1
|
Sarma A, Ronde M, Smit S, Meerlo P, Havekes R. Does It Matter What Keeps You Awake? Effects of Two Different Sleep Deprivation Methods on Object-Location Memory and Hippocampal c-Fos Expression in Mice. J Sleep Res 2025:e70079. [PMID: 40267993 DOI: 10.1111/jsr.70079] [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: 10/24/2024] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
In sleep research, various sleep deprivation methods have been used to examine the effects of sleep loss on memory. However, studies often overlook the distinct impacts each method may have on activity in specific neuronal circuits and memory storage. It remains unclear whether these changes following sleep deprivation result from extended wakefulness alone or from an interaction with the nature of the waking experience. To address this question, we examined how two commonly used sleep deprivation methods in mice-gentle handling and novelty-induced sleep deprivation-affect object-location memory and hippocampal c-Fos expression. Using either method, mice were sleep deprived for 3 or 6 h immediately after training in the object-location memory task, and spatial memory performance was assessed 1 day after training. Object-location memory was impaired after 3 and 6 h of novelty-induced sleep deprivation, but only after 6 h of sleep deprivation by gentle handling. Assessing c-Fos expression in separate groups of mice immediately after 3 or 6 h of sleep deprivation showed that both methods increased c-Fos expression in the CA1 and CA3 regions after 3 h of sleep deprivation, while effects in the dentate gyrus depended on the method and blade examined. After 6 h of sleep deprivation, no significant changes in hippocampal c-Fos expression were observed regardless of the method used. Overall, our findings show that the type of experience mice have while being kept awake and the duration of sleep deprivation can have different effects on spatial memory and neuronal activity in hippocampal subregions.
Collapse
Affiliation(s)
- Adithya Sarma
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Mirthe Ronde
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Soraya Smit
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Peter Meerlo
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Robbert Havekes
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| |
Collapse
|
2
|
Zeng S, Liu N, Zhang A, Duan N, Xu B, Ai C. Molecular basis identification and hypnotic drug interactions for cognitive impairment related to sleep deprivation. BMC Psychiatry 2025; 25:371. [PMID: 40229714 PMCID: PMC11995581 DOI: 10.1186/s12888-024-06395-7] [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: 05/28/2024] [Accepted: 12/09/2024] [Indexed: 04/16/2025] Open
Abstract
Chronic sleep deprivation can lead to cognitive impairment which makes it difficult to think, focus, and make comprehensive decisions. This in turn leads to the progression and increased risk of several diseases. This study aimed to explore potential drug targets and biomarkers underlying the increased disease risk due to sleep deprivation, including stress responses, immune dysfunction, and metabolic dysregulation. Four datasets namely GSE40562, GSE98566, GSE98582 for sleep deprivation, and GSE26576 normal brain cells were utilized to understand the molecular basis and potential drug targets associated with sleep deprivation. The GEO2R tool, Robust rank aggregations, and Venny were used to retrieve the common DEGs. Functional gene and pathway analyses were carried out via GO and the KEGG analyses. The STRING and CytoHuba plugins were utilized to identify the protein-protein interactions (PPIs) as well as the hub genes in the main PPI subnetworks following the drug interaction of the hub genes and GEPIA-based survival analysis of the DEGs. A total of 160 common DEGs were retrieved from all four datasets. Among them, 65 were down-regulated and 95 were up-regulated. TOP2A, AURKB, NEFL, CDC42, ASPM, GAP43, PVALB, NUF2, CALM1, TPR, KIF5B, KIF15, TROAP, NDC80, PBK, MKI67, SST, AHSP, ALAS2, and NEFH were retrieved as hub genes. While based on drug interaction, survival analysis and gene expression profile eight hub gene named TOP2A, AURKB, PVALB, CALM1, KIF5B, PBK, MKI67, and SST were found to be potential drug candidates and significantly correlated with infiltration levels of CD8 + T cells, B cells, macrophages, CD4 + T cells, neutrophils, and dendritic cells. These genes might play a role in sleep disorders via various pathways associated with neurodegeneration and diseases, potentially serving as biomarkers to support treatment and diagnosis.
Collapse
Affiliation(s)
- Shun Zeng
- Department of Mental Health Center, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Nannan Liu
- Department of Sleep Disorders Center, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Andong Zhang
- College of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Na Duan
- Obstetrical Department, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Bo Xu
- College of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Chunqi Ai
- Department of Mental Health Center, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China.
- College of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China.
| |
Collapse
|
3
|
Feeney SP, McCarthy JM, Petruconis CR, Tudor JC. Sleep loss is a metabolic disorder. Sci Signal 2025; 18:eadp9358. [PMID: 40198749 DOI: 10.1126/scisignal.adp9358] [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: 07/27/2024] [Accepted: 03/12/2025] [Indexed: 04/10/2025]
Abstract
Sleep loss dysregulates cellular metabolism and energy homeostasis. Highly metabolically active cells, such as neurons, enter a catabolic state during periods of sleep loss, which consequently disrupts physiological functioning. Specific to the central nervous system, sleep loss results in impaired synaptogenesis and long-term memory, effects that are also characteristic of neurodegenerative diseases. In this review, we describe how sleep deprivation increases resting energy expenditure, leading to the development of a negative energy balance-a state with insufficient metabolic resources to support energy expenditure-in highly active cells like neurons. This disruption of energetic homeostasis alters the balance of metabolites, including adenosine, lactate, and lipid peroxides, such that energetically costly processes, such as synapse formation, are attenuated. During sleep loss, metabolically active cells shunt energetic resources away from those processes that are not acutely essential, like memory formation, to support cell survival. Ultimately, these findings characterize sleep loss as a metabolic disorder.
Collapse
Affiliation(s)
- Sierra P Feeney
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Jordan M McCarthy
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Cecilia R Petruconis
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Jennifer C Tudor
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA 19131, USA
| |
Collapse
|
4
|
Tennin M, Matkins HT, Rexrode L, Bollavarapu R, Asplund SD, Pareek T, Kroeger D, Pantazopoulos H, Gisabella B. Sleep Deprivation Alters Hippocampal Dendritic Spines in a Contextual Fear Memory Engram. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.02.641043. [PMID: 40093122 PMCID: PMC11908145 DOI: 10.1101/2025.03.02.641043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Sleep is critically involved in strengthening memories. However, our understanding of the morphological changes underlying this process is still emerging. Recent studies suggest that specific subsets of dendritic spines are strengthened during sleep in specific neurons involved in recent learning. Contextual memories associated with traumatic experiences are involved in post-traumatic stress disorder (PTSD) and represent recent learning that may be strengthened during sleep. We tested the hypothesis that dendritic spines encoding contextual fear memories are selectively strengthened during sleep. Furthermore, we tested how sleep deprivation after initial fear learning impacts dendritic spines following re-exposure to fear conditioning. We used ArcCreERT2 mice to visualize neurons that encode contextual fear learning (Arc+ neurons), and concomitantly labeled neurons that did not encode contextual fear learning (Arc- neurons). Dendritic branches of Arc+ and Arc- neurons were sampled using confocal imaging to assess spine densities using three-dimensional image analysis from either sleep deprived (SD) or control mice allowed to sleep normally. Mushroom spines in Arc+ branches displayed decreased density in SD mice, indicating upscaling of mushroom spines during sleep following fear learning. In comparison, no changes were observed in dendritic spines from Arc- branches. When animals were re-exposed to contextual fear conditioning 4 weeks later, we observed lower density of mushroom spines in both Arc+ and Arc- branches, as well as lower density of thin spines in Arc- branches in mice that were SD following the initial fear conditioning trial. Our findings indicate that sleep strengthens dendritic spines in neurons that recently encoded fear memory, and sleep deprivation following initial fear learning impairs dendritic spine strengthening initially and following later re-exposure. SD following a traumatic experience thus may be a viable strategy in weakening the strength of contextual memories associated with trauma and PTSD.
Collapse
Affiliation(s)
- Matthew Tennin
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Hunter T. Matkins
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Lindsay Rexrode
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Ratna Bollavarapu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Samuel D. Asplund
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Tanya Pareek
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Daniel Kroeger
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
5
|
Raven F, Medina AV, Schmidt K, He A, Vankampen AA, Balendran V, Aton SJ. Brief sleep disruption alters synaptic structures among hippocampal and neocortical somatostatin-expressing interneurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.22.604591. [PMID: 39211205 PMCID: PMC11360998 DOI: 10.1101/2024.07.22.604591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Study objectives Brief sleep loss alters cognition and synaptic structures of principal neurons in hippocampus and neocortex. However, while in vivo recording and bioinformatic data suggest that inhibitory interneurons are more strongly affected by sleep loss, it is unclear how sleep and sleep deprivation affect interneurons' synapses. Disruption of the SST+ interneuron population seems to be a critical early sign of neuropathology in Alzheimer's dementia, schizophrenia, and bipolar disorder - and the risk of developing all three is increased by habitual sleep loss. We aimed to test how the synaptic structures of SST+ interneurons in various brain regions are affected by brief sleep disruption. Methods We used Brainbow 3.0 to label SST+ interneurons in the dorsal hippocampus, prefrontal cortex, and visual cortex of male SST-CRE transgenic mice, then compared synaptic structures in labeled neurons after a 6-h period of ad lib sleep, or gentle handling sleep deprivation (SD) starting at lights on. Results Dendritic spine density among SST+ interneurons in both hippocampus and neocortex was altered in a subregion-specific manner, with increased overall and thin spine density in CA1, dramatic increases in spine volume and surface area in CA3, and small but significant changes (primarily decreases) in spine size in CA1, PFC and V1. Conclusions Our suggest that the synaptic connectivity of SST+ interneurons is significantly altered in a brain region-specific manner by a few hours of sleep loss. This suggests a cell type-specific mechanism by which sleep loss disrupts cognition and alters excitatory-inhibitory balance in brain networks.
Collapse
Affiliation(s)
- Frank Raven
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Alexis Vega Medina
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Kailynn Schmidt
- University of New England College of Osteopathic Medicine, Biddeford, ME 04005
| | - Annie He
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Anna A. Vankampen
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Vinodh Balendran
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| | - Sara J. Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019
| |
Collapse
|
6
|
Bridi MCD, Peixoto L. Excitatory/Inhibitory imbalance as a mechanism linking autism and sleep problems. Curr Opin Neurobiol 2025; 90:102968. [PMID: 39754885 PMCID: PMC11839321 DOI: 10.1016/j.conb.2024.102968] [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: 08/25/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/06/2025]
Abstract
Sleep problems occur more frequently in individuals with autism spectrum disorder (ASD) than in typically developing individuals, and recent studies support a genetic link between ASD and sleep disturbances. However, it remains unclear how sleep problems may be mechanistically connected to ASD phenotypes. A longstanding hypothesis posits that an imbalance between excitatory and inhibitory (E/I) signaling in the brain underlies the behavioral characteristics of ASD. In recent years, emerging evidence has shown that regulation of the E/I ratio is coupled to sleep/wake states in wild-type animal models. In this review, we will explore the idea of altered E/I regulation over the sleep/wake cycle as a mechanism bridging sleep disruption and behavioral phenotypes in ASD.
Collapse
Affiliation(s)
- Michelle C D Bridi
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA
| | - Lucia Peixoto
- Department of Translational Medicine and Physiology, Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University Spokane, 99202, USA.
| |
Collapse
|
7
|
Chen J, Zhu Z, Xu F, Dou B, Sheng Z, Xu Y. Phosphodiesterase 4 Inhibition in Neuropsychiatric Disorders Associated with Alzheimer's Disease. Cells 2025; 14:164. [PMID: 39936956 PMCID: PMC11816594 DOI: 10.3390/cells14030164] [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: 11/10/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/13/2025] Open
Abstract
Cognitive disorders and psychiatric pathologies, particularly Alzheimer's disease (AD) and Major depressive disorder (MDD), represent a considerable health burden, impacting millions of people in the United States and worldwide. Notably, comorbidities of MDD and anxiety are prevalent in the early stages of mild cognitive impairment (MCI), which is the preceding phase of Alzheimer's disease and related dementia (ADRD). The symptoms of MDD and anxiety affect up to 80% of individuals in the advanced stages of the neurodegenerative conditions. Despite overlapping clinical manifestations, the pathogenesis of AD/ADRD and MDD remains inadequately elucidated. Until now, dozens of drugs for treating AD/ADRD have failed in clinical trials because they have not proven beneficial in reversing or preventing the progression of these neuropsychiatric indications. This underscores the need to identify new drug targets that could reverse neuropsychiatric symptoms and delay the progress of AD/ADRD. In this context, phosphodiesterase 4 (PDE4) arises as a primary enzyme in the modulation of cognition and mood disorders, particularly through its enzymatic action on cyclic adenosine monophosphate (cAMP) and its downstream anti-inflammatory pathways. Despite the considerable cognitive and antidepressant potential of PDE4 inhibitors, their translation into clinical practice is hampered by profound side effects. Recent studies have focused on the effects of PDE4 and its subtype-selective isoform inhibitors, aiming to delineate their precise mechanistic contributions to neuropsychiatric symptoms with greater specificity. This review aims to analyze the current advances regarding PDE4 inhibition-specifically the selective targeting of its isoforms and elucidate the therapeutic implications of enhanced cAMP signaling and the consequent anti-inflammatory responses in ameliorating the symptomatology associated with AD and ADRD.
Collapse
Affiliation(s)
- Jiming Chen
- Department of Anesthesiology, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; (J.C.); (F.X.); (B.D.); (Z.S.)
| | - Zhengyao Zhu
- School of Nursing and Rehabilitation, Nantong University, Nantong 226007, China;
| | - Fu Xu
- Department of Anesthesiology, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; (J.C.); (F.X.); (B.D.); (Z.S.)
| | - Baomin Dou
- Department of Anesthesiology, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; (J.C.); (F.X.); (B.D.); (Z.S.)
| | - Zhutao Sheng
- Department of Anesthesiology, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; (J.C.); (F.X.); (B.D.); (Z.S.)
| | - Ying Xu
- Department of Anesthesiology, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; (J.C.); (F.X.); (B.D.); (Z.S.)
| |
Collapse
|
8
|
Kelly MP, Nikolaev VO, Gobejishvili L, Lugnier C, Hesslinger C, Nickolaus P, Kass DA, Pereira de Vasconcelos W, Fischmeister R, Brocke S, Epstein PM, Piazza GA, Keeton AB, Zhou G, Abdel-Halim M, Abadi AH, Baillie GS, Giembycz MA, Bolger G, Snyder G, Tasken K, Saidu NEB, Schmidt M, Zaccolo M, Schermuly RT, Ke H, Cote RH, Mohammadi Jouabadi S, Roks AJM. Cyclic nucleotide phosphodiesterases as drug targets. Pharmacol Rev 2025; 77:100042. [PMID: 40081105 DOI: 10.1016/j.pharmr.2025.100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/13/2025] [Indexed: 03/15/2025] Open
Abstract
Cyclic nucleotides are synthesized by adenylyl and/or guanylyl cyclase, and downstream of this synthesis, the cyclic nucleotide phosphodiesterase families (PDEs) specifically hydrolyze cyclic nucleotides. PDEs control cyclic adenosine-3',5'monophosphate (cAMP) and cyclic guanosine-3',5'-monophosphate (cGMP) intracellular levels by mediating their quick return to the basal steady state levels. This often takes place in subcellular nanodomains. Thus, PDEs govern short-term protein phosphorylation, long-term protein expression, and even epigenetic mechanisms by modulating cyclic nucleotide levels. Consequently, their involvement in both health and disease is extensively investigated. PDE inhibition has emerged as a promising clinical intervention method, with ongoing developments aiming to enhance its efficacy and applicability. In this comprehensive review, we extensively look into the intricate landscape of PDEs biochemistry, exploring their diverse roles in various tissues. Furthermore, we outline the underlying mechanisms of PDEs in different pathophysiological conditions. Additionally, we review the application of PDE inhibition in related diseases, shedding light on current advancements and future prospects for clinical intervention. SIGNIFICANCE STATEMENT: Regulating PDEs is a critical checkpoint for numerous (patho)physiological conditions. However, despite the development of several PDE inhibitors aimed at controlling overactivated PDEs, their applicability in clinical settings poses challenges. In this context, our focus is on pharmacodynamics and the structure activity of PDEs, aiming to illustrate how selectivity and efficacy can be optimized. Additionally, this review points to current preclinical and clinical evidence that depicts various optimization efforts and indications.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Neurobiology, Center for Research on Aging, University of Maryland School of Medicine, Baltimore, Maryland
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leila Gobejishvili
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, Louisville
| | - Claire Lugnier
- Translational CardioVascular Medicine, CRBS, UR 3074, Strasbourg, France
| | | | - Peter Nickolaus
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Rodolphe Fischmeister
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Orsay, France
| | - Stefan Brocke
- Department of Immunology, UConn Health, Farmington, Connecticut
| | - Paul M Epstein
- Department of Cell Biology, UConn Health, Farmington, Connecticut
| | - Gary A Piazza
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Adam B Keeton
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Gang Zhou
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - George S Baillie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mark A Giembycz
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Gretchen Snyder
- Molecular Neuropharmacology, Intra-Cellular Therapies Inc (ITI), New York, New York
| | - Kjetil Tasken
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nathaniel E B Saidu
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics and National Institute for Health and Care Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Ralph T Schermuly
- Department of internal Medicine, Justus Liebig University of Giessen, Giessen, Germany
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina
| | - Rick H Cote
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, New Hampshire
| | - Soroush Mohammadi Jouabadi
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Anton J M Roks
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
9
|
Lim CR, Ogawa S, Kumari Y. Exploring β-caryophyllene: a non-psychotropic cannabinoid's potential in mitigating cognitive impairment induced by sleep deprivation. Arch Pharm Res 2025; 48:1-42. [PMID: 39653971 DOI: 10.1007/s12272-024-01523-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Sleep deprivation or sleep loss, a prevalent issue in modern society, is linked to cognitive impairment, leading to heightened risks of errors and accidents. Chronic sleep deprivation affects various cognitive functions, including memory, attention, and decision-making, and is associated with an increased risk of neurodegenerative diseases, cardiovascular issues, and metabolic disorders. This review examines the potential of β-caryophyllene, a dietary non-psychotropic cannabinoid, and FDA-approved flavoring agent, as a therapeutic solution for sleep loss-induced cognitive impairment. It highlights β-caryophyllene's ability to mitigate key contributors to sleep loss-induced cognitive impairment, such as inflammation, oxidative stress, neuronal death, and reduced neuroplasticity, by modulating various signaling pathways, including TLR4/NF-κB/NLRP3, MAPK, Nrf2/HO-1, PI3K/Akt, and cAMP/PKA/CREB. As a naturally occurring, non-psychotropic compound with low toxicity, β-caryophyllene emerges as a promising candidate for further investigation. The review underscores the therapeutic potential of β-caryophyllene for sleep loss-induced cognitive impairment and provides mechanistic insights into its action on crucial pathways, suggesting that β-caryophyllene could be a valuable addition to strategies aimed at combating cognitive impairment and other health issues due to sleep loss.
Collapse
Affiliation(s)
- Cher Ryn Lim
- Neurological Disorder and Aging Research Group (NDA), Neuroscience Research Strength (NRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Selangor, Malaysia
| | - Satoshi Ogawa
- Neuroscience Research Strength (NRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Selangor, Malaysia
| | - Yatinesh Kumari
- Neurological Disorder and Aging Research Group (NDA), Neuroscience Research Strength (NRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Selangor, Malaysia.
| |
Collapse
|
10
|
Özcan ÖÖ, Çevreli B, Kaşıkçı ES, Karahan M, Konuk M. Effects of Quetiapine on Novelty-Related Object Recognition Memory and Hippocampal BDNF Level in Sleep-Deprived Rats. Brain Behav 2025; 15:e70226. [PMID: 39829139 PMCID: PMC11744024 DOI: 10.1002/brb3.70226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND The underlying mechanism of quetiapine (QET) in treating cognitive impairment in sleep deprivation is unclear. The present study aimed to evaluate the effects of treatment with QET on novel object recognition and hippocampal (hippo) brain-derived neurotrophic factor (BDNF) levels in rats submitted to 72 h sleep deprivation (SD). MATERIALS AND METHODS A total of 42 adult male Wistar albino rats were assigned into six experimental groups: non-sleep-deprived (NSD) control, short-term control group (n = 7) received a single intraperitoneal (i.p.) injection 10 mg/kg QET of 1 mL saline (4 days) (NSD-STQET), long-term control group (n = 7) received single i.p. injection 10 mg/kg QET of 1 mL saline (30 days) (NSD-LTQET); 72 h sleep-deprived (SD) group, 72 h SD short-term group received short-term i.p. injection 10 mg/kg QET of either (n = 7) (SD-STQET), and 72 h SD long-term group received long-term i.p. injection 10 mg/kg QET of either (n = 7) QET (SD-LTQET). SD was performed using the modified multiple-platform technique in a water tank for 72 h. Additionally, we aim to reveal the consequences of 72 h SD and QET effects on memory processes with hippo BDNF levels by testing rats in the novel object recognition (NOR) test and ELISA method. RESULTS Long-term QET administration in healthy rats decreased NOR and BDNF protein expression in the hippocampus, as did 72 h SD. Long- and short-term QET administration reversed SD effects, but only short-term QET administration increased hippo BDNF. CONCLUSION These results suggest that the beneficial effects of QET on SD may be partly related to the upregulation of recognition memory and neuroprotective proteins such as BDNF. However, long-term QET treatment in the absence of a disease model may have the potential to negatively impact recognition memory and BDNF levels, which support synaptic plasticity and cognitive function.
Collapse
Affiliation(s)
- Öznur Özge Özcan
- Electro‐Neurophysiology, Vocational School of Health SciencesÜsküdar UniversityİstanbulTurkey
| | - Burcu Çevreli
- Department of Physiology, Faculty of MedicineÜsküdar UniversityİstanbulTurkey
| | - Emel Serdaroğlu Kaşıkçı
- Department of Molecular Biology, Faculty of Engineering and Natural SciencesÜsküdar UniversityİstanbulTurkey
| | - Mesut Karahan
- Medical Laboratory Techniques, Vocational School of Health SciencesÜsküdar UniversityİstanbulTurkey
| | - Muhsin Konuk
- Department of Molecular Biology, Faculty of Engineering and Natural SciencesÜsküdar UniversityİstanbulTurkey
| |
Collapse
|
11
|
Jia M, Li G, Chen J, Tang X, Zang Y, Yang G, Shi YS, Ma D, Ji M, Yang J. Hippocampal Nogo66-NgR1 signaling activation restricts postsynaptic assembly in aged mice with postoperative neurocognitive disorders. Aging Cell 2025; 24:e14366. [PMID: 39412367 PMCID: PMC11709113 DOI: 10.1111/acel.14366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 09/05/2024] [Accepted: 09/17/2024] [Indexed: 01/11/2025] Open
Abstract
Postoperative neurocognitive disorders (pNCD) are a common neurological complication, especially in elderly following anesthesia and surgery. Yet, the underlying mechanisms of pNCD remain elusive. This study aimed to investigate the molecular mechanisms that compromise synaptic metaplasticity in pNCD development with a focus on the involvement of Nogo-66 receptor 1 (NgR1) in the pathogenesis of pNCD in aged mice. Aged mice subjected to anesthesia and laparotomy surgery exhibited anxiety-like behavior and contextual fear memory impairment. Moreover, the procedure significantly increased NogoA and NgR1 expressions, particularly in the hippocampal CA1 and CA3 regions. This increase led to the depolymerization of F-actin, attributed to the activation of the RhoA-GTPase, resulting in a reduction of dendritic spines and changes in their morphology. Additionally, these changes hindered the efficient postsynaptic delivery of the subunit GluA1 and GluA2 of AMPA receptors (AMPARs), consequently diminishing excitatory neurotransmission in the hippocampus. Importantly, administering the competitive NgR1 antagonist peptide NEP1-40 (Nogo-A extracellular peptide residues 1-40 amino acids of Nogo-66) and Fasudil (a Rho-kinase inhibitor) effectively mitigated synaptic impairments and reversed neurocognitive deficits in aged mice following anesthesia and surgery. Our work indicates that high hippocampal Nogo66-NgR1 signaling disrupts postsynaptic AMPA receptor surface delivery due to F-actin depolymerization in the pathophysiology of pNCD.
Collapse
Affiliation(s)
- Min Jia
- Department of Anaesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Gui‐zhou Li
- Minister of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research CenterNanjing UniversityNanjingChina
| | - Jiang Chen
- Minister of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research CenterNanjing UniversityNanjingChina
| | - Xiao‐hui Tang
- Department of Anaesthesiology and Perioperative MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yan‐yu Zang
- Minister of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research CenterNanjing UniversityNanjingChina
| | - Guo‐lin Yang
- Minister of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research CenterNanjing UniversityNanjingChina
| | - Yun Stone Shi
- Minister of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research CenterNanjing UniversityNanjingChina
| | - Daqing Ma
- Perioperative and Systems Medicine LaboratoryNational Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of MedicineHangzhouChina
- Division of Anaesthetics, Pain Medicine & Intensive Care, Department of Surgery & Cancer, Faculty of MedicineImperial College London, Chelsea & Westminster HospitalLondonUK
| | - Mu‐huo Ji
- Department of AnaesthesiologyThe Second Affiliated Hospital, Nanjing Medical UniversityNanjingChina
| | - Jian‐jun Yang
- Department of Anaesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
12
|
Tadinada SM, Walsh EN, Mukherjee U, Abel T. Differential effects of Phosphodiesterase 4A5 on cAMP-dependent forms of long-term potentiation. J Physiol 2024. [PMID: 39693518 DOI: 10.1113/jp286801] [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: 04/22/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
cAMP signalling is critical for memory consolidation and certain forms of long-term potentiation (LTP). Phosphodiesterases (PDEs), enzymes that degrade the second messengers cAMP and cGMP, are highly conserved during evolution and represent a unique set of drug targets, given the involvement of these enzymes in several pathophysiological states including brain disorders. The PDE4 family of cAMP-selective PDEs exert regulatory roles in memory and synaptic plasticity, but the specific roles of distinct PDE4 isoforms in these processes are poorly understood. Building on our previous work demonstrating that spatial and contextual memory deficits were caused by expressing selectively the long isoform of the PDE4A subfamily, PDE4A5, in hippocampal excitatory neurons, we now investigate the effects of PDE4A isoforms on different cAMP-dependent forms of LTP. We found that PDE4A5 impairs long-lasting LTP induced by theta burst stimulation (TBS) while sparing long-lasting LTP induced by spaced four-train stimulation (4 × 100 Hz). This effect requires the unique N-terminus of PDE4A5 and is specific to this long isoform. Targeted overexpression of PDE4A5 in area CA1 is sufficient to impair TBS-LTP, suggesting that cAMP levels in the postsynaptic neuron are critical for TBS-LTP. Our results shed light on the inherent differences among the PDE4A subfamily isoforms, emphasizing the importance of the long isoforms, which have a unique N-terminal region. Advancing our understanding of the function of specific PDE isoforms will pave the way for developing isoform-selective approaches to treat the cognitive deficits that are debilitating aspects of psychiatric, neurodevelopmental and neurodegenerative disorders. KEY POINTS: Hippocampal overexpression of PDE4A5, but not PDE4A1 or the N-terminus-truncated PDE4A5 (PDE4A5Δ4), selectively impairs long-term potentiation (LTP) induced by theta burst stimulation (TBS-LTP). Expression of PDE4A5 in area CA1 is sufficient to cause deficits in TBS-LTP. Hippocampal overexpression of the PDE4A isoforms PDE4A1 and PDE4A5 does not impair LTP induced by repeated tetanic stimulation at the CA3-CA1 synapses. These results suggest that PDE4A5, through its N-terminus, regulates cAMP pools that are critical for memory consolidation and expression of specific forms of long-lasting synaptic plasticity at CA3-CA1 synapses.
Collapse
Affiliation(s)
- Satya Murthy Tadinada
- Department of Neuroscience and Pharmacology, The University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, USA
| | - Emily N Walsh
- Department of Neuroscience and Pharmacology, The University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, The University of Iowa, Iowa City, IA, USA
| | - Utsav Mukherjee
- Department of Neuroscience and Pharmacology, The University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, The University of Iowa, Iowa City, IA, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, The University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, The University of Iowa, Iowa City, IA, USA
| |
Collapse
|
13
|
Chen W, Liu M, Li Z, Luo Z, Wu J. Phloretin alleviates sleep deprivation-induced cognitive impairment by reducing inflammation through PPARγ/NF-κB signaling pathway. Exp Neurol 2024; 382:114949. [PMID: 39284540 DOI: 10.1016/j.expneurol.2024.114949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/29/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Sleep loss leads to significant pathophysiological consequences, including cognitive impairment. The neuroinflammation are pivotal factors in the pathogenesis of cognitive impairment induced by sleep loss. The phloretin (PHL), derived from peel of juicy fruits, has demonstrated potent anti-inflammatory properties. However, the precise influence of PHL on the cognitive impairment triggered by sleep loss and its underlying mechanism remain uncertain. In the present study, mice were subjected to sleep deprivation (SD) paradigm. Cognitive impairment induced by SD were significantly relieved by administration of PHL in a dose-dependent manner. Furthermore, PHL not only mitigated the synaptic losses but also enhanced dendritic spine density and neuronal activity within mice hippocampus following exposure to SD. Moreover, PHL treatment decreased the microglial numbers and altered microglial morphology in the hippocampus to restore the M1/M2 balances; these effects were accompanied by regulation of pro-/anti-inflammatory cytokine production and secretion in SD-exposed mice. Additionally, in vivo and in vitro studies showed PHL might attenuate the inflammation through the PPARγ/NF-κB pathway. Our findings suggest that PHL exerts inhibitory effects on microglia-mediated neuroinflammation, thereby providing protection against cognitive impairment induced by SD through a PPAR-γ dependent mechanism. The results indicate PHL is expected to provide a valuable candidate for new drug development for SD-induced cognitive impairment in the future.
Collapse
Affiliation(s)
- Wenjun Chen
- Research Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou 514031, China; Meizhou Clinical Medical College of Guangdong Medical University, Meizhou 514000, China; Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou 514000, China.
| | - Mei Liu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Afffliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Ziming Li
- Department of Neurobiology, Southern Medical University, Guangzhou 510515, China
| | - Zhoucai Luo
- National Canine Laboratory Animal Resources Center, Guangzhou General Pharmaceutical Research Institute Co., Ltd., Guangzhou 510240, China
| | - Jianlin Wu
- Research Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou 514031, China; Meizhou Clinical Medical College of Guangdong Medical University, Meizhou 514000, China; Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou 514000, China.
| |
Collapse
|
14
|
Al Doori L, Zaki PG, Joshi V. Impact of USMLE Step 1 transition to pass/fail scoring system on medical students' anxiety, sleep quality, and burnout. Ir J Med Sci 2024; 193:2155-2160. [PMID: 38954326 PMCID: PMC11450038 DOI: 10.1007/s11845-024-03738-x] [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: 03/18/2024] [Accepted: 06/12/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION Originally designed to evaluate the application of foundational scientific knowledge in clinical contexts, the United States Medical License Exam (USMLE) Step 1 evolved into a comparative tool for assessing candidates with similar educational foundations. This transition heightened the pressure on medical students to excel in the exam. In response, collaborative efforts involving the National Board of Medical Examiners prompted a change from reporting scores to a pass/fail system. The true impact of this shift remains insufficiently explored. This study aims to assess the emotional toll - encompassing burnout, anxiety, depression, and sleep quality - experienced prior to taking the Step 1 exam. Additionally, it aims to uncover potential gender-based disparities in perceived anxiety and depression. METHODS The study encompasses the entirety of third-year medical students at Drexel University College of Medicine, who were invited to participate in a comprehensive survey. Drawing from retrospection, the survey relies on self-reported data regarding anxiety, depression, sleep quality, and burnout. Data compilation was anonymized and executed via Qualtrics platform. RESULTS A total of 102 medical students completed the survey, with a 97% pass rate for the USMLE Step 1. Despite their excellent performance, 75% of students reported inadequate sleep quality, and 68% exhibited mild to moderate anxiety levels. Among them, a higher percentage of females (83%) experienced anxiety compared to their male counterparts (50%). Furthermore, 66% of students felt that their commitment to education exceeded what was reasonable for their well-being. CONCLUSIONS The transition from traditional scoring to a pass/fail system was ostensibly intended to enhance the mental well-being of medical students. Nevertheless, our findings underscore that students continue to endure heightened levels of stress, anxiety, and burnout during the pivotal month leading up to the Step 1 examination.
Collapse
Affiliation(s)
- Lana Al Doori
- Drexel University College of Medicine at Tower Health, Drexel University, West Reading, Wyomissing, PA, 19610, USA
| | - Peter G Zaki
- Drexel University College of Medicine at Tower Health, Drexel University, West Reading, Wyomissing, PA, 19610, USA
| | - Vivek Joshi
- Drexel University College of Medicine at Tower Health, Drexel University, West Reading, Wyomissing, PA, 19610, USA.
- Department of Biochemistry, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| |
Collapse
|
15
|
Popescu A, Ottaway C, Ford K, Patterson TW, Ingiosi A, Medina E, Hicks SC, Singletary K, Peixoto L. Transcriptional dynamics of sleep deprivation and subsequent recovery sleep in the male mouse cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.607983. [PMID: 39229182 PMCID: PMC11370348 DOI: 10.1101/2024.08.20.607983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Sleep is an essential, tightly regulated biological function. Sleep is also a homeostatic process, with the need to sleep increasing as a function of being awake. Acute sleep deprivation (SD) increases sleep need, and subsequent recovery sleep (RS) discharges it. SD is known to alter brain gene expression in rodents, but it remains unclear which changes are linked to sleep homeostasis, SD-related impairments, or non-sleep-specific effects. To investigate this question, we analyzed RNA-seq data from adult wild-type male mice subjected to 3 and 5-6 hours of SD and 2 and 6 hours of RS after SD. We hypothesized molecular changes associated with sleep homeostasis mirror sleep pressure dynamics as defined by brain electrical activity, peaking at 5-6 hours of SD, and are no longer differentially expressed after 2 hours of RS. We report 5-6 hours of SD produces the largest effect on gene expression, affecting approximately half of the cortical transcriptome, with most differentially expressed genes (DEGs) downregulated. The majority of DEGs normalize after 2 hours of RS and are involved in redox metabolism, chromatin regulation, and DNA damage/repair. Additionally, RS affects gene expression related to mitochondrial metabolism and Wnt-signaling, potentially contributing to its restorative effects. DEGs associated with cholesterol metabolism and stress response do not normalize within 6 hours and may be non-sleep-specific. Finally, DEGs involved in insulin signaling, MAPK signaling, and RNA-binding may mediate the impairing effects of SD. Overall, our results offer insight into the molecular mechanisms underlying sleep homeostasis and the broader effects of SD.
Collapse
Affiliation(s)
- Alexander Popescu
- Department of Translational Medicine and Physiology, Sleep and Performance Research Center. Elson S. Floyd College of Medicine. Washington State University, Spokane, WA, USA
| | - Caitlin Ottaway
- Department of Translational Medicine and Physiology, Sleep and Performance Research Center. Elson S. Floyd College of Medicine. Washington State University, Spokane, WA, USA
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Kaitlyn Ford
- Department of Translational Medicine and Physiology, Sleep and Performance Research Center. Elson S. Floyd College of Medicine. Washington State University, Spokane, WA, USA
| | - Taylor Wintler Patterson
- Department of Translational Medicine and Physiology, Sleep and Performance Research Center. Elson S. Floyd College of Medicine. Washington State University, Spokane, WA, USA
| | - Ashley Ingiosi
- Department of Translational Medicine and Physiology, Sleep and Performance Research Center. Elson S. Floyd College of Medicine. Washington State University, Spokane, WA, USA
| | - Elizabeth Medina
- Department of Translational Medicine and Physiology, Sleep and Performance Research Center. Elson S. Floyd College of Medicine. Washington State University, Spokane, WA, USA
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| | - Kristan Singletary
- Department of Translational Medicine and Physiology, Sleep and Performance Research Center. Elson S. Floyd College of Medicine. Washington State University, Spokane, WA, USA
| | - Lucia Peixoto
- Department of Translational Medicine and Physiology, Sleep and Performance Research Center. Elson S. Floyd College of Medicine. Washington State University, Spokane, WA, USA
| |
Collapse
|
16
|
Koukaroudi D, Qiu Z, Fransén E, Gokhale R, Bulovaite E, Komiyama NH, Seibt J, Grant SGN. Sleep maintains excitatory synapse diversity in the cortex and hippocampus. Curr Biol 2024; 34:3836-3843.e5. [PMID: 39096907 PMCID: PMC11359089 DOI: 10.1016/j.cub.2024.07.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/17/2024] [Accepted: 07/05/2024] [Indexed: 08/05/2024]
Abstract
Insufficient sleep is a global problem with serious consequences for cognition and mental health.1 Synapses play a central role in many aspects of cognition, including the crucial function of memory consolidation during sleep.2 Interference with the normal expression or function of synapse proteins is a cause of cognitive, mood, and other behavioral problems in over 130 brain disorders.3 Sleep deprivation (SD) has also been reported to alter synapse protein composition and synapse number, although with conflicting results.4,5,6,7 In our study, we conducted synaptome mapping of excitatory synapses in 125 regions of the mouse brain and found that sleep deprivation selectively reduces synapse diversity in the cortex and in the CA1 region of the hippocampus. Sleep deprivation targeted specific types and subtypes of excitatory synapses while maintaining total synapse density (synapse number/area). Synapse subtypes with longer protein lifetimes exhibited resilience to sleep deprivation, similar to observations in aging and genetic perturbations. Moreover, the altered synaptome architecture affected the responses to neural oscillations, suggesting that sleep plays a vital role in preserving cognitive function by maintaining the brain's synaptome architecture.
Collapse
Affiliation(s)
- Dimitra Koukaroudi
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Zhen Qiu
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; School of Science and Engineering, University of Dundee, Dundee DD1 4HN, UK
| | - Erik Fransén
- Department of Computational Science and Technology, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, 10044 Stockholm, Sweden; Science for Life Laboratory, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - Ragini Gokhale
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Edita Bulovaite
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Noboru H Komiyama
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK; The Patrick Wild Centre for Research into Autism, Fragile X Syndrome & Intellectual Disabilities, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK; Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Julie Seibt
- Surrey Sleep Research Centre, School of Biosciences, University of Surrey, Guildford, Surrey GU2 7XP, UK
| | - Seth G N Grant
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.
| |
Collapse
|
17
|
Kolobaric A, Saleska J, Hewlings SJ, Bryant C, Colwell CS, D'Adamo CR, Chen J, Pauli EK. A Randomized, Double-Blind, Placebo-Controlled Trial to Assess the Effectiveness and Safety of Melatonin and Three Formulations of Floraworks Proprietary TruCBN™ for Improving Sleep. Pharmaceuticals (Basel) 2024; 17:977. [PMID: 39204082 PMCID: PMC11357382 DOI: 10.3390/ph17080977] [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: 06/16/2024] [Revised: 07/18/2024] [Accepted: 07/21/2024] [Indexed: 09/03/2024] Open
Abstract
The phytocannabinoid cannabinol (CBN) has a potential mechanism of action as an alternative sleep aid but there is minimal evidence to support its effectiveness. The aim of this randomized, double-blind, placebo-controlled study was to assess the safety and effects of three formulations of a hemp-derived CBN sleep aid, TruCBN™ [25 mg (n = 206), 50 mg (n = 205), 100 mg (n = 203)], on sleep quality (PROMIS Sleep Disturbance 8A), relative to placebo (n = 204). The effectiveness and safety of these formulations relative to 4 mg of melatonin (n = 202) was assessed. Exploratory measures were stress (PROMIS Stress 4A), anxiety (Anxiety 4A), pain (PROMIS™ PEG), and well-being (WHO 5). All groups and the 4 mg melatonin group experienced significant improvement in sleep quality relative to the placebo group with no significant differences between any group and the melatonin group. Participants taking 100 mg showed a larger decrease in stress compared to the placebo group. There were no significant differences in anxiety, pain, well-being, or the frequency of side effects between any group and the placebo group. There was no significant difference in improvements in sleep quality between any of the treatment groups and the 4 mg melatonin group. Orally ingested CBN, at 25 mg, 50 mg, and 100 mg, is a safe and effective alternative for the improvement of sleep.
Collapse
Affiliation(s)
- Antonija Kolobaric
- Radicle Science, Inc., Del Mar, CA 92014, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | | | | | - Christopher S Colwell
- Department of Psychiatry & Biobehavioral Sciences, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Christopher R D'Adamo
- Department of Family and Community Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jeff Chen
- Radicle Science, Inc., Del Mar, CA 92014, USA
| | | |
Collapse
|
18
|
Pifer GC, Ferrara NC, Kwapis JL. Long-lasting effects of disturbing the circadian rhythm or sleep in adolescence. Brain Res Bull 2024; 213:110978. [PMID: 38759704 PMCID: PMC11197883 DOI: 10.1016/j.brainresbull.2024.110978] [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: 03/22/2024] [Revised: 05/02/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Circadian rhythms are endogenous, near 24-hour rhythms that regulate a multitude of biological and behavioral processes across the diurnal cycle in most organisms. Over the lifespan, a bell curve pattern emerges in circadian phase preference (i.e. chronotype), with children and adults generally preferring to wake earlier and fall asleep earlier, and adolescents and young adults preferring to wake later and fall asleep later than their adult counterparts. This well-defined shift speaks to the variability of circadian rhythmicity over the lifespan and the changing needs and demands of the brain as an organism develops, particularly in the adolescent period. Indeed, adolescence is known to be a critical period of development during which dramatic neuroanatomical changes are occurring to allow for improved decision-making. Due to the large amount of re-structuring occurring in the adolescent brain, circadian disruptions during this period could have adverse consequences that persist across the lifespan. While the detrimental effects of circadian disruptions in adults have been characterized in depth, few studies have longitudinally assessed the potential long-term impacts of circadian disruptions during adolescence. Here, we will review the evidence that disruptions in circadian rhythmicity during adolescence have effects that persist into adulthood. As biological and social time often conflict in modern society, with school start times misaligned with adolescents' endogenous rhythms, it is critical to understand the long-term impacts of disrupted circadian rhythmicity in adolescence.
Collapse
Affiliation(s)
- Gretchen C Pifer
- Department of Biology, The Pennsylvania State University, University Park, PA, USA
| | - Nicole C Ferrara
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Discipline of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Janine L Kwapis
- Department of Biology, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
19
|
Martinez JD, Wilson LG, Brancaleone WP, Peterson KG, Popke DS, Garzon VC, Perez Tremble RE, Donnelly MJ, Mendez Ortega SL, Torres D, Shaver JJ, Jiang S, Yang Z, Aton SJ. Hypnotic treatment improves sleep architecture and EEG disruptions and rescues memory deficits in a mouse model of fragile X syndrome. Cell Rep 2024; 43:114266. [PMID: 38787724 PMCID: PMC11910971 DOI: 10.1016/j.celrep.2024.114266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 12/20/2023] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Fragile X syndrome (FXS) is associated with disrupted cognition and sleep abnormalities. Sleep loss negatively impacts cognitive function, and one untested possibility is that disrupted cognition in FXS is exacerbated by abnormal sleep. We tested whether ML297, a hypnotic acting on G-protein-activated inward-rectifying potassium (GIRK) channels, could reverse sleep phenotypes and disrupted memory in Fmr1-/y mice. Fmr1-/y mice exhibit reduced non-rapid eye movement (NREM) sleep and fragmented NREM architecture, altered sleep electroencephalogram (EEG) oscillations, and reduced EEG coherence between cortical areas; these are partially reversed following ML297 administration. Treatment following contextual fear or spatial learning restores disrupted memory consolidation in Fmr1-/y mice. During memory recall, Fmr1-/y mice show an altered balance of activity among hippocampal principal neurons vs. parvalbumin-expressing interneurons; this is partially reversed by ML297. Because sleep disruption could impact neurophysiological phenotypes in FXS, augmenting sleep may improve disrupted cognition in this disorder.
Collapse
Affiliation(s)
- Jessy D Martinez
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lydia G Wilson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - William P Brancaleone
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kathryn G Peterson
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | - Donald S Popke
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | - Valentina Caicedo Garzon
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Roxanne E Perez Tremble
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marcus J Donnelly
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Daniel Torres
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J Shaver
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sha Jiang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhongying Yang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sara J Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
20
|
Chen C, Zhai R, Lan X, Yang S, Tang S, Xiong X, He Y, Lin J, Feng J, Chen D, Shi J. The influence of sleep disorders on perioperative neurocognitive disorders among the elderly: A narrative review. IBRAIN 2024; 10:197-216. [PMID: 38915944 PMCID: PMC11193868 DOI: 10.1002/ibra.12167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/26/2024]
Abstract
This review comprehensively assesses the epidemiology, interaction, and impact on patient outcomes of perioperative sleep disorders (SD) and perioperative neurocognitive disorders (PND) in the elderly. The incidence of SD and PND during the perioperative period in older adults is alarmingly high, with SD significantly contributing to the occurrence of postoperative delirium. However, the clinical evidence linking SD to PND remains insufficient, despite substantial preclinical data. Therefore, this study focuses on the underlying mechanisms between SD and PND, underscoring that potential mechanisms driving SD-induced PND include uncontrolled central nervous inflammation, blood-brain barrier disruption, circadian rhythm disturbances, glial cell dysfunction, neuronal and synaptic abnormalities, impaired central metabolic waste clearance, gut microbiome dysbiosis, hippocampal oxidative stress, and altered brain network connectivity. Additionally, the review also evaluates the effectiveness of various sleep interventions, both pharmacological and nonpharmacological, in mitigating PND. Strategies such as earplugs, eye masks, restoring circadian rhythms, physical exercise, noninvasive brain stimulation, dexmedetomidine, and melatonin receptor agonists have shown efficacy in reducing PND incidence. The impact of other sleep-improvement drugs (e.g., orexin receptor antagonists) and methods (e.g., cognitive-behavioral therapy for insomnia) on PND is still unclear. However, certain drugs used for treating SD (e.g., antidepressants and first-generation antihistamines) may potentially aggravate PND. By providing valuable insights and references, this review aimed to enhance the understanding and management of PND in older adults based on SD.
Collapse
Affiliation(s)
- Chao Chen
- Department of Anesthesiology/Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Rui‐Xue Zhai
- Department of Anesthesiology/Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Xin Lan
- Department of Anesthesiology/Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Sheng‐Feng Yang
- Department of Anesthesiology/Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Si‐Jie Tang
- Department of Anesthesiology/Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Xing‐Long Xiong
- Department of Anesthesiology/Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Yu‐Xin He
- Department of Gastroenterology and HepatologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Jing‐Fang Lin
- Department of Anesthesiology, Fujian Provincial HospitalSheng Li Clinical Medical College of Fujian Medical UniversityFuzhouChina
| | - Jia‐Rong Feng
- Khoury College of Computer SciencesNortheastern UniversityBostonAmerica
| | - Dong‐Xu Chen
- Department of Anesthesiology, West China Second HospitalSichuan UniversityChengduChina
| | - Jing Shi
- Department of Anesthesiology/Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| |
Collapse
|
21
|
Li Y, Hou S, Li F, Long S, Yang Y, Li Y, Zhao L, Yu Y. Preoperative recovery sleep ameliorates postoperative cognitive dysfunction aggravated by sleep fragmentation in aged mice by enhancing EEG delta-wave activity and LFP theta oscillation in hippocampal CA1. Brain Res Bull 2024; 211:110945. [PMID: 38608544 DOI: 10.1016/j.brainresbull.2024.110945] [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: 12/07/2023] [Revised: 03/10/2024] [Accepted: 04/08/2024] [Indexed: 04/14/2024]
Abstract
Sleep fragmentation (SF) is a common sleep problem experienced during the perioperative period by older adults, and is associated with postoperative cognitive dysfunction (POCD). Increasing evidence indicates that delta-wave activity during non-rapid eye movement (NREM) sleep is involved in sleep-dependent memory consolidation and that hippocampal theta oscillations are related to spatial exploratory memory. Recovery sleep (RS), a self-regulated state of sleep homeostasis, enhances delta-wave power and memory performance in sleep-deprived older mice. However, it remains unclear whether RS therapy has a positive effect on cognitive changes following SF in older mouse models. Therefore, this study aimed to explore whether preoperative RS can alleviate cognitive deficits in aged mice with SF. A model of preoperative 24-h SF combined with exploratory laparotomy-induced POCD was established in 18-month-old mice. Aged mice were treated with preoperative 6-h RS following SF and postoperative 6-h RS following surgery, respectively. The changes in hippocampus-dependent cognitive function were investigated using behavioral tests, electroencephalography (EEG), local field potential (LFP), magnetic resonance imaging, and neuromorphology. Mice that underwent 24-h SF combined with surgery exhibited severe spatial memory impairment; impaired cognitive performance could be alleviated by preoperative RS treatment. In addition, preoperative RS increased NREM sleep; enhanced EEG delta-wave activity and LFP theta oscillation in the hippocampal CA1; and improved hippocampal perfusion, microstructural integrity, and neuronal damage. Taken together, these results provide evidence that preoperative RS may ameliorate the severity of POCD aggravated by SF by enhancing delta slow-wave activity and hippocampal theta oscillation, and by ameliorating the reduction in regional cerebral blood flow and white matter microstructure integrity in the hippocampus.
Collapse
Affiliation(s)
- Yun Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Shaowei Hou
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin 300070, China
| | - Feixiang Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Siwen Long
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Yue Yang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Yize Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Lina Zhao
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China.
| |
Collapse
|
22
|
Giri B, Kinsky N, Kaya U, Maboudi K, Abel T, Diba K. Sleep loss diminishes hippocampal reactivation and replay. Nature 2024; 630:935-942. [PMID: 38867049 PMCID: PMC11472378 DOI: 10.1038/s41586-024-07538-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/07/2024] [Indexed: 06/14/2024]
Abstract
Memories benefit from sleep1, and the reactivation and replay of waking experiences during hippocampal sharp-wave ripples (SWRs) are considered to be crucial for this process2. However, little is known about how these patterns are impacted by sleep loss. Here we recorded CA1 neuronal activity over 12 h in rats across maze exploration, sleep and sleep deprivation, followed by recovery sleep. We found that SWRs showed sustained or higher rates during sleep deprivation but with lower power and higher frequency ripples. Pyramidal cells exhibited sustained firing during sleep deprivation and reduced firing during sleep, yet their firing rates were comparable during SWRs regardless of sleep state. Despite the robust firing and abundance of SWRs during sleep deprivation, we found that the reactivation and replay of neuronal firing patterns was diminished during these periods and, in some cases, completely abolished compared to ad libitum sleep. Reactivation partially rebounded after recovery sleep but failed to reach the levels found in natural sleep. These results delineate the adverse consequences of sleep loss on hippocampal function at the network level and reveal a dissociation between the many SWRs elicited during sleep deprivation and the few reactivations and replays that occur during these events.
Collapse
Affiliation(s)
- Bapun Giri
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Nathaniel Kinsky
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Utku Kaya
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Kourosh Maboudi
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Kamran Diba
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
23
|
Pandi-Perumal SR, Saravanan KM, Paul S, Namasivayam GP, Chidambaram SB. Waking Up the Sleep Field: An Overview on the Implications of Genetics and Bioinformatics of Sleep. Mol Biotechnol 2024; 66:919-931. [PMID: 38198051 DOI: 10.1007/s12033-023-01009-1] [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: 08/04/2023] [Accepted: 11/28/2023] [Indexed: 01/11/2024]
Abstract
Sleep genetics is an intriguing, as yet less understood, understudied, emerging area of biological and medical discipline. A generalist may not be aware of the current status of the field given the variety of journals that have published studies on the genetics of sleep and the circadian clock over the years. For researchers venturing into this fascinating area, this review thus includes fundamental features of circadian rhythm and genetic variables impacting sleep-wake cycles. Sleep/wake pathway medication exposure and susceptibility are influenced by genetic variations, and the responsiveness of sleep-related medicines is influenced by several functional polymorphisms. This review highlights the features of the circadian timing system and then a genetic perspective on wakefulness and sleep, as well as the relationship between sleep genetics and sleep disorders. Neurotransmission genes, as well as circadian and sleep/wake receptors, exhibit functional variability. Experiments on animals and humans have shown that these genetic variants impact clock systems, signaling pathways, nature, amount, duration, type, intensity, quality, and quantity of sleep. In this regard, the overview covers research on sleep genetics, the genomic properties of several popular model species used in sleep studies, homologs of mammalian genes, sleep disorders, and related genes. In addition, the study includes a brief discussion of sleep, narcolepsy, and restless legs syndrome from the viewpoint of a model organism. It is suggested that the understanding of genetic clues on sleep function and sleep disorders may, in future, result in an evidence-based, personalized treatment of sleep disorders.
Collapse
Affiliation(s)
- Seithikurippu R Pandi-Perumal
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru, Karnataka, 570015, India
- Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, 602105, India
- Division of Research and Development, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Konda Mani Saravanan
- Department of Biotechnology, Bharath Institute of Higher Education and Research, Chennai, Tamil Nadu, 600073, India
| | - Sayan Paul
- Department of Biochemistry & Molecular Biology, The University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA
| | - Ganesh Pandian Namasivayam
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), A210, Kyoto University Institute for Advanced Study, Yoshida Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Saravana Babu Chidambaram
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru, Karnataka, 570015, India.
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka, 570015, India.
- Special Interest Group - Brain, Behaviour and Cognitive Neurosciences, JSS Academy of Higher Education & Research, Mysuru, Karnataka, 570015, India.
| |
Collapse
|
24
|
Cowen MH, Raizen DM, Hart MP. Structural neuroplasticity after sleep loss modifies behavior and requires neurexin and neuroligin. iScience 2024; 27:109477. [PMID: 38551003 PMCID: PMC10973677 DOI: 10.1016/j.isci.2024.109477] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/05/2023] [Accepted: 03/08/2024] [Indexed: 02/08/2025] Open
Abstract
Structural neuroplasticity (changes in the size, strength, number, and targets of synaptic connections) can be modified by sleep and sleep disruption. However, the causal relationships between genetic perturbations, sleep loss, neuroplasticity, and behavior remain unclear. The C. elegans GABAergic DVB neuron undergoes structural plasticity in adult males in response to adolescent stress, which rewires synaptic connections, alters behavior, and is dependent on conserved autism-associated genes NRXN1/nrx-1 and NLGN3/nlg-1. We find that four methods of sleep deprivation transiently induce DVB neurite extension in day 1 adults and increase the time to spicule protraction, which is the functional and behavioral output of the DVB neuron. Loss of nrx-1 and nlg-1 prevent DVB structural plasticity and behavioral changes at day 1 caused by adolescent sleep loss. Therefore, nrx-1 and nlg-1 mediate the morphologic and behavioral consequences of sleep loss, providing insight into the relationship between sleep, neuroplasticity, behavior, and neurologic disease.
Collapse
Affiliation(s)
- Mara H. Cowen
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David M. Raizen
- Department of Neurology and the Chronobiology and Sleep Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Michael P. Hart
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
25
|
Wang L, Park L, Wu W, King D, Vega-Medina A, Raven F, Martinez J, Ensing A, McDonald K, Yang Z, Jiang S, Aton SJ. Sleep-dependent engram reactivation during hippocampal memory consolidation associated with subregion-specific biosynthetic changes. iScience 2024; 27:109408. [PMID: 38523798 PMCID: PMC10957462 DOI: 10.1016/j.isci.2024.109408] [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: 10/11/2023] [Revised: 01/14/2024] [Accepted: 02/29/2024] [Indexed: 03/26/2024] Open
Abstract
Post-learning sleep is essential for hippocampal memory processing, including contextual fear memory consolidation. We labeled context-encoding engram neurons in the hippocampal dentate gyrus (DG) and assessed reactivation of these neurons after fear learning. Post-learning sleep deprivation (SD) selectively disrupted reactivation of inferior blade DG engram neurons, linked to SD-induced suppression of neuronal activity in the inferior, but not superior DG blade. Subregion-specific spatial profiling of transcripts revealed that transcriptomic responses to SD differed greatly between hippocampal CA1, CA3, and DG inferior blade, superior blade, and hilus. Activity-driven transcripts, and those associated with cytoskeletal remodeling, were selectively suppressed in the inferior blade. Critically, learning-driven transcriptomic changes differed dramatically between the DG blades and were absent from all other regions. Together, these data suggest that the DG is critical for sleep-dependent memory consolidation, and that the effects of sleep loss on the hippocampus are highly subregion-specific.
Collapse
Affiliation(s)
- Lijing Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lauren Park
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Weisheng Wu
- Bioinformatics Core, Biomedical Research Core Facilities, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dana King
- Bioinformatics Core, Biomedical Research Core Facilities, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alexis Vega-Medina
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Frank Raven
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jessy Martinez
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Amy Ensing
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Katherine McDonald
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhongying Yang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sha Jiang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sara J. Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
26
|
Riemersma IW, Ike KGO, Sollie T, Meijer EL, Havekes R, Kas MJH. Suppression of Cofilin function in the somatosensory cortex alters social contact behavior in the BTBR mouse inbred line. Cereb Cortex 2024; 34:bhae136. [PMID: 38602737 PMCID: PMC11008688 DOI: 10.1093/cercor/bhae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 03/08/2024] [Accepted: 03/09/2024] [Indexed: 04/12/2024] Open
Abstract
Sensory differences are a core feature of autism spectrum disorders (ASD) and are predictive of other ASD core symptoms such as social difficulties. However, the neurobiological substrate underlying the functional relationship between sensory and social functioning is poorly understood. Here, we examined whether misregulation of structural plasticity in the somatosensory cortex modulates aberrant social functioning in BTBR mice, a mouse model for autism spectrum disorder-like phenotypes. By locally expressing a dominant-negative form of Cofilin (CofilinS3D; a key regulator of synaptic structure) in the somatosensory cortex, we tested whether somatosensory suppression of Cofilin activity alters social functioning in BTBR mice. Somatosensory Cofilin suppression altered social contact and nest-hide behavior of BTBR mice in a social colony, assessed for seven consecutive days. Subsequent behavioral testing revealed that altered social functioning is related to altered tactile sensory perception; CofilinS3D-treated BTBR mice showed a time-dependent difference in the sensory bedding preference task. These findings show that Cofilin suppression in the somatosensory cortex alters social functioning in BTBR mice and that this is associated with tactile sensory processing, a critical indicator of somatosensory functioning.
Collapse
Affiliation(s)
- Iris W Riemersma
- Groningen Institute for Evolutionary Life Sciences, Neurobiology, University of Groningen , Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Kevin G O Ike
- Groningen Institute for Evolutionary Life Sciences, Neurobiology, University of Groningen , Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Thomas Sollie
- Groningen Institute for Evolutionary Life Sciences, Neurobiology, University of Groningen , Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Elroy L Meijer
- Groningen Institute for Evolutionary Life Sciences, Neurobiology, University of Groningen , Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Robbert Havekes
- Groningen Institute for Evolutionary Life Sciences, Neurobiology, University of Groningen , Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Martien J H Kas
- Groningen Institute for Evolutionary Life Sciences, Neurobiology, University of Groningen , Nijenborgh 7, 9747 AG Groningen, The Netherlands
| |
Collapse
|
27
|
Dalferth TF, Nunes ML, Furini CRG. Sleep deprivation in early life: Cellular and behavioral impacts. Neurosci Biobehav Rev 2024; 159:105597. [PMID: 38387838 DOI: 10.1016/j.neubiorev.2024.105597] [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: 09/18/2023] [Revised: 02/08/2024] [Accepted: 02/18/2024] [Indexed: 02/24/2024]
Abstract
Sleep deprivation has become increasingly prevalent in contemporary society, and the consequences of this reality such as cognitive impairment and metabolic disorders, are widely investigated in the scientific scenario. However, the impact of sleep deprivation on the health of future generations is a challenge, and researchers are focusing their attention on this issue. Thus, this review aims to describe the impact of sleep deprivation in early life in animal models, particularly rodents, discussing the molecular physiology impacted by prolonged wakefulness in early life, as well as the changes that interfere with neurodevelopmental processes. Additionally, it explores the changes impacting metabolic mechanisms and discusses both the short- and long-term consequences of these processes on endocrine, behavioral, and cognitive functions. Finally, we briefly address some strategies to mitigate the adverse effects of sleep deprivation.
Collapse
Affiliation(s)
- Thais F Dalferth
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, Porto Alegre, RS 90619-900, Brazil; Laboratory of Cognition and Memory Neurobiology, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 3rd floor, Porto Alegre, RS 90610-000, Brazil
| | - Magda L Nunes
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, Porto Alegre, RS 90619-900, Brazil; Brain Institute (InsCer), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690, Porto Alegre, RS 90610-000, Brazil
| | - Cristiane R G Furini
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, Porto Alegre, RS 90619-900, Brazil; Laboratory of Cognition and Memory Neurobiology, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 3rd floor, Porto Alegre, RS 90610-000, Brazil.
| |
Collapse
|
28
|
Lutfy RH, Essawy AE, Mohammed HS, Shakweer MM, Salam SA. Transcranial Irradiation Mitigates Paradoxical Sleep Deprivation Effect in an Age-Dependent Manner: Role of BDNF and GLP-1. Neurochem Res 2024; 49:919-934. [PMID: 38114728 PMCID: PMC10902205 DOI: 10.1007/s11064-023-04071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/15/2023] [Accepted: 11/18/2023] [Indexed: 12/21/2023]
Abstract
The growing prevalence of aged sleep-deprived nations is turning into a pandemic state. Acute sleep deprivation (SD) accompanies aging, changing the hippocampal cellular pattern, neurogenesis pathway expression, and aggravating cognitive deterioration. The present study investigated the ability of Near Infra Red (NIR) light laser to ameliorate cognitive impairment induced by SD in young and senile rats. Wistar rats ≤ 2 months (young) and ≥ 14 months (senile) were sleep-deprived for 72 h with or without transcranial administration of NIR laser of 830 nm. Our results showed that NIR photobiomodulation (PBM) attenuated cognitive deterioration made by SD in young, but not senile rats, while both sleep-deprived young and senile rats exhibited decreased anxiety (mania)-like behavior in response to PBM. NIR PBM had an inhibitory effect on AChE, enhanced the production of ACh, attenuated ROS, and regulated cell apoptosis factors such as Bax and Bcl-2. NIR increased mRNA expression of BDNF and GLP-1 in senile rats, thus facilitating neuronal survival and differentiation. The present findings also revealed that age exerts an additive factor to the cellular assaults produced by SD where hippocampal damages made in 2-month rats were less severe than those of the aged one. In conclusion, NIR PBM seems to promote cellular longevity of senile hippocampal cells by combating ROS, elevating neurotrophic factors, thus improving cognitive performance. The present findings provide NIR as a possible candidate for hippocampal neuronal insults accompanying aging and SD.
Collapse
Affiliation(s)
- Radwa H Lutfy
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, 11829, Egypt
| | - Amina E Essawy
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
| | - Haitham S Mohammed
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt
| | - Marwa M Shakweer
- Department of Pathology, Faculty of Medicine, Badr University in Cairo (BUC), Cairo, Egypt
- Department of Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sherine Abdel Salam
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt.
| |
Collapse
|
29
|
Misrani A, Tabassum S, Zhang ZY, Tan SH, Long C. Urolithin A Prevents Sleep-deprivation-induced Neuroinflammation and Mitochondrial Dysfunction in Young and Aged Mice. Mol Neurobiol 2024; 61:1448-1466. [PMID: 37725214 DOI: 10.1007/s12035-023-03651-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 09/10/2023] [Indexed: 09/21/2023]
Abstract
Sleep deprivation (SD) has reached epidemic proportions worldwide and negatively affects people of all ages. Cognitive impairment induced by SD involves neuroinflammation and mitochondrial dysfunction, but the underlying mechanisms are largely unknown. Urolithin A (UA) is a natural compound that can reduce neuroinflammation and improve mitochondrial health, but its therapeutic effects in a SD model have not yet been studied. Young (3-months old) and aged (12-months old) mice were sleep deprived for 24 h, and UA (2.5 mg/kg or 10 mg/kg) was injected intraperitoneally for 7 consecutive days before the SD period. Immunofluorescent staining, western blotting, and RT-PCR were employed to evaluate levels of proteins involved in neuroinflammation and mitochondrial function. Transmission electron microscope and Golgi-Cox staining were used to evaluate mitochondrial and neuronal morphology, respectively. Finally, contextual fear conditioning and the Morris water maze test were conducted to assess hippocampal learning and memory. In the hippocampus of young (3 months-old) and aged (12 months-old) mice subjected to 24 h SD, pretreatment with UA prevented the activation of microglia and astrocytes, NF-κB-NLRP3 signaling and IL-1β, IL6, TNF-α cytokine production, thus ameliorating neuroinflammation. Furthermore, UA also attenuated SD-induced mitochondrial dysfunction, normalized autophagy and mitophagy and protected hippocampal neuronal morphology. Finally, UA prevented SD-induced hippocampal memory impairment. Cumulatively, the results show that UA imparts cognitive protection by reducing neuroinflammation and enhancing mitochondrial function in SD mice. This suggests that UA shows promise as a therapeutic for the treatment of SD-induced neurological disorders.
Collapse
Affiliation(s)
- Afzal Misrani
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, 511400, China
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Sidra Tabassum
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, 511400, China
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Zai-Yong Zhang
- Department of Cardiology, Panyu Central Hospital, Guangzhou, 511400, China
- Cardiovascular Institute of Panyu District, Guangzhou, 511400, China
| | - Shao-Hua Tan
- Department of Neurology, Panyu District Central Hospital, Guangzhou, 511400, China
| | - Cheng Long
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, 511400, China.
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
30
|
Zhao H, Blokland A, Prickaerts J, Havekes R, Heckman PRA. Treatment with the selective PDE4B inhibitor A-33 or PDE4D inhibitor zatolmilast prevents sleep deprivation-induced deficits in spatial pattern separation. Behav Brain Res 2024; 459:114798. [PMID: 38056709 DOI: 10.1016/j.bbr.2023.114798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/22/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023]
Abstract
Sleep deprivation (SD) disrupts hippocampus-dependent memory, particularly in the dentate gyrus (DG) region, an area crucial for pattern separation. Previous research showed that non-selective phosphodiesterase type 4 (PDE4) inhibitors like roflumilast can alleviate these deficits. However, it remains unclear whether these outcomes are specific to a particular subfamily of PDE4. Hence, this study examined the specific impact of PDE4B inhibitor (A-33) and PDE4D inhibitor (zatolmilast) on spatial pattern separation in sleep deprived mice. Results demonstrated that SD impairs pattern separation, but both zatolmilast and A-33 alleviate these effects. However, A-33 impaired pattern separation in non-sleep deprived animals. The cognitive benefits of these inhibitors after SD may arise from alterations in relevant signaling pathways in the DG. This study provides initial evidence that inhibiting PDE4B or PDE4D holds promise for mitigating memory deficits due to SD.
Collapse
Affiliation(s)
- Hongyu Zhao
- Dept. Neuropsychology & Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Arjan Blokland
- Dept. Neuropsychology & Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Jos Prickaerts
- Dept. Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Robbert Havekes
- Neurobiology Expert Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Pim R A Heckman
- Dept. Neuropsychology & Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
31
|
Oliveira MM, Mohamed M, Elder MK, Banegas-Morales K, Mamcarz M, Lu EH, Golhan EAN, Navrange N, Chatterjee S, Abel T, Klann E. The integrated stress response effector GADD34 is repurposed by neurons to promote stimulus-induced translation. Cell Rep 2024; 43:113670. [PMID: 38219147 PMCID: PMC10964249 DOI: 10.1016/j.celrep.2023.113670] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/11/2023] [Accepted: 12/26/2023] [Indexed: 01/16/2024] Open
Abstract
Neuronal protein synthesis is required for long-lasting plasticity and long-term memory consolidation. Dephosphorylation of eukaryotic initiation factor 2α is one of the key translational control events that is required to increase de novo protein synthesis that underlies long-lasting plasticity and memory consolidation. Here, we interrogate the molecular pathways of translational control that are triggered by neuronal stimulation with brain-derived neurotrophic factor (BDNF), which results in eukaryotic initiation factor 2α (eIF2α) dephosphorylation and increases in de novo protein synthesis. Primary rodent neurons exposed to BDNF display elevated translation of GADD34, which facilitates eIF2α dephosphorylation and subsequent de novo protein synthesis. Furthermore, GADD34 requires G-actin generated by cofilin to dephosphorylate eIF2α and enhance protein synthesis. Finally, GADD34 is required for BDNF-induced translation of synaptic plasticity-related proteins. Overall, we provide evidence that neurons repurpose GADD34, an effector of the integrated stress response, as an orchestrator of rapid increases in eIF2-dependent translation in response to plasticity-inducing stimuli.
Collapse
Affiliation(s)
| | - Muhaned Mohamed
- Center for Neural Science, New York University, New York, NY, USA
| | - Megan K Elder
- Center for Neural Science, New York University, New York, NY, USA
| | | | - Maggie Mamcarz
- Center for Neural Science, New York University, New York, NY, USA
| | - Emily H Lu
- Center for Neural Science, New York University, New York, NY, USA
| | - Ela A N Golhan
- Center for Neural Science, New York University, New York, NY, USA
| | - Nishika Navrange
- Center for Neural Science, New York University, New York, NY, USA
| | - Snehajyoti Chatterjee
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY, USA; NYU Neuroscience Institute, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
32
|
Berisha DE, Rizvi B, Chappel-Farley MG, Tustison N, Taylor L, Dave A, Sattari NS, Chen IY, Lui KK, Janecek JC, Keator D, Neikrug AB, Benca RM, Yassa MA, Mander BA. Cerebrovascular pathology mediates associations between hypoxemia during rapid eye movement sleep and medial temporal lobe structure and function in older adults. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.28.577469. [PMID: 38328085 PMCID: PMC10849660 DOI: 10.1101/2024.01.28.577469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Obstructive sleep apnea (OSA) is common in older adults and is associated with medial temporal lobe (MTL) degeneration and memory decline in aging and Alzheimer's disease (AD). However, the underlying mechanisms linking OSA to MTL degeneration and impaired memory remains unclear. By combining magnetic resonance imaging (MRI) assessments of cerebrovascular pathology and MTL structure with clinical polysomnography and assessment of overnight emotional memory retention in older adults at risk for AD, cerebrovascular pathology in fronto-parietal brain regions was shown to statistically mediate the relationship between OSA-related hypoxemia, particularly during rapid eye movement (REM) sleep, and entorhinal cortical thickness. Reduced entorhinal cortical thickness was, in turn, associated with impaired overnight retention in mnemonic discrimination ability across emotional valences for high similarity lures. These findings identify cerebrovascular pathology as a contributing mechanism linking hypoxemia to MTL degeneration and impaired sleep-dependent memory in older adults.
Collapse
Affiliation(s)
- Destiny E. Berisha
- Department of Neurobiology and Behavior, University of California Irvine, Irvine CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine CA, 92697, USA
| | - Batool Rizvi
- Department of Neurobiology and Behavior, University of California Irvine, Irvine CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine CA, 92697, USA
| | - Miranda G. Chappel-Farley
- Department of Neurobiology and Behavior, University of California Irvine, Irvine CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine CA, 92697, USA
| | - Nicholas Tustison
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine CA, 92697, USA
| | - Lisa Taylor
- Department of Neurobiology and Behavior, University of California Irvine, Irvine CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine CA, 92697, USA
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine CA, 92697, USA
| | - Abhishek Dave
- Department of Cognitive Sciences, University of California Irvine, Irvine CA, 92697, USA
| | - Negin S. Sattari
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine CA, 92697, USA
| | - Ivy Y. Chen
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine CA, 92697, USA
| | - Kitty K. Lui
- San Diego State University/University of California San Diego, Joint Doctoral Program in Clinical Psychology, San Diego, CA, 92093, USA
| | - John C. Janecek
- Department of Neurobiology and Behavior, University of California Irvine, Irvine CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine CA, 92697, USA
| | - David Keator
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine CA, 92697, USA
| | - Ariel B. Neikrug
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine CA, 92697, USA
| | - Ruth M. Benca
- Department of Neurobiology and Behavior, University of California Irvine, Irvine CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine CA, 92697, USA
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine CA, 92697, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Department of Psychiatry, University of Wisconsin-Madison, Madison, 53706, WI, USA
- Department of Psychiatry and Behavioral Medicine, Wake Forest University, Winston-Salem, NC, 27109, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine CA, 92697, USA
| | - Michael A. Yassa
- Department of Neurobiology and Behavior, University of California Irvine, Irvine CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine CA, 92697, USA
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine CA, 92697, USA
- Department of Neurology, University of California Irvine, Irvine CA, 92697, USA
| | - Bryce A. Mander
- Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine CA, 92697, USA
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine CA, 92697, USA
- Department of Cognitive Sciences, University of California Irvine, Irvine CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine CA, 92697, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine CA, 92697, USA
| |
Collapse
|
33
|
Picard K, Dolhan K, Watters JJ, Tremblay MÈ. Microglia and Sleep Disorders. ADVANCES IN NEUROBIOLOGY 2024; 37:357-377. [PMID: 39207702 DOI: 10.1007/978-3-031-55529-9_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Sleep is a physiological state that is essential for maintaining physical and mental health. Sleep disorders and sleep deprivation therefore have many adverse effects, including an increased risk of metabolic diseases and a decline in cognitive function that may be implicated in the long-term development of neurodegenerative diseases. There is increasing evidence that microglia, the resident immune cells of the central nervous system (CNS), are involved in regulating the sleep-wake cycle and the CNS response to sleep alteration and deprivation. In this chapter, we will discuss the involvement of microglia in various sleep disorders, including sleep-disordered breathing, insomnia, narcolepsy, myalgic encephalomyelitis/chronic fatigue syndrome, and idiopathic rapid-eye-movement sleep behavior disorder. We will also explore the impact of acute and chronic sleep deprivation on microglial functions. Moreover, we will look into the potential involvement of microglia in sleep disorders as a comorbidity to Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Katherine Picard
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Kira Dolhan
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Department of Psychology, University of Victoria, Victoria, BC, Canada
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | - Jyoti J Watters
- Department of Comparative Biosciences, University of Wisconsin Madison, Madison, WI, USA
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada.
- Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
34
|
Gao H, Zhang Y, Luo D, Xu J, Tan S, Li Y, Qi W, Zhai Q, Wang Q. Activation of the Hippocampal DRD2 Alleviates Neuroinflammation, Synaptic Plasticity Damage and Cognitive Impairment After Sleep Deprivation. Mol Neurobiol 2023; 60:7208-7221. [PMID: 37543530 DOI: 10.1007/s12035-023-03514-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/15/2023] [Indexed: 08/07/2023]
Abstract
Sleep loss is commonplace nowadays and profoundly impacts cognition. Dopamine receptor D2 (DRD2) makes a specific contribution to cognition, although the precise mechanism underlying how DRD2 affects the cognitive process after sleep deprivation remains unclear. Herein, we observed cognitive impairment and impaired synaptic plasticity, including downregulation of synaptophysin and PSD95, decreased postsynaptic density thickness, neuron complexity, and spine density in chronic sleep restriction (CSR) mice. We also observed downregulated hippocampal DRD2 and Cryab expression in the CSR mice. Meanwhile, NF-κB translocation from the cytoplasm to the nucleus occurred, indicating that neuroinflammation ensued. However, hippocampal delivery of the DRD2 agonist quinpirole effectively rescued these changes. In vitro, quinpirole treatment significantly decreased the release of proinflammatory cytokines in microglial supernatant, indicating a potential anti-neuroinflammatory effect of Drd2/Cryab/NF-κB in CSR mice. Our study provided the evidence that activation of the Drd2 may relieve neuroinflammation and improve sleep deprivation-induced cognitive deficits.
Collapse
Affiliation(s)
- Hui Gao
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Department of Anaesthesiology, Yan'an University Affiliated Hospital, Yan'an, 716000, China
| | - Yuxin Zhang
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200434, China
| | - Danlei Luo
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jing Xu
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shuwen Tan
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ying Li
- Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Wanling Qi
- Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Qian Zhai
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Qiang Wang
- Department of Anaesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
35
|
Ripoli C, Dagliyan O, Renna P, Pastore F, Paciello F, Sollazzo R, Rinaudo M, Battistoni M, Martini S, Tramutola A, Sattin A, Barone E, Saneyoshi T, Fellin T, Hayashi Y, Grassi C. Engineering memory with an extrinsically disordered kinase. SCIENCE ADVANCES 2023; 9:eadh1110. [PMID: 37967196 PMCID: PMC10651130 DOI: 10.1126/sciadv.adh1110] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 10/13/2023] [Indexed: 11/17/2023]
Abstract
Synaptic plasticity plays a crucial role in memory formation by regulating the communication between neurons. Although actin polymerization has been linked to synaptic plasticity and dendritic spine stability, the causal link between actin polymerization and memory encoding has not been identified yet. It is not clear whether actin polymerization and structural changes in dendritic spines are a driver or a consequence of learning and memory. Using an extrinsically disordered form of the protein kinase LIMK1, which rapidly and precisely acts on ADF/cofilin, a direct modifier of actin, we induced long-term enlargement of dendritic spines and enhancement of synaptic transmission in the hippocampus on command. The activation of extrinsically disordered LIMK1 in vivo improved memory encoding and slowed cognitive decline in aged mice exhibiting reduced cofilin phosphorylation. The engineered memory by an extrinsically disordered LIMK1 supports a direct causal link between actin-mediated synaptic transmission and memory.
Collapse
Affiliation(s)
- Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Onur Dagliyan
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17165 Stockholm, Sweden
| | - Pietro Renna
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Pastore
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Raimondo Sollazzo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marco Rinaudo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Martina Battistoni
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Sara Martini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185 Rome, Italy
| | - Andrea Sattin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185 Rome, Italy
| | - Takeo Saneyoshi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Tommaso Fellin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Yasunori Hayashi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
36
|
Sengupta A, Tudor JC, Cusmano D, Baur JA, Abel T, Weljie AM. Sleep deprivation and aging are metabolically linked across tissues. Sleep 2023; 46:zsad246. [PMID: 37738102 PMCID: PMC11502955 DOI: 10.1093/sleep/zsad246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/21/2023] [Indexed: 09/24/2023] Open
Abstract
STUDY OBJECTIVES Insufficient sleep is a concerning hallmark of modern society because sleep deprivation (SD) is a risk factor for neurodegenerative and cardiometabolic disorders. SD imparts an aging-like effect on learning and memory, although little is known about possible common molecular underpinnings of SD and aging. Here, we examine this question by profiling metabolic features across different tissues after acute SD in young adult and aged mice. METHODS Young adult and aged mice were subjected to acute SD for 5 hours. Blood plasma, hippocampus, and liver samples were subjected to UPLC-MS/MS-based metabolic profiling. RESULTS SD preferentially impacts peripheral plasma and liver profiles (e.g. ketone body metabolism) whereas the hippocampus is more impacted by aging. We further demonstrate that aged animals exhibit SD-like metabolic features at baseline. Hepatic alterations include parallel changes in nicotinamide metabolism between aging and SD in young animals. Overall, metabolism in young adult animals is more impacted by SD, which in turn induces aging-like features. A set of nine metabolites was classified (79% correct) based on age and sleep status across all four groups. CONCLUSIONS Our metabolic observations demonstrate striking parallels to previous observations in studies of learning and memory and define a molecular metabolic signature of sleep loss and aging.
Collapse
Affiliation(s)
- Arjun Sengupta
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer C Tudor
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
- Current affiliation: Department of Biology, Saint Joseph’s University, Philadelphia, PA, USA
| | - Danielle Cusmano
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph A Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ted Abel
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
- Current Affiliation: Iowa Neuroscience Institute, Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 2312 PBDB, Iowa City, IA, USA
| | - Aalim M Weljie
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
37
|
Vanrobaeys Y, Peterson ZJ, Walsh EN, Chatterjee S, Lin LC, Lyons LC, Nickl-Jockschat T, Abel T. Spatial transcriptomics reveals unique gene expression changes in different brain regions after sleep deprivation. Nat Commun 2023; 14:7095. [PMID: 37925446 PMCID: PMC10625558 DOI: 10.1038/s41467-023-42751-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 10/20/2023] [Indexed: 11/06/2023] Open
Abstract
Sleep deprivation has far-reaching consequences on the brain and behavior, impacting memory, attention, and metabolism. Previous research has focused on gene expression changes in individual brain regions, such as the hippocampus or cortex. Therefore, it is unclear how uniformly or heterogeneously sleep loss affects the brain. Here, we use spatial transcriptomics to define the impact of a brief period of sleep deprivation across the brain in male mice. We find that sleep deprivation induced pronounced differences in gene expression across the brain, with the greatest changes in the hippocampus, neocortex, hypothalamus, and thalamus. Both the differentially expressed genes and the direction of regulation differed markedly across regions. Importantly, we developed bioinformatic tools to register tissue sections and gene expression data into a common anatomical space, allowing a brain-wide comparison of gene expression patterns between samples. Our results suggest that distinct molecular mechanisms acting in discrete brain regions underlie the biological effects of sleep deprivation.
Collapse
Affiliation(s)
- Yann Vanrobaeys
- Interdisciplinary Graduate Program in Genetics, University of Iowa, 357 Medical Research Center Iowa City, Iowa, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA, USA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 51 Newton Road, 2-417B Bowen Science Building, Iowa City, IA, USA
| | - Zeru J Peterson
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Emily N Walsh
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA, USA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 51 Newton Road, 2-417B Bowen Science Building, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, 356 Medical Research Center, Iowa City, IA, USA
| | - Snehajyoti Chatterjee
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA, USA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 51 Newton Road, 2-417B Bowen Science Building, Iowa City, IA, USA
| | - Li-Chun Lin
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA, USA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 51 Newton Road, 2-417B Bowen Science Building, Iowa City, IA, USA
- Department of Neurology, University of Iowa, Iowa City, IA, USA
| | - Lisa C Lyons
- Program in Neuroscience, Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Thomas Nickl-Jockschat
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA, USA.
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 51 Newton Road, 2-417B Bowen Science Building, Iowa City, IA, USA.
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA.
| | - Ted Abel
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA, USA.
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 51 Newton Road, 2-417B Bowen Science Building, Iowa City, IA, USA.
| |
Collapse
|
38
|
Blokland A, Jackson M, Puustinen K, Soeterboek J, Heckman PRA. Acute sleep loss impairs object but not spatial pattern separation in humans. Neurosci Lett 2023; 818:137535. [PMID: 39491126 DOI: 10.1016/j.neulet.2023.137535] [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: 06/21/2023] [Revised: 10/12/2023] [Accepted: 10/23/2023] [Indexed: 11/05/2024]
Abstract
Pattern separation allows us to form discrete representations of information in memory. Pattern separation can be measured in several domains including spatial and object-based discrimination. The brain area largely involved in this process is the dentate gyrus of the hippocampus, which has been shown to be particularly sensitive to the effects of sleep loss. However, methodology in rodent and human studies varies greatly making translational conclusions difficult. Therefore, the aim of the current study was to measure the effects of sleep deprivation on human hippocampal function, using well-validated spatial and object-based pattern separation tests. The effects of acute sleep loss were examined, as this method is frequently used in rodent research but not human studies. Results show that sleep loss impaired performance on the object-based version of the test, but not spatial pattern separation. The findings support the notion that these discrimination projections represent separate but complimentary hippocampal processes, and further elucidates how they may be discretely affected by acute sleep loss.
Collapse
Affiliation(s)
- Arjan Blokland
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Meyra Jackson
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Kia Puustinen
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, The Netherlands; REVAL Rehabilitation Research Center, Hasselt University, Hasselt, Belgium; Department of Movement Sciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jens Soeterboek
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, The Netherlands; Alzheimer Centrum Limburg, School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Pim R A Heckman
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
39
|
Pagano R, Salamian A, Skonieczna E, Wojtas B, Gielniewski B, Harda Z, Cały A, Havekes R, Abel T, Radwanska K. Molecular fingerprints in the hippocampus of alcohol seeking during withdrawal. RESEARCH SQUARE 2023:rs.3.rs-3337670. [PMID: 37841864 PMCID: PMC10571638 DOI: 10.21203/rs.3.rs-3337670/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Alcohol use disorder (AUD) is characterized by pathological motivation to consume alcohol and cognitive inflexibility, leading to excessive alcohol seeking and use. Due to limited understanding of the molecular basis of the disease, there are few pharmacological interventions available to combat AUD. In this study, we aimed to investigate the molecular correlates of impaired extinction of alcohol seeking during alcohol withdrawal using a mouse model of AUD implemented in the automated IntelliCage social system. This model enabled us to distinguish between animals exhibiting AUD-prone and AUD-resistant phenotypes, based on the presence of ≥ 2 or < 2 criteria of AUD, respectively. We utilized new generation RNA sequencing to identify genes that were differentially expressed in the hippocampus and amygdala of mice meeting ≥ 2 or < 2 criteria, as these brain regions are implicated in alcohol motivation, seeking, consumption and the cognitive inflexibility characteristic of AUD. To complement the sequencing studies, we conducted ex vivo electrophysiology experiments. Our findings revealed significant dysregulation of the hippocampal genes associated with the actin cytoskeleton and synaptic function, including actin binding molecule cofilin, during alcohol withdrawal in mice meeting ≥ 2 criteria compared to those meeting < 2 criteria. Moreover, this dysregulation was accompanied by impaired synaptic transmission in the molecular layer of the hippocampal dentate gyrus (ML-DG). Additionally, we demonstrated that overexpression of cofilin in the polymorphic layer of the hippocampal dentate gyrus (PoDG) inhibited ML-DG synapses, increased motivation to seek alcohol, impaired extinction of alcohol seeking and increased correlation between AUD behaviors, resembling the phenotype observed in mice meeting ≥ 2 criteria. Overall, our study uncovers a novel mechanism linking increased hippocampal cofilin expression with the AUD phenotype.
Collapse
Affiliation(s)
- Roberto Pagano
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Ahmad Salamian
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Edyta Skonieczna
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Bartosz Wojtas
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Bartek Gielniewski
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Zofia Harda
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
- current address: Department Molecular Neuropharmacology, Maj Institute of Pharmacology of Polish Academy of Sciences, Krakow, Poland
| | - Anna Cały
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Robbert Havekes
- Neurobiology expertise group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Ted Abel
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kasia Radwanska
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| |
Collapse
|
40
|
Pagano R, Salamian A, Skonieczna E, Wojtas B, Gielniewski B, Harda Z, Cały A, Havekes R, Abel T, Radwanska K. Molecular fingerprints in the hippocampus of alcohol seeking during withdrawal. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554622. [PMID: 37662388 PMCID: PMC10473700 DOI: 10.1101/2023.08.24.554622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Alcohol use disorder (AUD) is characterized by excessive alcohol seeking and use. Here, we investigated the molecular correlates of impaired extinction of alcohol seeking using a multidimentional mouse model of AUD. We distinguished AUD-prone and AUD-resistant mice, based on the presence of ≥ 2 or < 2 criteria of AUD and utilized RNA sequencing to identify genes that were differentially expressed in the hippocampus and amygdala of mice meeting ≥ 2 or < 2 criteria, as these brain regions are implicated in alcohol motivation, seeking, consumption and the cognitive inflexibility characteristic of AUD. Our findings revealed dysregulation of the genes associated with the actin cytoskeleton, including actin binding molecule cofilin, and impaired synaptic transmission in the hippocampi of mice meeting ≥ 2 criteria. Overexpression of cofilin in the polymorphic layer of the dentate gyrus (PoDG) inhibited ML-DG synapses, increased motivation to seek alcohol and impaired extinction of alcohol seeking, resembling the phenotype observed in mice meeting ≥ 2 criteria. Overall, our study uncovers a novel mechanism linking increased hippocampal cofilin expression with the AUD phenotype.
Collapse
Affiliation(s)
- Roberto Pagano
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Ahmad Salamian
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Edyta Skonieczna
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Bartosz Wojtas
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Bartek Gielniewski
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Zofia Harda
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
- current address: Department Molecular Neuropharmacology, Maj Institute of Pharmacology of Polish Academy of Sciences, Krakow, Poland
| | - Anna Cały
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| | - Robbert Havekes
- Neurobiology expertise group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Ted Abel
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kasia Radwanska
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., Warsaw 02-093, Poland
| |
Collapse
|
41
|
Zaki Y, Pennington ZT, Morales-Rodriguez D, Francisco TR, LaBanca AR, Dong Z, Lamsifer S, Segura SC, Chen HT, Wick ZC, Silva AJ, van der Meer M, Shuman T, Fenton A, Rajan K, Cai DJ. Aversive experience drives offline ensemble reactivation to link memories across days. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.13.532469. [PMID: 36993254 PMCID: PMC10054942 DOI: 10.1101/2023.03.13.532469] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Memories are encoded in neural ensembles during learning and stabilized by post-learning reactivation. Integrating recent experiences into existing memories ensures that memories contain the most recently available information, but how the brain accomplishes this critical process remains unknown. Here we show that in mice, a strong aversive experience drives the offline ensemble reactivation of not only the recent aversive memory but also a neutral memory formed two days prior, linking the fear from the recent aversive memory to the previous neutral memory. We find that fear specifically links retrospectively, but not prospectively, to neutral memories across days. Consistent with prior studies, we find reactivation of the recent aversive memory ensemble during the offline period following learning. However, a strong aversive experience also increases co-reactivation of the aversive and neutral memory ensembles during the offline period. Finally, the expression of fear in the neutral context is associated with reactivation of the shared ensemble between the aversive and neutral memories. Taken together, these results demonstrate that strong aversive experience can drive retrospective memory-linking through the offline co-reactivation of recent memory ensembles with memory ensembles formed days prior, providing a neural mechanism by which memories can be integrated across days.
Collapse
Affiliation(s)
- Yosif Zaki
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Zachary T. Pennington
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | | | - Taylor R. Francisco
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Alexa R. LaBanca
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Zhe Dong
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Sophia Lamsifer
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Simón Carrillo Segura
- Graduate Program in Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, NY, 11201
| | - Hung-Tu Chen
- Department of Psychological & Brain Sciences, Dartmouth College, Hanover, NH, 03755
| | - Zoé Christenson Wick
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Alcino J. Silva
- Department of Neurobiology, Psychiatry & Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, Brain Research Institute, UCLA, Los Angeles, CA 90095
| | | | - Tristan Shuman
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - André Fenton
- Center for Neural Science, New York University, New York, NY, 10003
- Neuroscience Institute at the NYU Langone Medical Center, New York, NY, 10016
| | - Kanaka Rajan
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Denise J. Cai
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| |
Collapse
|
42
|
Raven F, Riemersma IW, Olthuis MF, Rybakovaite I, Meijer EL, Meerlo P, Van der Zee EA, Havekes R. Cofilin overactivation improves hippocampus-dependent short-term memory. Front Behav Neurosci 2023; 17:1243524. [PMID: 37638111 PMCID: PMC10448394 DOI: 10.3389/fnbeh.2023.1243524] [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: 06/20/2023] [Accepted: 07/28/2023] [Indexed: 08/29/2023] Open
Abstract
Many living organisms of the animal kingdom have the fundamental ability to form and retrieve memories. Most information is initially stored as short-term memory, which is then converted to a more stable long-term memory through a process called memory consolidation. At the neuronal level, synaptic plasticity is crucial for memory storage. It includes the formation of new spines, as well as the modification of existing spines, thereby tuning and shaping synaptic efficacy. Cofilin critically contributes to memory processes as upon activation, it regulates the shape of dendritic spines by targeting actin filaments. We previously found that prolonged activation of cofilin in hippocampal neurons attenuated the formation of long-term object-location memories. Because the modification of spine shape and structure is also essential for short-term memory formation, we determined whether overactivation of hippocampal cofilin also influences the formation of short-term memories. To this end, mice were either injected with an adeno-associated virus expressing catalytically active cofilin, or an eGFP control, in the hippocampus. We show for the first time that cofilin overactivation improves short-term memory formation in the object-location memory task, without affecting anxiety-like behavior. Surprisingly, we found no effect of cofilin overactivation on AMPA receptor expression levels. Altogether, while cofilin overactivation might negatively impact the formation of long-lasting memories, it may benefit short-term plasticity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Robbert Havekes
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, Netherlands
| |
Collapse
|
43
|
Martinez JD, Wilson LG, Brancaleone WP, Peterson KG, Popke DS, Garzon VC, Perez Tremble RE, Donnelly MJ, Mendez Ortega SL, Torres D, Shaver JJ, Clawson BC, Jiang S, Yang Z, Aton SJ. Hypnotic treatment reverses NREM sleep disruption and EEG desynchronization in a mouse model of Fragile X syndrome to rescue memory consolidation deficits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.14.549070. [PMID: 37502832 PMCID: PMC10370139 DOI: 10.1101/2023.07.14.549070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Fragile X syndrome (FXS) is a highly-prevalent genetic cause of intellectual disability, associated with disrupted cognition and sleep abnormalities. Sleep loss itself negatively impacts cognitive function, yet the contribution of sleep loss to impaired cognition in FXS is vastly understudied. One untested possibility is that disrupted cognition in FXS is exacerbated by abnormal sleep. We hypothesized that restoration of sleep-dependent mechanisms could improve functions such as memory consolidation in FXS. We examined whether administration of ML297, a hypnotic drug acting on G-protein-activated inward-rectifying potassium channels, could restore sleep phenotypes and improve disrupted memory consolidation in Fmr1 -/y mice. Using 24-h polysomnographic recordings, we found that Fmr1 -/y mice exhibit reduced non-rapid eye movement (NREM) sleep and fragmented NREM sleep architecture, alterations in NREM EEG spectral power (including reductions in sleep spindles), and reduced EEG coherence between cortical areas. These alterations were reversed in the hours following ML297 administration. Hypnotic treatment following contextual fear or spatial learning also ameliorated disrupted memory consolidation in Fmr1 -/y mice. Hippocampal activation patterns during memory recall was altered in Fmr1 -/y mice, reflecting an altered balance of activity among principal neurons vs. parvalbumin-expressing (PV+) interneurons. This phenotype was partially reversed by post-learning ML297 administration. These studies suggest that sleep disruption could have a major impact on neurophysiological and behavioral phenotypes in FXS, and that hypnotic therapy may significantly improve disrupted cognition in this disorder.
Collapse
|
44
|
Lyons LC, Vanrobaeys Y, Abel T. Sleep and memory: The impact of sleep deprivation on transcription, translational control, and protein synthesis in the brain. J Neurochem 2023; 166:24-46. [PMID: 36802068 PMCID: PMC10919414 DOI: 10.1111/jnc.15787] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/20/2023]
Abstract
In countries around the world, sleep deprivation represents a widespread problem affecting school-age children, teenagers, and adults. Acute sleep deprivation and more chronic sleep restriction adversely affect individual health, impairing memory and cognitive performance as well as increasing the risk and progression of numerous diseases. In mammals, the hippocampus and hippocampus-dependent memory are vulnerable to the effects of acute sleep deprivation. Sleep deprivation induces changes in molecular signaling, gene expression and may cause changes in dendritic structure in neurons. Genome wide studies have shown that acute sleep deprivation alters gene transcription, although the pool of genes affected varies between brain regions. More recently, advances in research have drawn attention to differences in gene regulation between the level of the transcriptome compared with the pool of mRNA associated with ribosomes for protein translation following sleep deprivation. Thus, in addition to transcriptional changes, sleep deprivation also affects downstream processes to alter protein translation. In this review, we focus on the multiple levels through which acute sleep deprivation impacts gene regulation, highlighting potential post-transcriptional and translational processes that may be affected by sleep deprivation. Understanding the multiple levels of gene regulation impacted by sleep deprivation is essential for future development of therapeutics that may mitigate the effects of sleep loss.
Collapse
Affiliation(s)
- Lisa C Lyons
- Program in Neuroscience, Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Yann Vanrobaeys
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, Iowa City, Iowa, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, Iowa City, Iowa, USA
| |
Collapse
|
45
|
Wang L, Ling H, He H, Hu N, Xiao L, Zhang Y, Xie L, You Z. Dysfunctional synaptic pruning by microglia correlates with cognitive impairment in sleep-deprived mice: Involvement of CX3CR1 signaling. Neurobiol Stress 2023; 25:100553. [PMID: 37547773 PMCID: PMC10401339 DOI: 10.1016/j.ynstr.2023.100553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/08/2023] [Accepted: 06/23/2023] [Indexed: 08/08/2023] Open
Abstract
Microglia are involved in sleep/wake cycles and the response to sleep loss. Synaptic pruning by microglia is necessary for central nervous system circuit refinement and contributes to cognitive function. Here, we investigated whether and how microglia-mediated synaptic pruning may be involved in cognitive deficits induced by sleep deprivation in mice. Mice were deprived of sleep by leaving them in a spontaneously rotating rod for 72 h, after which their cognitive function was assessed using an object location test, Y maze, and novel object recognition test. Sleep deprivation lowered the discrimination index for familiar locations in the object location test and Y maze. Microglial morphology was assessed using immunostaining Iba1, while microglia-mediated synaptic pruning was examined based on immunostaining PSD95, CD68, and Iba1. Sleep deprivation also activated microglial cells in the hippocampus, as reflected in bigger soma as well as fewer and shorter branches than normal sleep. Sleep deprivation downregulated phagocytic markers and internalization of postsynaptic protein 95 (PSD95), suggesting impaired synaptic pruning. CX3C motif chemokine receptor 1 (CX3CR1) signaling was detected in in vitro experiments. Sleep deprivation also downregulated CX3CR1. Activation of CX3CR1 signaling increased phagocytosis activity of BV2 microglia in vitro. Sleep deprivation dysregulates microglial CX3CR1 signaling and inhibits synaptic pruning, contributing to associated cognitive deficits. These findings identify CX3CR1-dependent synaptic pruning as a potential therapeutic target in which sleep deprivation causes recognition impairments.
Collapse
Affiliation(s)
- Lu Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, 610054, China
- School of Life Science and Technology, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Hanyi Ling
- School of Life Science and Technology, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Hui He
- School of Life Science and Technology, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Nan Hu
- School of Life Science and Technology, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Lin Xiao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Yue Zhang
- School of Life Science and Technology, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Lei Xie
- School of Life Science and Technology, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Zili You
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, 610054, China
- School of Life Science and Technology, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| |
Collapse
|
46
|
Gerstner JR, Flores CC, Lefton M, Rogers B, Davis CJ. FABP7: a glial integrator of sleep, circadian rhythms, plasticity, and metabolic function. Front Syst Neurosci 2023; 17:1212213. [PMID: 37404868 PMCID: PMC10315501 DOI: 10.3389/fnsys.2023.1212213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/02/2023] [Indexed: 07/06/2023] Open
Abstract
Sleep and circadian rhythms are observed broadly throughout animal phyla and influence neural plasticity and cognitive function. However, the few phylogenetically conserved cellular and molecular pathways that are implicated in these processes are largely focused on neuronal cells. Research on these topics has traditionally segregated sleep homeostatic behavior from circadian rest-activity rhythms. Here we posit an alternative perspective, whereby mechanisms underlying the integration of sleep and circadian rhythms that affect behavioral state, plasticity, and cognition reside within glial cells. The brain-type fatty acid binding protein, FABP7, is part of a larger family of lipid chaperone proteins that regulate the subcellular trafficking of fatty acids for a wide range of cellular functions, including gene expression, growth, survival, inflammation, and metabolism. FABP7 is enriched in glial cells of the central nervous system and has been shown to be a clock-controlled gene implicated in sleep/wake regulation and cognitive processing. FABP7 is known to affect gene transcription, cellular outgrowth, and its subcellular localization in the fine perisynaptic astrocytic processes (PAPs) varies based on time-of-day. Future studies determining the effects of FABP7 on behavioral state- and circadian-dependent plasticity and cognitive processes, in addition to functional consequences on cellular and molecular mechanisms related to neural-glial interactions, lipid storage, and blood brain barrier integrity will be important for our knowledge of basic sleep function. Given the comorbidity of sleep disturbance with neurological disorders, these studies will also be important for our understanding of the etiology and pathophysiology of how these diseases affect or are affected by sleep.
Collapse
Affiliation(s)
- Jason R. Gerstner
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Carlos C. Flores
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Micah Lefton
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Brooke Rogers
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Christopher J. Davis
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| |
Collapse
|
47
|
Chen P, Ban W, Wang W, You Y, Yang Z. The Devastating Effects of Sleep Deprivation on Memory: Lessons from Rodent Models. Clocks Sleep 2023; 5:276-294. [PMID: 37218868 DOI: 10.3390/clockssleep5020022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 05/24/2023] Open
Abstract
In this narrative review article, we discuss the role of sleep deprivation (SD) in memory processing in rodent models. Numerous studies have examined the effects of SD on memory, with the majority showing that sleep disorders negatively affect memory. Currently, a consensus has not been established on which damage mechanism is the most appropriate. This critical issue in the neuroscience of sleep remains largely unknown. This review article aims to elucidate the mechanisms that underlie the damaging effects of SD on memory. It also proposes a scientific solution that might explain some findings. We have chosen to summarize literature that is both representative and comprehensive, as well as innovative in its approach. We examined the effects of SD on memory, including synaptic plasticity, neuritis, oxidative stress, and neurotransmitters. Results provide valuable insights into the mechanisms by which SD impairs memory function.
Collapse
Affiliation(s)
- Pinqiu Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
| | - Weikang Ban
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Wenyan Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
| | - Yuyang You
- School of Automation, Beijing Institute of Technology, Beijing 100081, China
| | - Zhihong Yang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| |
Collapse
|
48
|
Rexrode L, Tennin M, Babu J, Young C, Bollavarapu R, Lawson LA, Valeri J, Pantazopoulos H, Gisabella B. Regulation of dendritic spines in the amygdala following sleep deprivation. FRONTIERS IN SLEEP 2023; 2:1145203. [PMID: 37928499 PMCID: PMC10624159 DOI: 10.3389/frsle.2023.1145203] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
The amygdala is a hub of emotional circuits involved in the regulation of cognitive and emotional behaviors and its critically involved in emotional reactivity, stress regulation, and fear memory. Growing evidence suggests that the amygdala plays a key role in the consolidation of emotional memories during sleep. Neuroimaging studies demonstrated that the amygdala is selectively and highly activated during rapid eye movement sleep (REM) and sleep deprivation induces emotional instability and dysregulation of the emotional learning process. Regulation of dendritic spines during sleep represents a morphological correlate of memory consolidation. Several studies indicate that dendritic spines are remodeled during sleep, with evidence for broad synaptic downscaling and selective synaptic upscaling in several cortical areas and the hippocampus. Currently, there is a lack of information regarding the regulation of dendritic spines in the amygdala during sleep. In the present work, we investigated the effect of 5 h of sleep deprivation on dendritic spines in the mouse amygdala. Our data demonstrate that sleep deprivation results in differential dendritic spine changes depending on both the amygdala subregions and the morphological subtypes of dendritic spines. We observed decreased density of mushroom spines in the basolateral amygdala of sleep deprived mice, together with increased neck length and decreased surface area and volume. In contrast, we observed greater densities of stubby spines in sleep deprived mice in the central amygdala, indicating that downscaling selectively occurs in this spine type. Greater neck diameters for thin spines in the lateral and basolateral nuclei of sleep deprived mice, and decreases in surface area and volume for mushroom spines in the basolateral amygdala compared to increases in the cental amygdala provide further support for spine type-selective synaptic downscaling in these areas during sleep. Our findings suggest that sleep promotes synaptic upscaling of mushroom spines in the basolateral amygdala, and downscaling of selective spine types in the lateral and central amygdala. In addition, we observed decreased density of phosphorylated cofilin immunoreactive and growth hormone immunoreactive cells in the amygdala of sleep deprived mice, providing further support for upscaling of dendritic spines during sleep. Overall, our findings point to region-and spine type-specific changes in dendritic spines during sleep in the amygdala, which may contribute to consolidation of emotional memories during sleep.
Collapse
Affiliation(s)
- Lindsay Rexrode
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
| | - Matthew Tennin
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jobin Babu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Caleb Young
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
| | - Ratna Bollavarapu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
| | - Lamiorkor Ameley Lawson
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jake Valeri
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
49
|
Zhang F, Niu L, Zhong R, Li S, Le W. Chronic Sleep Disturbances Alters Sleep Structure and Tau Phosphorylation in AβPP/PS1 AD Mice and Their Wild-Type Littermates. J Alzheimers Dis 2023; 92:1341-1355. [PMID: 37038814 DOI: 10.3233/jad-221048] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
Background: Emerging evidence indicates that sleep disorders are the common non-cognitive symptoms of Alzheimer’s disease (AD), and they may contribute to the pathogenesis of this disease. Objective: In this study, we aim to investigate the effect of chronic sleep deprivation (CSD) on AD-related pathologies with a focus on tau phosphorylation and the underlying DNA methylation regulation. Methods: AβPPswe/PS1ΔE9 AD mice and their wild-type (WT) littermates were subjected to a two-month CSD followed by electroencephalography and electromyography recording. The mice were examined for learning and memory evaluation, then pathological, biochemical, and epigenetic assessments including western blotting, immunofluorescence, dot blotting, and bisulfite sequencing. Results: The results show that CSD caused sleep disorders shown as sleep pattern change, poor sleep maintenance, and increased sleep fragmentation. CSD increased tau phosphorylation at different sites and increased the level of tau kinases in AD and WT mice. The increased expression of cyclin-dependent kinase 5 (CDK5) may result from decreased DNA methylation of CpG sites in the promoter region of CDK5 gene, which might be associated with the downregulation of DNA methyltransferase 3A and 3B. Conclusion: CSD altered AD-related tau phosphorylation through epigenetic modification of tau kinase gene. The findings in this study may give insights into the mechanisms underlying the effects of sleep disorders on AD pathology and provide new therapeutic targets for the treatment of this disease.
Collapse
Affiliation(s)
- Feng Zhang
- Center for Clinical and Translational Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Long Niu
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Rujia Zhong
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Song Li
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Weidong Le
- Center for Clinical and Translational Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
50
|
Cyclin Y regulates spatial learning and memory flexibility through distinct control of the actin pathway. Mol Psychiatry 2023; 28:1351-1364. [PMID: 36434054 PMCID: PMC10005959 DOI: 10.1038/s41380-022-01877-0] [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: 02/08/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 11/27/2022]
Abstract
Spatial learning and memory flexibility are known to require long-term potentiation (LTP) and long-term depression (LTD), respectively, on a cellular basis. We previously showed that cyclin Y (CCNY), a synapse-remodeling cyclin, is a novel actin-binding protein and an inhibitory regulator of functional and structural LTP in vitro. In this study, we report that Ccny knockout (KO) mice exhibit enhanced LTP and weak LTD at Schaffer collateral-CA1 synapses in the hippocampus. In accordance with enhanced LTP, Ccny KO mice showed improved spatial learning and memory. However, although previous studies reported that normal LTD is necessary for memory flexibility, Ccny KO mice intriguingly showed improved memory flexibility, suggesting that weak LTD could exert memory flexibility when combined with enhanced LTP. At the molecular level, CCNY modulated spatial learning and memory flexibility by distinctively affecting the cofilin-actin signaling pathway in the hippocampus. Specifically, CCNY inhibited cofilin activation by original learning, but reversed such inhibition by reversal learning. Furthermore, viral-mediated overexpression of a phosphomimetic cofilin-S3E in hippocampal CA1 regions enhanced LTP, weakened LTD, and improved spatial learning and memory flexibility, thus mirroring the phenotype of Ccny KO mice. In contrast, the overexpression of a non-phosphorylatable cofilin-S3A in hippocampal CA1 regions of Ccny KO mice reversed the synaptic plasticity, spatial learning, and memory flexibility phenotypes observed in Ccny KO mice. Altogether, our findings demonstrate that LTP and LTD cooperatively regulate memory flexibility. Moreover, CCNY suppresses LTP while facilitating LTD in the hippocampus and negatively regulates spatial learning and memory flexibility through the control of cofilin-actin signaling, proposing CCNY as a learning regulator modulating both memorizing and forgetting processes.
Collapse
|