-0198, Saitama, Japan; RIKEN CBS-Kao Collaboration Center (BKCC), Wako-shi 351-0198, Saitama, Japan; Department of Biology, Graduate School of Science, Kobe University, Kobe-shi 657-8501, Hyogo, Japan
| | - Yuma Osako
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Wako-shi 351-0198, Saitama, Japan; Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Maya Odagawa
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Wako-shi 351-0198, Saitama, Japan; RIKEN CBS-Kao Collaboration Center (BKCC), Wako-shi 351-0198, Saitama, Japan
| | - Yasuhiro Oisi
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Wako-shi 351-0198, Saitama, Japan; RIKEN CBS-Kao Collaboration Center (BKCC), Wako-shi 351-0198, Saitama, Japan
| | - Chie Matsubara
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Wako-shi 351-0198, Saitama, Japan; RIKEN CBS-Kao Collaboration Center (BKCC), Wako-shi 351-0198, Saitama, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, School of Medicine, Fukushima Medical University, Fukushima-shi 960-1295, Fukushima, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, School of Medicine, Fukushima Medical University, Fukushima-shi 960-1295, Fukushima, Japan
| | - Mitsuhiro Morita
- Department of Biology, Graduate School of Science, Kobe University, Kobe-shi 657-8501, Hyogo, Japan
| | - Joshua P Johansen
- Laboratory for the Neural Circuitry of Learning and Memory, RIKEN Center for Brain Science, Wako-shi 351-0198, Saitama, Japan
| | - Masanori Murayama
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Wako-shi 351-0198, Saitama, Japan; RIKEN CBS-Kao Collaboration Center (BKCC), Wako-shi 351-0198, Saitama, Japan.
| |
Collapse
|
18
|
Silva A, Girardeau G. Sweet dreams are made of this: How emotions influence sleep-dependent consolidation of perceptual memories. Neuron 2025; 113:803-805. [PMID: 40112774 DOI: 10.1016/j.neuron.2025.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
Emotions strongly modulate long-term memory retention, yet the underlying mechanisms remain unclear. Using causal and correlational approaches, Saito et al.1 found that basolateral amygdala inputs to the M2-S1 cortical loop enhance the sleep-dependent consolidation of a perceptual-emotional associative memory.
Collapse
Affiliation(s)
- Azul Silva
- Sorbonne Université, CNRS, Inserm, Center of Neuroscience Neuro-SU, 75005 Paris, France; Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-Seine, IBPS, 75005 Paris, France
| | - Gabrielle Girardeau
- Sorbonne Université, CNRS, Inserm, Center of Neuroscience Neuro-SU, 75005 Paris, France; Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-Seine, IBPS, 75005 Paris, France.
| |
Collapse
|
19
|
Smolensky MH, Hermida RC, Castriotta RJ, Geng YJ. Findings and Methodological Shortcomings of Investigations Concerning the Relationship Between Sleep Duration and Blood Pressure: A Comprehensive Narrative Review. J Cardiovasc Dev Dis 2025; 12:95. [PMID: 40137093 PMCID: PMC11943021 DOI: 10.3390/jcdd12030095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/03/2025] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Cardiology and sleep societies recommend 7-9 h sleep/night for adults (7-8 h for seniors) and more for youngsters; nonetheless, short sleep duration (SSD) of <7 h/night is epidemic. We searched PubMed for representative investigations, including those cited by meta-analyses, that reported association between SSD and long sleep duration (LSD) of >9 h/night and blood pressure (BP) levels to assess shortcomings of their methods. Studies indicate both SSD and LSD negatively impact BP despite major deficiencies, such as (i) reliance mainly on cross-sectional rather than longitudinal protocols, (ii) inclusion of participants diagnosed with hypertension (HTN) and/or taking antihypertension medications, (iii) assessment of BP and diagnosis of HTN performed by single wake-time office measurement rather than multiple measurements performed by 24 h ambulatory BP monitoring (ABPM), and (iv) determination of SD by subjective recall, single-night polysomnography, or diary recordings rather than objective wrist actigraphy of sufficient duration. The limited number of ABPM-based studies, despite evidencing major shortcomings, particularly (i) assessment for 24 h rather than preferred ≥48 h and (ii) inclusion of subjects diagnosed with HTN and/or taking antihypertension medications, also report association between abnormal SD and elevated 24 h 'daytime'/wake-time diastolic and systolic (SBP) means plus 'nighttime'/sleep-time SBP mean and dipping-the latter two indices, in combination, the strongest predictors of major adverse cardiovascular events.
Collapse
Affiliation(s)
- Michael H. Smolensky
- Department of Biomedical Engineering, Cockell School of Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (R.C.H.); (Y.-J.G.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern School of Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ramón C. Hermida
- Department of Biomedical Engineering, Cockell School of Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (R.C.H.); (Y.-J.G.)
- Bioengineering & Chronobiology Laboratories, Atlantic Research Center for Telecommunication Technologies, Universidade de Vigo, 36310 Vigo, Spain
- Bioengineering & Chronobiology Research Group, Galicia Sur Health Research Institute, 36310 Vigo, Spain
| | - Richard J. Castriotta
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
| | - Yong-Jian Geng
- Department of Biomedical Engineering, Cockell School of Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (R.C.H.); (Y.-J.G.)
- Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern School of Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- The Texas Heart Institute at Baylor St. Luke’s Medical Center, Houston, TX 77030, USA
| |
Collapse
|
20
|
Solbi A, Earle FS. The Role of Sleep in Memory Consolidation and Reading in Dyslexia. J Cogn Neurosci 2025; 37:532-542. [PMID: 39620967 PMCID: PMC12097521 DOI: 10.1162/jocn_a_02282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
Dyslexia is a neurodevelopmental disorder characterized by reading difficulty, which has long been attributed to a phonological processing deficit. However, recent research suggests that general difficulties with learning and memory, but also in memory consolidation, may underlie disordered reading. This review article provides an overview of the relationship between learning and memory, memory consolidation during sleep, and reading and explores the emerging literature on consolidation during sleep in individuals with dyslexia. We consider evidence that sleep appears to be less effective for memory consolidation in children with dyslexia and how this may be related to their deficits in reading. This discussion highlights the need for further research to determine the extent to which atypical sleep patterns may contribute to learning deficits associated with disordered reading.
Collapse
|
21
|
Lüthi A, Nedergaard M. Anything but small: Microarousals stand at the crossroad between noradrenaline signaling and key sleep functions. Neuron 2025; 113:509-523. [PMID: 39809276 DOI: 10.1016/j.neuron.2024.12.009] [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: 01/29/2024] [Revised: 09/16/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025]
Abstract
Continuous sleep restores the brain and body, whereas fragmented sleep harms cognition and health. Microarousals (MAs), brief (3- to 15-s-long) wake intrusions into sleep, are clinical markers for various sleep disorders. Recent rodent studies show that MAs during healthy non-rapid eye movement (NREM) sleep are driven by infraslow fluctuations of noradrenaline (NA) in coordination with electrophysiological rhythms, vasomotor activity, cerebral blood volume, and glymphatic flow. MAs are hence part of healthy sleep dynamics, raising questions about their biological roles. We propose that MAs bolster NREM sleep's benefits associated with NA fluctuations, according to an inverted U-shaped curve. Weakened noradrenergic fluctuations, as may occur in neurodegenerative diseases or with sleep aids, reduce MAs, whereas exacerbated fluctuations caused by stress fragment NREM sleep and collapse NA signaling. We suggest that MAs are crucial for the restorative and plasticity-promoting functions of sleep and advance our insight into normal and pathological arousal dynamics from sleep.
Collapse
Affiliation(s)
- Anita Lüthi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.
| | - Maiken Nedergaard
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
22
|
Liu YC, Deng YC, Zhu ZT, Rao B, Shang HL, Wang LK, Li T, Wang YR, Wang JZ, Zhang QP, Gao Y, Xu HB. Oxytocin modulates inhibitory balance in the prelimbic cortex to support social memory consolidation during REM sleep. Theranostics 2025; 15:3257-3274. [PMID: 40093885 PMCID: PMC11905142 DOI: 10.7150/thno.109104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 01/26/2025] [Indexed: 03/19/2025] Open
Abstract
Rationale: The prelimbic cortex (PrL), enriched with oxytocin (OXT) receptors, plays a critical role in memory consolidation. However, the role of OXT in social memory consolidation within the PrL microcircuit remains poorly understood. Methods: To examine the role of OXT signaling in social memory consolidation, we used OXT biosensors and loss-of-function approaches, including tetanus toxin-mediated silencing of OXT neurons in the paraventricular nucleus (PVNOXT), optogenetic inhibition of the PVNOXT-PrL pathway during rapid-eye-movement (REM) sleep, and local administration of an OXT receptor antagonist in the PrL. In vivo molecular biosensors for vasoactive intestinal peptide (VIP), somatostatin, and presynaptic calcium imaging were employed to assess inhibitory signaling in the PrL microcircuit. Optogenetic activation of the PVNOXT-PrL pathway and intranasal OXT were used to evaluate resilience to chronic sleep deprivation-induced social memory deficits. Results: We identified that REM-sleep OXT release via the PVN to PrL pathway supports social memory consolidation. OXT signaling deficiency reduces the activity of VIP and parvalbumin (PV) neurons, thereby disrupting the inhibitory balance between somatic inhibition mediated by PV neurons and dendritic disinhibition mediated by VIP neurons in PrL microcircuits during REM sleep. Chronic sleep deprivation (SD) disrupts OXT release and inhibitory balance, leading to pyramidal neuron hyperactivity and social memory impairments. Notably, REM-sleep-specific activation of the PVNOXT-PrL pathway or intranasal OXT restores inhibitory balance and rescues social memory deficits in SD mice. Conclusion: Our results reveal how OXT modulates inhibitory balance in the PrL microcircuit to support social memory consolidation during REM sleep, suggesting potential therapeutic strategies for treating sleep-related memory disorders.
Collapse
Affiliation(s)
- Yan-chao Liu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yu-chen Deng
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Zi-tao Zhu
- Second Clinical College, Wuhan University, Wuhan, 430071, China
| | - Bo Rao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Hong-lei Shang
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Li-ke Wang
- Second Clinical College, Wuhan University, Wuhan, 430071, China
| | - Tao Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Ya-rong Wang
- Li-Yuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qing-ping Zhang
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yang Gao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Hai-bo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan, 430071, China
- Wuhan clinical research and development center of brain resuscitation and functional imaging, Wuhan, 430071, China
| |
Collapse
|
23
|
Maski K, Heckler G, Worhach J, Mylonas D, Wang G, Szilagyi K, Zhang B, Diniz Behn C, Scammell TE, Stickgold R. Impaired sleep-dependent memory consolidation in pediatric narcolepsy type 1. Sleep 2025; 48:zsae238. [PMID: 39420719 PMCID: PMC11807881 DOI: 10.1093/sleep/zsae238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 09/15/2024] [Indexed: 10/19/2024] Open
Abstract
STUDY OBJECTIVES Disrupted nighttime sleep is common in pediatric narcolepsy type 1, yet its cognitive impact is unknown. As N2 sleep spindles are necessary for sleep-dependent memory consolidation, we hypothesized that narcolepsy type 1 impairs memory consolidation via N2 sleep fragmentation and N2 sleep spindle alterations. METHODS We trained 28 pediatric narcolepsy type 1 participants and 27 healthy controls (HCs) on a spatial declarative memory task before a nocturnal in-lab polysomnogram and then gave them a cued recall test upon awakening in the morning. We extracted wake and sleep stage bout numbers and N2 spindle characteristics from the polysomnogram and conducted mixed model analysis of sleep-dependent memory consolidation to identify group differences. RESULTS Narcolepsy type 1 participants had shorter N2 bout durations and associated shorter N2 spindles versus HC, but other N2 spindle features were similar. Narcolepsy type 1 participants had worse memory performance postsleep than HCs after adjusting for age and gender (mean memory consolidation HC: -3.1% ± 18.7, NT1: -15.6 ± 24.8, main effect group × time of testing F = 5.3, p = .03). We did not find significant relationships between sleep-dependent memory consolidation and N2 spindle characteristics. Notably, increased N1% was associated with worse sleep-dependent memory consolidation with results driven by the narcolepsy type 1 group. CONCLUSIONS Sleep-dependent memory consolidation is mildly impaired in youth with narcolepsy type 1 and findings may be attributed to increases in N1 sleep. Further studies are needed to determine if these findings are generalizable and reversible with sleep-based therapies.
Collapse
Affiliation(s)
- Kiran Maski
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
| | - Gillian Heckler
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
| | - Jennifer Worhach
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
| | - Dimitrios Mylonas
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MAUSA
| | - Grace Wang
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
| | - Klara Szilagyi
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
| | - Bo Zhang
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
| | - Cecilia Diniz Behn
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Robert Stickgold
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
24
|
Balsamo F, Meneo D, Berretta E, Baglioni C, Gelfo F. Could sleep be a brain/cognitive/neural reserve-builder factor? A systematic review on the cognitive effects of sleep modulation in animal models. Neurosci Biobehav Rev 2025; 169:106015. [PMID: 39828234 DOI: 10.1016/j.neubiorev.2025.106015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
The brain/cognitive/neural reserve concept suggests that lifelong experiences, from early life through adulthood, make the brain more resilient to neuronal damage. Modifiable lifestyle factors, such as sleep, can support the development and enhance such a reserve, helping to counteract age- or disease-related brain changes and their impact on cognition. Sleep plays a crucial role in cognitive functioning, and disruptions or disorders may increase neurodegenerative risks. This systematic review aims to explore how functional and disturbed sleep impacts cognitive functions and neuromorphological mechanisms in rodents, aiming to better understand its role in brain/cognitive/neural reserve development. This systematic review, registered on PROSPERO (ID: CRD42023423901) and conducted according to PRISMA-P guidelines, searched PubMed, Scopus, Web of Science, and Embase databases for studies up to June 2022, with terms related to sleep, rodents, and cognitive functions. Of the 28,666 articles identified, 142 met the inclusion criteria. Main results showed significant cognitive decline after sleep deprivation, especially in memory performance. These findings supports the importance of sleep as a critical factor in modulating brain/cognitive/neural reserve.
Collapse
Affiliation(s)
- Francesca Balsamo
- Department of Human Sciences, Guglielmo Marconi University, Rome 00193, Italy; IRCCS Fondazione Santa Lucia, Rome 00179, Italy.
| | - Debora Meneo
- Department of Human Sciences, Guglielmo Marconi University, Rome 00193, Italy
| | | | - Chiara Baglioni
- Department of Human Sciences, Guglielmo Marconi University, Rome 00193, Italy; Department of Clinical Psychology and Psychophysiology/Sleep, Medicine, Centre for Mental Disorders, University Medical Centre, Freiburg, Germany
| | - Francesca Gelfo
- Department of Human Sciences, Guglielmo Marconi University, Rome 00193, Italy; IRCCS Fondazione Santa Lucia, Rome 00179, Italy.
| |
Collapse
|
25
|
Zhu L, Yao D. Research advances in children's sleep and vitamin D levels. Ann Pediatr Endocrinol Metab 2025; 30:3-10. [PMID: 40049669 PMCID: PMC11917401 DOI: 10.6065/apem.2448076.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/05/2024] [Indexed: 03/20/2025] Open
Abstract
In recent years, studies have revealed that vitamin D, a steroid hormone essential for calcium and phosphorus metabolism, also plays a role in sleep. Adequate levels of vitamin D have been linked to improved sleep quality in children and effective prevention of sleep problems. This report is a review and summary of research on the significance of sleep, the stages of children's sleep, and the impact of vitamin D levels on sleep problems. Additionally, this report explores the mechanisms through which vitamin D improves sleep.
Collapse
Affiliation(s)
- Liuyan Zhu
- Department of Pediatric Health Care, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Clinical Research Center for Child Health, Hangzhou, China
| | - Dan Yao
- Department of Pediatric Health Care, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
26
|
Arnold JC, Occelli Hanbury-Brown CV, Anderson LL, Bedoya-Pérez MA, Udoh M, Sharman LA, Raymond JS, Doohan PT, Ametovski A, McGregor IS. A sleepy cannabis constituent: cannabinol and its active metabolite influence sleep architecture in rats. Neuropsychopharmacology 2025; 50:586-595. [PMID: 39528623 PMCID: PMC11736144 DOI: 10.1038/s41386-024-02018-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Medicinal cannabis is being used worldwide and there is increasing use of novel cannabis products in the community. Cannabis contains the major cannabinoids, Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD), but also an array of minor cannabinoids that have undergone much less pharmacological characterization. Cannabinol (CBN) is a minor cannabinoid used in the community in "isolate' products and is claimed to have pro-sleep effects comparable to conventional sleep medications. However, no study has yet examined whether it impacts sleep architecture using objective sleep measures. The effects of CBN on sleep in rats using polysomnography were therefore examined. CBN increased total sleep time, although there was evidence of biphasic effects with initial sleep suppression before a dramatic increase in sleep. CBN increased both non-rapid eye movement (NREM) and rapid eye movement (REM) sleep. The magnitude of the effect of CBN on NREM was comparable to the sleep aid zolpidem, although, unlike CBN, zolpidem did not influence REM sleep. Following CBN dosing, 11-hydroxy-CBN, a primary metabolite of CBN surprisingly attained equivalently high brain concentrations to CBN. 11-hydroxy-CBN was active at cannabinoid CB1 receptors with comparable potency and efficacy to Δ9-THC, however, CBN had much lower activity. We then discovered that the metabolite 11-hydroxy-CBN also influenced sleep architecture, albeit with some subtle differences from CBN itself. This study shows CBN affects sleep using objective sleep measures and suggests an active metabolite may contribute to its hypnotic action.
Collapse
Affiliation(s)
- Jonathon C Arnold
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia.
- Discipline of Pharmacology, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.
| | - Cassandra V Occelli Hanbury-Brown
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Discipline of Pharmacology, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Lyndsey L Anderson
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Discipline of Pharmacology, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Miguel A Bedoya-Pérez
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- School of Psychology, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Michael Udoh
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Discipline of Pharmacology, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Laura A Sharman
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Discipline of Pharmacology, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Joel S Raymond
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- School of Psychology, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Peter T Doohan
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Discipline of Pharmacology, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Adam Ametovski
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- School of Chemistry, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Iain S McGregor
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- School of Psychology, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
27
|
Turi GF, Teng S, Chen X, Lim ECY, Dias C, Hu R, Wang R, Zhen F, Peng Y. Serotonin modulates infraslow oscillation in the dentate gyrus during Non-REM sleep. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.05.12.540575. [PMID: 38854102 PMCID: PMC11160574 DOI: 10.1101/2023.05.12.540575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Synchronous neuronal activity is organized into neuronal oscillations with various frequency and time domains across different brain areas and brain states. For example, hippocampal theta, gamma and sharp wave oscillations are critical for memory formation and communication between hippocampal subareas and the cortex. In this study, we investigated the neuronal activity of the dentate gyrus (DG) with optical imaging tools during sleep-wake cycles. We found that the activity of major glutamatergic cell populations in the DG is organized into infraslow oscillations (0.01 - 0.03 Hz) during NREM sleep. Although the DG is considered a sparsely active network during wakefulness, we found that 50% of granule cells and about 25% of mossy cells exhibit increased activity during NREM sleep, compared to that during wakefulness. Further experiments revealed that the infraslow oscillation in the DG was correlated with rhythmic serotonin release during sleep, which oscillates at the same frequency but in an opposite phase. Genetic manipulation of 5-HT receptors revealed that this neuromodulatory regulation is mediated by 5-HT1a receptors and the knockdown of these receptors leads to memory impairment. Together, our results provide novel mechanistic insights into how the 5-HT system can influence hippocampal activity patterns during sleep.
Collapse
Affiliation(s)
- Gergely F. Turi
- New York State Psychiatric Institute, Division of Systems Neuroscience New York, NY 10032, USA
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Sasa Teng
- Institute for Genomic Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Xinyue Chen
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Department of Neuroscience, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Emily CY Lim
- Columbia College, Columbia University, New York, NY 10027, USA
| | - Carla Dias
- New York State Psychiatric Institute, Division of Systems Neuroscience New York, NY 10032, USA
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Ruining Hu
- Institute for Genomic Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Ruizhi Wang
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Fenghua Zhen
- Institute for Genomic Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Present address: National Institute of Allergy and Infectious Diseases, Bethesda, MD, 20894, USA
| | - Yueqing Peng
- Institute for Genomic Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
28
|
E Said S, Miyamoto D. Multi-region processing during sleep for memory and cognition. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2025; 101:107-128. [PMID: 40074337 DOI: 10.2183/pjab.101.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Over the past decades, the understanding of sleep has evolved to be a fundamental physiological mechanism integral to the processing of different types of memory rather than just being a passive brain state. The cyclic sleep substates, namely, rapid eye movement (REM) sleep and non-REM (NREM) sleep, exhibit distinct yet complementary oscillatory patterns that form inter-regional networks between different brain regions crucial to learning, memory consolidation, and memory retrieval. Technical advancements in imaging and manipulation approaches have provided deeper understanding of memory formation processes on multi-scales including brain-wide, synaptic, and molecular levels. The present review provides a short background and outlines the current state of research and future perspectives in understanding the role of sleep and its substates in memory processing from both humans and rodents, with a focus on cross-regional brain communication, oscillation coupling, offline reactivations, and engram studies. Moreover, we briefly discuss how sleep contributes to other higher-order cognitive functions.
Collapse
Affiliation(s)
- Salma E Said
- Laboratory for Sleeping-Brain Dynamics, Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Daisuke Miyamoto
- Laboratory for Sleeping-Brain Dynamics, Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| |
Collapse
|
29
|
Ognjanovski N, Kim DS, Charlett-Green E, Goldiez E, van Koppen S, Aton SJ, Watson BO. Daily rhythms drive dynamism in sleep, oscillations and interneuron firing, while excitatory firing remains stable across 24 h. Eur J Neurosci 2025; 61:e16619. [PMID: 39663213 DOI: 10.1111/ejn.16619] [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: 06/04/2024] [Revised: 10/25/2024] [Accepted: 11/10/2024] [Indexed: 12/13/2024]
Abstract
The adaptation to the daily 24-h light-dark cycle is ubiquitous across animal species and is crucial for maintaining fitness. This free-running cycle occurs innately within multiple bodily systems, such as endogenous circadian rhythms in clock-gene expression and synaptic plasticity. These phenomena are well studied; however, it is unknown if and how the 24-h clock affects electrophysiologic network function in vivo. The hippocampus is a region of interest for long timescale (>8 h) studies because it is critical for cognitive function and exhibits time-of-day effects in learning. We recorded single cell spiking activity and local field potentials (LFPs) in mouse hippocampus across the 24-h (12:12-h light/dark) cycle to quantify how electrophysiological network function is modulated across the 24-h day. We found that while inhibitory population firing rates and LFP oscillations exhibit modulation across the day, average excitatory population firing is static. This excitatory stability, despite inhibitory dynamism, may enable consistent around-the-clock function of neural circuits.
Collapse
Affiliation(s)
- Nicolette Ognjanovski
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
- Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - David S Kim
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| | - Emma Charlett-Green
- Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Ethan Goldiez
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| | - Sofie van Koppen
- Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Sara J Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Brendon O Watson
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
30
|
Cheng C, Jia M, Peng X, Sun Y, Jiao Y, Zhang M, Song X, Chu Z, Zeng X, Sun JB, Yang XJ, Qin W. Different regulative effects of high- and low-frequency external trigeminal nerve stimulation (eTNS) on sleep activity: Preliminary study. Sleep Med 2025; 125:136-145. [PMID: 39608185 DOI: 10.1016/j.sleep.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/28/2024] [Accepted: 11/07/2024] [Indexed: 11/30/2024]
Abstract
STUDY OBJECTIVE With the growing prominence of peripheral nerve stimulation technology, the clinical applications and potential neurophysiological mechanisms of external trigeminal nerve stimulation (eTNS) have garnered increasing attention. Despite its status as the sole neuromodulation method commonly employed in sleep, no studies have explored the effects of eTNS at varying frequencies on sleep activities. This study aims to investigate the regulatory effects of high-frequency and low-frequency eTNS on sleep activities using polysomnography. METHODS In this within-subjects experiment, 20 participants underwent a night of adaptation sleep, followed by 8-h sessions of sham, 120Hz-, and 2Hz-eTNS interventions in a randomized order in the sleep laboratory, with polysomnographic signals collected throughout. RESULTS The results indicated that 120Hz-eTNS significantly improved sleep efficiency, increased N2 sleep proportion, and reduced sleep latency, without significantly affecting sleep stage transition probabilities, sleep duration, or sleep-specific wave activities. Conversely, while 2Hz-eTNS did not impact sleep efficiency or latency, it increased the proportion of N3 sleep, stabilizes N3 sleep, and enhanced the survival probability of N3 and REM sleep duration. Additionally, it increases the density of slow oscillations (SOs), improved the coupling ratio of SO-spindles, and enhanced coupling timing accuracy. CONCLUSIONS These findings suggest that eTNS during sleep can indeed modulate sleep activities, with different frequencies exerting distinct regulatory effects. This may hold significant value for advancing the clinical application and efficacy of eTNS.
Collapse
Affiliation(s)
- Chen Cheng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, Shaan xi, 710126, China
| | - Mengnan Jia
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, Shaan xi, 710126, China
| | - Xiangmiao Peng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, Shaan xi, 710126, China
| | - Yuchen Sun
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, Shaan xi, 710126, China
| | - Yunyun Jiao
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, Shaan xi, 710126, China
| | - Mengkai Zhang
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, Shaan xi, 710126, China
| | - Xiaoyu Song
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, Shaan xi, 710126, China
| | - Zhaoyang Chu
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, Shaan xi, 710126, China
| | - Xiao Zeng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, Shaan xi, 710126, China; Guangzhou Institute of Technology, Xidian University, Xi'an, Shaanxi, China
| | - Jin-Bo Sun
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, Shaan xi, 710126, China; Guangzhou Institute of Technology, Xidian University, Xi'an, Shaanxi, China
| | - Xue-Juan Yang
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, Shaan xi, 710126, China; Guangzhou Institute of Technology, Xidian University, Xi'an, Shaanxi, China.
| | - Wei Qin
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, Shaan xi, 710126, China; Guangzhou Institute of Technology, Xidian University, Xi'an, Shaanxi, China
| |
Collapse
|
31
|
Rina AS, Moniruzzaman M, Muzammil MA. Commentary to the Editor regarding "Association between sleep disorders and constipation risk: A systematic review and meta-analysis". J Clin Neurosci 2025; 131:110842. [PMID: 39289104 DOI: 10.1016/j.jocn.2024.110842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
|
32
|
Sheriff A, Zhou G, Sagar V, Morgenthaler JB, Cyr C, Hauner KK, Omidbeigi M, Rosenow JM, Schuele SU, Lane G, Zelano C. Breathing orchestrates synchronization of sleep oscillations in the human hippocampus. Proc Natl Acad Sci U S A 2024; 121:e2405395121. [PMID: 39680758 PMCID: PMC11670218 DOI: 10.1073/pnas.2405395121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 11/07/2024] [Indexed: 12/18/2024] Open
Abstract
Nested sleep oscillations, emerging from asynchronous states in coordinated bursts, are critical for memory consolidation. Whether these bursts emerge intrinsically or from an underlying rhythm is unknown. Here, we show a previously undescribed respiratory-driven oscillation in the human hippocampus that couples with cardinal sleep oscillations. Further, breathing promotes nesting of ripples in slow oscillations, together suggesting that respiration acts as an intrinsic rhythm to coordinate synchronization of sleep oscillations, providing a unique framework to characterize sleep-related respiratory and memory processes.
Collapse
Affiliation(s)
- Andrew Sheriff
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Guangyu Zhou
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Vivek Sagar
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Justin B. Morgenthaler
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Christopher Cyr
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Katherina K. Hauner
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Mahmoud Omidbeigi
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Joshua M. Rosenow
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Stephan U. Schuele
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Gregory Lane
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Christina Zelano
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| |
Collapse
|
33
|
Mittermaier FX, Kalbhenn T, Xu R, Onken J, Faust K, Sauvigny T, Thomale UW, Kaindl AM, Holtkamp M, Grosser S, Fidzinski P, Simon M, Alle H, Geiger JRP. Membrane potential states gate synaptic consolidation in human neocortical tissue. Nat Commun 2024; 15:10340. [PMID: 39668146 PMCID: PMC11638263 DOI: 10.1038/s41467-024-53901-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 10/22/2024] [Indexed: 12/14/2024] Open
Abstract
Synaptic mechanisms that contribute to human memory consolidation remain largely unexplored. Consolidation critically relies on sleep. During slow wave sleep, neurons exhibit characteristic membrane potential oscillations known as UP and DOWN states. Coupling of memory reactivation to these slow oscillations promotes consolidation, though the underlying mechanisms remain elusive. Here, we performed axonal and multineuron patch-clamp recordings in acute human brain slices, obtained from neurosurgeries, to show that sleep-like UP and DOWN states modulate axonal action potentials and temporarily enhance synaptic transmission between neocortical pyramidal neurons. Synaptic enhancement by UP and DOWN state sequences facilitates recruitment of postsynaptic action potentials, which in turn results in long-term stabilization of synaptic strength. In contrast, synapses undergo lasting depression if presynaptic neurons fail to recruit postsynaptic action potentials. Our study offers a mechanistic explanation for how coupling of neural activity to slow waves can cause synaptic consolidation, with potential implications for brain stimulation strategies targeting memory performance.
Collapse
Affiliation(s)
- Franz X Mittermaier
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Berlin, Germany
| | - Thilo Kalbhenn
- Department of Neurosurgery (Evangelisches Klinikum Bethel), University of Bielefeld Medical Center OWL, Bielefeld, Germany
| | - Ran Xu
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julia Onken
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Katharina Faust
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Sauvigny
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich W Thomale
- Pediatric Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Angela M Kaindl
- Department of Pediatric Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Martin Holtkamp
- Department of Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sabine Grosser
- Institute for Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Pawel Fidzinski
- Neuroscience Clinical Research Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, Berlin, Germany
| | - Matthias Simon
- Department of Neurosurgery (Evangelisches Klinikum Bethel), University of Bielefeld Medical Center OWL, Bielefeld, Germany
| | - Henrik Alle
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Berlin, Germany
| | - Jörg R P Geiger
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Berlin, Germany.
| |
Collapse
|
34
|
Di Pierro F, Sisti D, Rocchi M, Belli A, Bertuccioli A, Cazzaniga M, Palazzi CM, Tanda ML, Zerbinati N. Effects of Melissa officinalis Phytosome on Sleep Quality: Results of a Prospective, Double-Blind, Placebo-Controlled, and Cross-Over Study. Nutrients 2024; 16:4199. [PMID: 39683592 DOI: 10.3390/nu16234199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Melissa officinalis standardised extracts, characterised by the presence of hydroxycinnamic acids, have been experimentally demonstrated to be endowed with anti-anxiety and anti-insomnia pharmacological actions. These effects, probably attributable, at least in part, to the role played by rosmarinic acid on GABA-T, have not always been observed in a reproducible manner in humans, perhaps due to the poor bioavailability of these compounds. METHODS as nutraceuticals and botanicals could be an alternative option to prescription medications for alleviating symptoms of mild anxiety and insomnia, we have verified in a prospective, double-blind, placebo-controlled, and cross-over study the supporting role on sleep quality played by a Melissa officinalis highly standardised extract, formulated as Phytosome™ (MOP) to improve the oral bioavailability of its active polyphenolic components. RESULTS results showed a significant reduction in the ISI score in the treated group, with an average of 6.8 ± 4.1 compared to 9.7 ± 3.7 in the placebo group, indicating a significant reduction of 2.9 points (p = 0.003). The SWS phase duration increased by an average of 15%, while the REM phase decreased by 10%. Additionally, 87% of participants in the treated group reported improved sleep quality, compared to 30% in the placebo group, with significant differences measured by chi-square test (χ2(4) = 21.01, p = 0.0003), highlighting the effects due to Melissa officinalis L. No significant changes in physical activity or anxiety levels were observed. CONCLUSIONS these findings suggest that MOP may represent a natural and safe alternative to traditional pharmacological treatments for insomnia.
Collapse
Affiliation(s)
- Francesco Di Pierro
- Department of Medicine and Technological Innovation, University of Insubria, 21100 Varese, Italy
- Microbiota International Clinical Society, 10123 Torino, Italy
- Scientific & Research Department, Velleja Research, 20125 Milano, Italy
| | - Davide Sisti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Marco Rocchi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Annalisa Belli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Alexander Bertuccioli
- Microbiota International Clinical Society, 10123 Torino, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Massimiliano Cazzaniga
- Microbiota International Clinical Society, 10123 Torino, Italy
- Scientific & Research Department, Velleja Research, 20125 Milano, Italy
| | | | - Maria Laura Tanda
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Nicola Zerbinati
- Department of Medicine and Technological Innovation, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
35
|
Cheng Y, Huang P, Lin L, Zhang J, Cheng Y, Zheng J, Wang Y, Pan X. Abnormal brain-heart electrophysiology in mild and severe orthostatic hypotension. J Hypertens 2024; 42:2094-2106. [PMID: 39207017 DOI: 10.1097/hjh.0000000000003838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION This study investigated the changes in cardiocerebral electrophysiology in patients with mild orthostatic hypotension (MOH) and severe orthostatic hypotension (SOH) and their relationship with the severity of orthostatic hypotension, psychiatric symptoms, and cognitive dysfunction. METHODS This study included 72 nonorthostatic hypotension (NOH), 17 with MOH, and 11 with SOH. Seated resting-state heart rate variability (HRV) and quantitative electroencephalogram parameters were synchronized and recorded. HRV measures in the time and frequency domains were analyzed, along with the peak frequency and power of the brain waves. RESULTS Abnormal neuronal activity was found in FP1 in patients with MOH, whereas it was more widespread in FP1, FP2, and O2 in patients with SOH ( P < 0.05). Cardiac and cerebral electrophysiological abnormalities were significantly associated with orthostatic hypotension severity, psychiatric symptoms, and cognitive dysfunction. CONCLUSION Abnormal EEG activity in patients are mainly manifested in the prefrontal and occipital lobes, especially in patients with SOH. These results may help patients to better understand the mechanisms underlying orthostatic hypotension severity and psychiatric and cognitive impairment in orthostatic hypotension.
Collapse
Affiliation(s)
- Yingzhe Cheng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
- Institute of Clinical Neurology
- Four Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou City
| | - Peilin Huang
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
- Institute of Clinical Neurology
- Four Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou City
| | - Lin Lin
- Department of Neurology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jiejun Zhang
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
- Institute of Clinical Neurology
- Four Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou City
- Center for Geriatrics, Hainan General Hospital, Hainan Province
| | - Yahui Cheng
- Shandong Second Medical University, Weifang City
| | - Jiahao Zheng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
- Institute of Clinical Neurology
- Four Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou City
| | - Yanping Wang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou
| | - Xiaodong Pan
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
- Institute of Clinical Neurology
- Four Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou City
| |
Collapse
|
36
|
Fan X, Mao X, Yu P, Han D, Chen C, Wang H, Zhang X, Liu S, Chen W, Chen Z, Du X, Jin L, Song Y, Li H, Zhang N, Wu Y, Chang L, Wang C. Sleep disturbance impaired memory consolidation via lateralized disruption of metabolite in the thalamus and hippocampus: A cross-sectional proton magnetic resonance spectroscopy study. J Alzheimers Dis 2024; 102:1057-1073. [PMID: 39584303 DOI: 10.1177/13872877241295401] [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] [Indexed: 11/26/2024]
Abstract
BACKGROUND Memory consolidation in sleep-dependent individuals involves the circuitry connections of cortex, thalamus and hippocampus, regulating via neural metabolites. However, the disruption of metabolic pattern in thalamus and hippocampus remains unclear. OBJECTIVE We aim to explore the disruptive effects of insomnia on the metabolites during memory consolidation, particularly the underlying neurometabolic mechanisms in comorbidity of failed memory consolidation. METHODS This study integrates clinical research with animal experiment. In clinical research, 49 participants were divided into four groups: healthy controls (HC, n = 11), insomnia with normal cognition (IS, n = 14), mild cognitive impairment without insomnia (MCI, n = 10), and insomnia with mild cognitive impairment (IS-MCI, n = 14). Magnetic resonance spectroscopy (MRS) was used to evaluate the neural γ-aminobutyric acid (GABA) and glutamate-glutamine (Glx) in bilateral thalamus. In experimental studies, the rat model of sleep deprivation combined with amyloid-β (Aβ) injection was established, after behavior testing, the levels of Glx, choline (Cho) and N-acetyl aspartate (NAA) in the bilateral hippocampus were evaluated with MRS. RESULTS The patients in the IS-MCI group exhibited significantly lower GABA level than IS, MCI and HC groups. Results from rat studies showed that sleep deprivation exacerbated asymmetric alterations in Aβ-induced bilateral hippocampal metabolite abnormalities, which correlated with cognition. These neuro-metabolite disruption accompanied with synaptic loss and activation of astrocytes. CONCLUSIONS The lateralized decrease in GABA levels of thalamus and NAA, Cho, and Glx levels of hippocampus under conditions of sleep disturbance with cognitive decline may provide evidence for the neural metabolic mechanisms underlying the disruption of memory consolidation.
Collapse
Affiliation(s)
- Xiaowei Fan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
| | - Xin Mao
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Ping Yu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
| | - Ding Han
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Chuxin Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
| | - Hongqi Wang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xinyi Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
| | - Siyu Liu
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Weijing Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
| | - Ziyan Chen
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xiaoqiang Du
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Liangyun Jin
- Electron Microscope Room of Central Laboratory, Capital Medical University, Beijing, China
| | - Yizhi Song
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Hui Li
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Ning Zhang
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
- Department of Neuropsychiatry and Behavioral Neurology and Clinical Psychology, Capital Medical University, Beijing, China
| | - Yan Wu
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Lirong Chang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Chunxue Wang
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
- Department of Neuropsychiatry and Behavioral Neurology and Clinical Psychology, Capital Medical University, Beijing, China
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
37
|
McHugh SB, Lopes-Dos-Santos V, Castelli M, Gava GP, Thompson SE, Tam SKE, Hartwich K, Perry B, Toth R, Denison T, Sharott A, Dupret D. Offline hippocampal reactivation during dentate spikes supports flexible memory. Neuron 2024; 112:3768-3781.e8. [PMID: 39321790 PMCID: PMC7616703 DOI: 10.1016/j.neuron.2024.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 06/21/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
Stabilizing new memories requires coordinated neuronal spiking activity during sleep. Hippocampal sharp-wave ripples (SWRs) in the cornu ammonis (CA) region and dentate spikes (DSs) in the dentate gyrus (DG) are prime candidate network events for supporting this offline process. SWRs have been studied extensively, but the contribution of DSs remains unclear. By combining triple-ensemble (DG-CA3-CA1) recordings and closed-loop optogenetics in mice, we show that, like SWRs, DSs synchronize spiking across DG and CA principal cells to reactivate population-level patterns of neuronal coactivity expressed during prior waking experience. Notably, the population coactivity structure in DSs is more diverse and higher dimensional than that seen during SWRs. Importantly, suppressing DG granule cell spiking selectively during DSs impairs subsequent flexible memory performance during multi-object recognition tasks and associated hippocampal patterns of neuronal coactivity. We conclude that DSs constitute a second offline network event central to hippocampal population dynamics serving memory-guided behavior.
Collapse
Affiliation(s)
- Stephen B McHugh
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK.
| | - Vítor Lopes-Dos-Santos
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Manfredi Castelli
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Giuseppe P Gava
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Sophie E Thompson
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Shu K E Tam
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Katja Hartwich
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Brook Perry
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Robert Toth
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Timothy Denison
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Andrew Sharott
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - David Dupret
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK.
| |
Collapse
|
38
|
Wang W, Wang Z, Cao J, Dong Y, Chen Y. Melatonin ameliorates chronic sleep deprivation against memory encoding vulnerability: Involvement of synapse regulation via the mitochondrial-dependent redox homeostasis-induced autophagy inhibition. Free Radic Biol Med 2024; 225:398-414. [PMID: 39396581 DOI: 10.1016/j.freeradbiomed.2024.10.279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/13/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Voluntary sleep curtailment is increasingly more rampant in modern society and compromises healthy cognition, including memory, to varying degrees. However, whether memory encoding is impaired after chronic sleep deprivation (CSD) and the underlying molecular mechanisms involved remain unclear. Here, using the mice, we tested the impact of CSD on the encoding abilities of social recognition-dependent memory and object recognition-dependent memory. We found that memory encoding was indeed vulnerable to CSD, while memory retrieval remained unaffected. The hippocampal neurons of mice with memory encoding deficits exhibited significant synapse damage and hyperactive autophagy, which dissipates during regular sleep cycles. This excessive autophagy appeared to be triggered by damage to mitochondrial DNA (mtDNA), resulting from oxidative stress within the mitochondria. The relief at the behavioral and molecular biological levels can be achieved with intraperitoneal injections of the antioxidant compound melatonin. Moreover, our in vitro experiments using HT-22 cells demonstrated that oxidative stress induced by hydrogen peroxide led to oxidative damage, including mtDNA damage, and activation of autophagy. Melatonin treatment effectively countered these effects, restoring redox homeostasis and reducing excessive autophagic activity. Notably, this protective effect was not observed when melatonin was administered as a pre-treatment. Together, our findings reveal the vulnerability of memory encoding during chronic sleep curtailment, which is caused by oxidative stress and consequent enhancement of autophagy, suggest a potential therapeutic strategy for addressing these effects following prolonged wakefulness through melatonin intervention, and reiterate the significance of adequate sleep for memory formation and retention.
Collapse
Affiliation(s)
- Wei Wang
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China
| | - Zixu Wang
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China
| | - Jing Cao
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China
| | - Yulan Dong
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China
| | - Yaoxing Chen
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
39
|
Zheng Q, Huang Y, Mu C, Hu X, Lai CSW. Selective Modulation of Fear Memory in Non-Rapid Eye Movement Sleep. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400662. [PMID: 39382074 PMCID: PMC11600212 DOI: 10.1002/advs.202400662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 08/20/2024] [Indexed: 10/10/2024]
Abstract
Sleep stabilizes memories for their consolidation, but how to modify specific fear memory during sleep remains unclear. Here, it is reported that using targeted memory reactivation (TMR) to reactivate prior fear learning experience in non-slow wave sleep (NS) inhibits fear memory consolidation, while TMR during slow wave sleep (SWS) enhances fear memory in mice. Replaying conditioned stimulus (CS) during sleep affects sleep spindle occurrence, leading to the reduction or enhancement of slow oscillation-spindle (SO-spindle) coupling in NS and SWS, respectively. Optogenetic inhibition of pyramidal neurons in the frontal association cortex (FrA) during TMR abolishes the behavioral effects of NS-TMR and SWS-TMR by modulating SO-spindle coupling. Notably, calcium imaging of the L2/3 pyramidal neurons in the FrA shows that CS during SWS selectively enhances the activity of neurons previously activated during fear conditioning (FC+ neurons), which significantly correlates with CS-elicited spindle power spectrum density. Intriguingly, these TMR-induced calcium activity changes of FC+ neurons further correlate with mice freezing behavior, suggesting their contributions to the consolidation of fear memories. The findings indicate that TMR can selectively weaken or strengthen fear memory, in correlation with modulating SO-spindle coupling and the reactivation of FC+ neurons during substages of non-rapid eye movement (NREM) sleep.
Collapse
Affiliation(s)
- Qiyu Zheng
- School of Biomedical SciencesLKS Faculty of MedicineThe University of Hong KongHong KongSARChina
- Advanced Biomedical Instrumentation CentreHong Kong Science ParkShatin, New TerritoriesHong KongChina
| | - Yuhua Huang
- School of Biomedical SciencesLKS Faculty of MedicineThe University of Hong KongHong KongSARChina
| | - Changrui Mu
- School of Biomedical SciencesLKS Faculty of MedicineThe University of Hong KongHong KongSARChina
| | - Xiaoqing Hu
- Department of PsychologyFaculty of Social SciencesThe University of Hong KongHong KongSARChina
- The State Key Laboratory of Brain and Cognitive SciencesThe University of Hong KongHong KongSARChina
| | - Cora Sau Wan Lai
- School of Biomedical SciencesLKS Faculty of MedicineThe University of Hong KongHong KongSARChina
- Advanced Biomedical Instrumentation CentreHong Kong Science ParkShatin, New TerritoriesHong KongChina
- The State Key Laboratory of Brain and Cognitive SciencesThe University of Hong KongHong KongSARChina
| |
Collapse
|
40
|
Kiss MG, Cohen O, McAlpine CS, Swirski FK. Influence of sleep on physiological systems in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1284-1300. [PMID: 39528718 PMCID: PMC11567060 DOI: 10.1038/s44161-024-00560-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Sleep is a fundamental requirement of life and is integral to health. Deviation from optimal sleep associates with numerous diseases including those of the cardiovascular system. Studies, spanning animal models to humans, show that insufficient, disrupted or inconsistent sleep contribute to poor cardiovascular health by disrupting body systems. Fundamental experiments have begun to uncover the molecular and cellular links between sleep and heart health while large-scale human studies have associated sleep with cardiovascular outcomes in diverse populations. Here, we review preclinical and clinical findings that demonstrate how sleep influences the autonomic nervous, metabolic and immune systems to affect atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Máté G Kiss
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Oren Cohen
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S McAlpine
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Filip K Swirski
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
41
|
Hou R, Liu Z, Jin Z, Huang D, Hu Y, Du W, Zhu D, Yang L, Weng Y, Yuan T, Lu B, Wang Y, Ping Y, Xiao X. Coordinated Interactions between the Hippocampus and Retrosplenial Cortex in Spatial Memory. RESEARCH (WASHINGTON, D.C.) 2024; 7:0521. [PMID: 39483173 PMCID: PMC11525046 DOI: 10.34133/research.0521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/05/2024] [Accepted: 10/12/2024] [Indexed: 11/03/2024]
Abstract
While a hippocampal-cortical dialogue is generally thought to mediate memory consolidation, which is crucial for engram function, how it works remains largely unknown. Here, we examined the interplay of neural signals from the retrosplenial cortex (RSC), a neocortical region, and from the hippocampus in memory consolidation by simultaneously recording sharp-wave ripples (SWRs) of dorsal hippocampal CA1 and neural signals of RSC in free-moving mice during the delayed spatial alternation task (DSAT) and subsequent sleep. Hippocampal-RSC coordination during SWRs was identified in nonrapid eye movement (NREM) sleep, reflecting neural reactivation of decision-making in the task, as shown by a peak reactivation strength within SWRs. Using modified generalized linear models (GLMs), we traced information flow through the RSC-CA1-RSC circuit around SWRs during sleep following DSAT. Our findings show that after spatial training, RSC excitatory neurons typically increase CA1 activity prior to hippocampal SWRs, potentially initiating hippocampal memory replay, while inhibitory neurons are activated by hippocampal outputs in post-SWRs. We further identified certain excitatory neurons in the RSC that encoded spatial information related to the DSAT. These neurons, classified as splitters and location-related cells, showed varied responses to hippocampal SWRs. Overall, our study highlights the complex dynamics between the RSC and hippocampal CA1 region during SWRs in NREM sleep, underscoring their critical interplay in spatial memory consolidation.
Collapse
Affiliation(s)
- Ruiqing Hou
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Ziyue Liu
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Zichen Jin
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Dongxue Huang
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Yue Hu
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Wenjie Du
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Danyi Zhu
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Leiting Yang
- School of Life Science,
Fudan University, Shanghai 200032, China
| | - Yuanfeng Weng
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Tifei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center,
Shanghai Jiaotong University School of Medicine, Shanghai 200030, China
| | - Bin Lu
- Department of Endocrinology and Metabolism, Huadong Hospital,
Fudan University, Shanghai 200040, China
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Yong Ping
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education),
Shanghai JiaoTong University, Shanghai 200240, China
| | - Xiao Xiao
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| |
Collapse
|
42
|
Yan L, Wu L, Wiggin TD, Su X, Yan W, Li H, Li L, Lu Z, Li Y, Meng Z, Guo F, Li F, Griffith LC, Liu C. Brief disruption of activity in a subset of dopaminergic neurons during consolidation impairs long-term memory by fragmenting sleep. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.23.563499. [PMID: 37961167 PMCID: PMC10634733 DOI: 10.1101/2023.10.23.563499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Sleep disturbances are associated with poor long-term memory (LTM) formation, yet the underlying cell types and neural circuits involved have not been fully decoded. Dopamine neurons (DANs) are involved in memory processing at multiple stages. Here, using both male and female flies, Drosophila melanogaster , we show that, during the first few hours of memory consolidation, disruption of basal activity of a small subset of protocerebral anterior medial DANs (PAM-DANs), by either brief activation or inhibition of the two dorsal posterior medial (DPM) neurons, impairs 24 h LTM. Interestingly, these brief changes in activity using female flies result in sleep loss and fragmentation, especially at night. Pharmacological rescue of sleep after manipulation restores LTM. A specific subset of PAM-DANs (PAM-α1) that synapse onto DPM neurons specify the microcircuit that links sleep and memory. PAM-DANs, including PAM-α1, form functional synapses onto DPM mainly via multiple dopamine receptor subtypes. This PAM-α1 to DPM microcircuit exhibits a synchronized, transient, post-training increase in activity during the critical memory consolidation window, suggesting an effect of this microcircuit on maintaining the sleep necessary for LTM consolidation. Our results provide a new cellular and circuit basis for the complex relationship between sleep and memory.
Collapse
|
43
|
Xiao Q, Lu M, Zhang X, Guan J, Li X, Wen R, Wang N, Qian L, Liao Y, Zhang Z, Liao X, Jiang C, Yue F, Ren S, Xia J, Hu J, Luo F, Hu Z, He C. Isolated theta waves originating from the midline thalamus trigger memory reactivation during NREM sleep in mice. Nat Commun 2024; 15:9231. [PMID: 39455583 PMCID: PMC11511994 DOI: 10.1038/s41467-024-53522-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
During non-rapid eye movement (NREM) sleep, neural ensembles in the entorhinal-hippocampal circuit responsible for encoding recent memories undergo reactivation to facilitate the process of memory consolidation. This reactivation is widely acknowledged as pivotal for the formation of stable memory and its impairment is closely associated with memory dysfunction. To date, the neural mechanisms driving the reactivation of neural ensembles during NREM sleep remain poorly understood. Here, we show that the neural ensembles in the medial entorhinal cortex (MEC) that encode spatial experiences exhibit reactivation during NREM sleep. Notably, this reactivation consistently coincides with isolated theta waves. In addition, we found that the nucleus reuniens (RE) in the midline thalamus exhibits typical theta waves during NREM sleep, which are highly synchronized with those occurring in the MEC in male mice. Closed-loop optogenetic inhibition of the RE-MEC pathway specifically suppressed these isolated theta waves, resulting in impaired reactivation and compromised memory consolidation following a spatial memory task in male mice. The findings suggest that theta waves originating from the ventral midline thalamus play a role in initiating memory reactivation and consolidation during sleep.
Collapse
Affiliation(s)
- Qin Xiao
- Department of Physiology, Third Military Medical University, Chongqing, China
| | - Minmin Lu
- Department of Physiology, Third Military Medical University, Chongqing, China
| | - Xiaolong Zhang
- Department of Physiology, Third Military Medical University, Chongqing, China
| | - Jiangheng Guan
- Department of Neurosurgery, General Hospital of Chinese PLA Central Theater Command, Wuhan, China
| | - Xin Li
- Department of Physiology, Third Military Medical University, Chongqing, China
| | - Ruyi Wen
- Department of Physiology, Third Military Medical University, Chongqing, China
| | - Na Wang
- Department of Physiology, Third Military Medical University, Chongqing, China
| | - Ling Qian
- Department of Physiology, Third Military Medical University, Chongqing, China
| | - Yixiang Liao
- Department of Physiology, Third Military Medical University, Chongqing, China
| | - Zehui Zhang
- Department of Physiology, College of Basic Medical Sciences of Jilin University, Changchun, China
| | - Xiang Liao
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China
| | - Chenggang Jiang
- Department of Sleep and Psychology, Chongqing Health Center for Women and Children, Chongqing, China
| | - Faguo Yue
- Sleep and Psychology Center, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Shuancheng Ren
- Department of Physiology, Third Military Medical University, Chongqing, China
| | - Jianxia Xia
- Department of Physiology, Third Military Medical University, Chongqing, China
| | - Jun Hu
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Fenlan Luo
- Department of Physiology, Third Military Medical University, Chongqing, China.
| | - Zhian Hu
- Department of Physiology, Third Military Medical University, Chongqing, China.
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China.
| | - Chao He
- Department of Physiology, Third Military Medical University, Chongqing, China.
| |
Collapse
|
44
|
Qi Z, Zeng W, Zang D, Wang Z, Luo L, Wu X, Yu J, Mao Y. Classifying disorders of consciousness using a novel dual-level and dual-modal graph learning model. J Transl Med 2024; 22:950. [PMID: 39434088 PMCID: PMC11492684 DOI: 10.1186/s12967-024-05729-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Disorders of consciousness (DoC) are a group of conditions that affect the level of awareness and communication in patients. While neuroimaging techniques can provide useful information about the brain structure and function in these patients, most existing methods rely on a single modality for analysis and rarely account for brain injury. To address these limitations, we propose a novel method that integrates two neuroimaging modalities, functional magnetic resonance imaging (fMRI) and diffusion tensor imaging (DTI), to enhance the classification of subjects into different states of consciousness. METHOD AND RESULTS The main contributions of our work are threefold: first, after constructing a dual-model individual graph using functional magnetic resonance imaging (fMRI) and diffusion tensor imaging (DTI), we introduce a brain injury mask mechanism that consolidates damaged brain regions into a single graph node, enhancing the modeling of brain injuries and reducing deformation effects. Second, to address over-smoothing, we construct a dual-level graph that dynamically construct a population-level graph with node features from individual graphs, to promote the clustering of similar subjects while distinguishing dissimilar ones. Finally, we employ a subgraph exploration model with task-fMRI data to validate the interpretability of our model, confirming that the selected brain regions are task-relevant in cognition. Our experimental results on data from 89 healthy participants and 204 patients with DoC from Huashan Hospital, Fudan University, demonstrate that our method achieves high accuracy in classifying patients into unresponsive wakefulness syndrome (UWS), minimally conscious state (MCS), or normal conscious state, outperforming current state-of-the-art methods. The explainability results of our method identified a subset of brain regions that are important for consciousness, such as the default mode network, the salience network, the dorsal attention network, and the visual network. Our method also revealed the relationship between brain networks and language processing in consciousness, and showed that language-related subgraphs can distinguish MCS from UWS patients. CONCLUSION We proposed a novel graph learning method for classifying DoC based on fMRI and DTI data, introducing a brain injury mask mechanism to effectively handle damaged brains. The classification results demonstrate the effectiveness of our method in distinguishing subjects across different states of consciousness, while the explainability results identify key brain regions relevant to this classification. Our study provides new evidence for the role of brain networks and language processing in consciousness, with potential implications for improving the diagnosis and prognosis of patients with DoC.
Collapse
Affiliation(s)
- Zengxin Qi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200030, China
- National Center for Neurological Disorders, Shanghai, 200030, China
- Shanghai Key laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200030, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, 200030, China
| | - Wenwen Zeng
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Di Zang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200030, China.
- National Center for Neurological Disorders, Shanghai, 200030, China.
- Shanghai Key laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200030, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, 200030, China.
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China.
| | - Zhe Wang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200030, China
- National Center for Neurological Disorders, Shanghai, 200030, China
- Shanghai Key laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200030, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, 200030, China
| | - Lanqin Luo
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200030, China
- National Center for Neurological Disorders, Shanghai, 200030, China
- Shanghai Key laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200030, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, 200030, China
| | - Xuehai Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200030, China.
- National Center for Neurological Disorders, Shanghai, 200030, China.
- Shanghai Key laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200030, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, 200030, China.
| | - Jinhua Yu
- School of Information Science and Technology, Fudan University, Shanghai, China.
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200030, China.
- National Center for Neurological Disorders, Shanghai, 200030, China.
- Shanghai Key laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200030, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, 200030, China.
| |
Collapse
|
45
|
Madrid-Valero JJ, Scott EM, Boughton CK, Allen JM, Ware J, Wilinska ME, Hartnell S, Thankamony A, Randell T, Ghatak A, Besser RE, Elleri D, Trevelyan N, Campbell FM, Hovorka R, Gregory AM. Closed-Loop Therapy and Sleep in Young People Newly Diagnosed With T1D and Their Parents. J Diabetes Sci Technol 2024:19322968241286816. [PMID: 39397750 PMCID: PMC11571613 DOI: 10.1177/19322968241286816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
BACKGROUND A diagnosis of type 1 diabetes in a young person can create vulnerability for sleep. Historically it has been rare for young people to be offered a closed-loop system soon after diagnosis meaning that studies examining sleep under these circumstances in comparison with standard treatment have not been possible. In this study, we examine sleep in young people (and their parents) who were provided with hybrid closed-loop therapy at diagnosis of type 1 diabetes versus those who receive standard treatment over a 2-year period. METHODS The sample comprised 97 participants (mean age = 12.0 years; SD = 1.7) from a multicenter, open-label, randomized, parallel trial, where young people were randomized to either hybrid closed-loop insulin delivery or standard care at diagnosis. Sleep was measured using actigraphy and the Pittsburgh Sleep Quality Index (PSQI) in the young people, and using the PSQI in parents. RESULTS Sleep in young people using hybrid closed-loop insulin delivery did not differ significantly compared with those receiving standard care (although there were nonsignificant trends for better sleep in the closed-loop group for 4 of the 5 sleep actigraphy measures and PSQI). Similarly, there were nonsignificant differences for sleep between the groups at 24 months (with mixed direction of effects). CONCLUSIONS This study assessed for the first time sleep in young people using a closed-loop system soon after diagnosis. Although sleep was not significantly different for young people using closed-loop insulin delivery as compared with those receiving standard care, the direction of effects of the nonsignificant results indicates a possible tendency for better sleep quality in the hybrid closed-loop insulin delivery group at the beginning of the treatment.
Collapse
Affiliation(s)
- Juan J. Madrid-Valero
- Department of Human Anatomy and Psychobiology, University of Murcia, Murcia, Spain
- Biomedical Research Institute of Murcia, IMIB-Arrixaca, Murcia, Spain
| | - Eleanor M. Scott
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Charlotte K. Boughton
- Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Wolfson Diabetes and Endocrine Clinic, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Janet M. Allen
- Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Julia Ware
- Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Malgorzata E. Wilinska
- Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Sara Hartnell
- Wolfson Diabetes and Endocrine Clinic, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ajay Thankamony
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Tabitha Randell
- Department of Paediatric Diabetes and Endocrinology, Nottingham Children’s Hospital, Nottingham, UK
| | - Atrayee Ghatak
- Department of Diabetes, Alder Hey Children’s NHS Foundation Trust, Liverpool, UK
| | - Rachel E.J. Besser
- Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK
| | - Daniela Elleri
- Department of Diabetes, Royal Hospital for Sick Children, Edinburgh, UK
| | - Nicola Trevelyan
- Paediatric Diabetes, Southampton Children’s Hospital, Southampton, UK
| | - Fiona M. Campbell
- Department of Paediatric Diabetes, Leeds Children’s Hospital, Leeds, UK
| | - Roman Hovorka
- Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Alice M. Gregory
- Department of Psychology, Royal Holloway, University of London, Egham, UK
| |
Collapse
|
46
|
Mu X, Qu L, Yin L, Wang L, Liu X, Liu D. Pichia pastoris secreted peptides crossing the blood-brain barrier and DSIP fusion peptide efficacy in PCPA-induced insomnia mouse models. Front Pharmacol 2024; 15:1439536. [PMID: 39444618 PMCID: PMC11498945 DOI: 10.3389/fphar.2024.1439536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
Background Pichia pastoris-secreted delta sleep inducing peptide and crossing the blood-brain barrier peptides (DSIP-CBBBP) fusion peptides holds significant promise for its potential sleep-enhancing and neurotransmitter balancing effects. This study investigates these properties using a p-chlorophenylalanine (PCPA) -induced insomnia model in mice, an approach akin to traditional methods evaluating sleep-promoting activities in fusion peptides. Aim of the study The research aims to elucidate the sleep-promoting mechanism of DSIP-CBBBP, exploring its impact on neurotransmitter levels and sleep regulation, and to analyze its composition and structure. Materials and methods Using a PCPA-induced insomnia mouse model, the study evaluates the sleep-promoting effects of DSIP-CBBBP. The peptide's influence on neurotransmitters such as 5-HT, glutamate, dopamine, and melatonin is assessed. The functions of DSIP-CBBBP are characterized using biochemical and animal insomnia-induced behavior tests and compared without CBBBP. Results DSIP-CBBBP demonstrates a capacity to modulate neurotransmitter levels, indicated by changes in 5-HT, glutamate, DA, and melatonin. DSIP-CBBBP shows a better restorative effect than DSIP on neurotransmitter imbalance and the potential to enhance sleep. Conclusion The study underscores DSIP-CBBBP potential in correcting neurotransmitter dysregulation and promoting sleep, hinting at its utility in sleep-related therapies.
Collapse
Affiliation(s)
- Xiaoxiao Mu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Lijun Qu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Liquan Yin
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Libo Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiaoyang Liu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Dingxi Liu
- Department of Clinical medicine, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| |
Collapse
|
47
|
Hahn MA, Lendner JD, Anwander M, Slama KSJ, Knight RT, Lin JJ, Helfrich RF. A tradeoff between efficiency and robustness in the hippocampal-neocortical memory network during human and rodent sleep. Prog Neurobiol 2024; 242:102672. [PMID: 39369838 DOI: 10.1016/j.pneurobio.2024.102672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/30/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Sleep constitutes a brain state of disengagement from the external world that supports memory consolidation and restores cognitive resources. The precise mechanisms how sleep and its varied stages support information processing remain largely unknown. Synaptic scaling models imply that daytime learning accumulates neural information, which is then consolidated and downregulated during sleep. Currently, there is a lack of in-vivo data from humans and rodents that elucidate if, and how, sleep renormalizes information processing capacities. From an information-theoretical perspective, a consolidation process should entail a reduction in neural pattern variability over the course of a night. Here, in a cross-species intracranial study, we identify a tradeoff in the neural population code during sleep where information coding efficiency is higher in the neocortex than in hippocampal archicortex in humans than in rodents as well as during wakefulness compared to sleep. Critically, non-REM sleep selectively reduces information coding efficiency through pattern repetition in the neocortex in both species, indicating a transition to a more robust information coding regime. Conversely, the coding regime in the hippocampus remained consistent from wakefulness to non-REM sleep. These findings suggest that new information could be imprinted to the long-term mnemonic storage in the neocortex through pattern repetition during sleep. Lastly, our results show that task engagement increased coding efficiency, while medically-induced unconsciousness disrupted the population code. In sum, these findings suggest that neural pattern variability could constitute a fundamental principle underlying cognitive engagement and memory formation, while pattern repetition reflects robust coding, possibly underlying the consolidation process.
Collapse
Affiliation(s)
- Michael A Hahn
- Hertie-Institute for Clinical Brain Research, University Medical Center Tübingen, Otfried-Müller Str. 27, Tübingen 72076, Germany.
| | - Janna D Lendner
- Hertie-Institute for Clinical Brain Research, University Medical Center Tübingen, Otfried-Müller Str. 27, Tübingen 72076, Germany; Department of Anesthesiology and Intensive Care Medicine, University Medical Center Tübingen, Hoppe-Seyler-Str 3, Tübingen 72076, Germany
| | - Matthias Anwander
- Hertie-Institute for Clinical Brain Research, University Medical Center Tübingen, Otfried-Müller Str. 27, Tübingen 72076, Germany
| | - Katarina S J Slama
- Department of Psychology and the Helen Wills Neuroscience Institute, UC Berkeley, 130 Barker Hall, Berkeley, CA 94720, USA
| | - Robert T Knight
- Department of Psychology and the Helen Wills Neuroscience Institute, UC Berkeley, 130 Barker Hall, Berkeley, CA 94720, USA
| | - Jack J Lin
- Department of Neurology, UC Davis, 3160 Folsom Blvd, Sacramento, CA 95816, USA; Center for Mind and Brain, UC Davis, 267 Cousteau Pl, Davis, CA 95618, USA
| | - Randolph F Helfrich
- Hertie-Institute for Clinical Brain Research, University Medical Center Tübingen, Otfried-Müller Str. 27, Tübingen 72076, Germany.
| |
Collapse
|
48
|
Wan Y, Gao H, Zhou K, Zhang X, Xue R, Zhang N. Virtual reality improves sleep quality and associated symptoms in patients with chronic insomnia. Sleep Med 2024; 122:230-236. [PMID: 39213857 DOI: 10.1016/j.sleep.2024.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/16/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE The present study aimed to explore the effectiveness of virtual reality (VR) therapy on sleep quality and associated symptoms, such as depression and anxiety, cognitive decline and autonomic nervous dysfunction, in chronic insomnia patients. METHODS Sixty-three chronic insomnia patients were randomly divided into VR group (n = 32) and control group (n = 20) based on a standard drug therapy. Patients were instructed to use VR at home once a day at evening for 6-week treatment. All participants received evaluations of subjective sleep quality measured with the Pittsburgh Sleep Quality Index (PSQI), the Insomnia Severity Index (ISI), and the Epworth Sleepiness Scale (ESS), depression and anxiety symptoms measured with the Hamilton Depression Scale (HAMD) and Hamilton Anxiety Scale (HAMA), cognitive function, and objective sleep structure and autonomic nerve function examination measured with the sleep respiration monitoring device at baseline and after 6-week treatment. The main objective of this study was sleep quality assessment as the primary outcome. RESULTS After 6-week treatment, the decreases in PSQI score (-5.60 ± 2.37 vs -4.10 ± 1.80, P = 0.020) and ISI score (-8.81 ± 4.52 vs -6.35 ± 2.89, P = 0.038) of the VR group were significantly greater compared with the control group. The VR group showed more reduction in HAMD score (-9.96 ± 4.41 vs -7.50 ± 2.89, P = 0.035) and HAMA score (-8.96 ± 3.80 vs -6.80 ± 3.22, P = 0.046), and more increase in processing speed (0.54 ± 0.60 vs 0.00 ± 0.79, P = 0.011) than the control group. Moreover, the low-frequency coupling (-10.00 ± 17.40 vs. 8.25 ± 20.03, P = 0.001) was lowered and the high-frequency coupling (9.99 ± 17.40 vs. -8.24 ± 20.03, P = 0.001) was elevated in the VR group relative to the control group. CONCLUSION Our findings offered preliminary evidence that VR therapy enhanced sleep quality and also lessened depressive and anxious symptoms, and improved cognitive and autonomic functioning in patients with chronic insomnia.
Collapse
Affiliation(s)
- Yahui Wan
- Departments of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, 300308, China
| | - Haijing Gao
- Departments of Neurology, The First Affiliated Hospital of Hebei North University, Hebei, 075000, China
| | - Kaili Zhou
- Departments of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, 300308, China
| | - Xuan Zhang
- Departments of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, 300308, China
| | - Rong Xue
- Departments of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, 300308, China; Departments of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Nan Zhang
- Departments of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
49
|
Li Z, Wang J, Tang C, Wang P, Ren P, Li S, Yi L, Liu Q, Sun L, Li K, Ding W, Bao H, Yao L, Na M, Luan G, Liang X. Coordinated NREM sleep oscillations among hippocampal subfields modulate synaptic plasticity in humans. Commun Biol 2024; 7:1236. [PMID: 39354050 PMCID: PMC11445409 DOI: 10.1038/s42003-024-06941-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
The integration of hippocampal oscillations during non-rapid eye movement (NREM) sleep is crucial for memory consolidation. However, how cardinal sleep oscillations bind across various subfields of the human hippocampus to promote information transfer and synaptic plasticity remains unclear. Using human intracranial recordings from 25 epilepsy patients, we find that hippocampal subfields, including DG/CA3, CA1, and SUB, all exhibit significant delta and spindle power during NREM sleep. The DG/CA3 displays strong coupling between delta and ripple oscillations with all the other hippocampal subfields. In contrast, the regions of CA1 and SUB exhibit more precise coordination, characterized by event-level triple coupling between delta, spindle, and ripple oscillations. Furthermore, we demonstrate that the synaptic plasticity within the hippocampal circuit, as indexed by delta-wave slope, is linearly modulated by spindle power. In contrast, ripples act as a binary switch that triggers a sudden increase in delta-wave slope. Overall, these results suggest that different subfields of the hippocampus regulate one another through diverse layers of sleep oscillation synchronization, collectively facilitating information processing and synaptic plasticity during NREM sleep.
Collapse
Affiliation(s)
- Zhipeng Li
- School of Life Science and Technology, HIT Faculty of Life Science and Medicine, Harbin Institute of Technology, Harbin, 150001, China
- Laboratory for Space Environment and Physical Sciences, Harbin Institute of Technology, Harbin, 150001, China
| | - Jing Wang
- Department of Neurology, SanBo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Chongyang Tang
- Department of Neurosurgery, SanBo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Peng Wang
- Institute of Psychology, University of Greifswald, Greifswald, Germany
| | - Peng Ren
- Institute of Science and Technology for Brain-Inspired Intelligence and Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Siyang Li
- Zhejiang Lab, Hangzhou, Zhejiang, 311100, China
| | - Liye Yi
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiuyi Liu
- School of Life Science and Technology, HIT Faculty of Life Science and Medicine, Harbin Institute of Technology, Harbin, 150001, China
- Laboratory for Space Environment and Physical Sciences, Harbin Institute of Technology, Harbin, 150001, China
| | - Lili Sun
- School of Life Science and Technology, HIT Faculty of Life Science and Medicine, Harbin Institute of Technology, Harbin, 150001, China
- Laboratory for Space Environment and Physical Sciences, Harbin Institute of Technology, Harbin, 150001, China
| | - Kaizhou Li
- School of Life Science and Technology, HIT Faculty of Life Science and Medicine, Harbin Institute of Technology, Harbin, 150001, China
- Laboratory for Space Environment and Physical Sciences, Harbin Institute of Technology, Harbin, 150001, China
| | - Wencai Ding
- Department of Neurology, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Hongbo Bao
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, 150081, Harbin, China
- Department of Neurosurgery, BeijingTiantan Hospital, Capital Medical University, 100070, Beijing, China
| | - Lifen Yao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Meng Na
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| | - Guoming Luan
- Department of Neurosurgery, SanBo Brain Hospital, Capital Medical University, Beijing, 100093, China.
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, 100093, China.
| | - Xia Liang
- School of Life Science and Technology, HIT Faculty of Life Science and Medicine, Harbin Institute of Technology, Harbin, 150001, China.
- Laboratory for Space Environment and Physical Sciences, Harbin Institute of Technology, Harbin, 150001, China.
- Frontiers Science Center for Matter Behave in Space Environment, Harbin Institute of Technology, Harbin, 150001, China.
- Research Center for Social Computing and Information Retrieval, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
50
|
Mir FA, Jha SK. The Kir channel in the nucleus tractus solitarius integrates the chemosensory system with REM sleep executive machinery for homeostatic balance. Sci Rep 2024; 14:21651. [PMID: 39289431 PMCID: PMC11408532 DOI: 10.1038/s41598-024-71818-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/30/2024] [Indexed: 09/19/2024] Open
Abstract
The locus coeruleus (LC), nucleus tractus solitarius (NTS), and retrotrapezoid nucleus (RTN) are critical chemosensory regions in the brainstem. In the LC, acid-sensing ion channels and proton pumps serve as H+ sensors and facilitate the transition from non-rapid eye movement (NREM) to rapid eye movement (REM) sleep. Interestingly, the potassium inward rectifier (KIR) channels in the LC, NTS, and RTN also act as H+-sensors and are a primary target for improving sleep in obstructive sleep apnea and Rett syndrome patients. However, the role of Kir channels in NREM to REM sleep transition for H+ homeostasis is not known. Male Wistar rats were surgically prepared for chronic sleep-wake recording and drug delivery into the LC, NTS, and RTN. In different animal cohorts, microinjections of the Kir channel inhibitor, barium chloride (BaCl2), at concentrations of 1 mM (low dose) and 2 mM (high dose) in the LC and RTN significantly increased wakefulness and decreased NREM sleep. However, BaCl2 microinjection into the LC notably reduced REM sleep, whereas it didn't change in the RTN-injected group. Interestingly, BaCl2 microinjections into the NTS significantly decreased wakefulness and increased the percent amount of NREM and REM sleep. Additionally, with the infusion of BaCl2 into the NTS, the mean REM sleep episode numbers significantly increased, but the length of the REM sleep episode didn't change. These findings suggest that the Kir channels in the NTS, but not in the LC and RTN, modulate state transition from NREM to REM sleep.
Collapse
Affiliation(s)
- Fayaz A Mir
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
- Department of Anesthesia, Critical Care & Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Sushil K Jha
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|