Decoding the nexus: branched-chain amino acids and their connection with sleep, circadian rhythms, and cardiometabolic health.

IF 5.9 2区 医学 Q2 CELL BIOLOGY
Neural Regeneration Research Pub Date : 2025-05-01 Epub Date: 2024-06-03 DOI:10.4103/NRR.NRR-D-23-02020
Hui Li, Laurent Seugnet
{"title":"Decoding the nexus: branched-chain amino acids and their connection with sleep, circadian rhythms, and cardiometabolic health.","authors":"Hui Li, Laurent Seugnet","doi":"10.4103/NRR.NRR-D-23-02020","DOIUrl":null,"url":null,"abstract":"<p><p>The sleep-wake cycle stands as an integrative process essential for sustaining optimal brain function and, either directly or indirectly, overall body health, encompassing metabolic and cardiovascular well-being. Given the heightened metabolic activity of the brain, there exists a considerable demand for nutrients in comparison to other organs. Among these, the branched-chain amino acids, comprising leucine, isoleucine, and valine, display distinctive significance, from their contribution to protein structure to their involvement in overall metabolism, especially in cerebral processes. Among the first amino acids that are released into circulation post-food intake, branched-chain amino acids assume a pivotal role in the regulation of protein synthesis, modulating insulin secretion and the amino acid sensing pathway of target of rapamycin. Branched-chain amino acids are key players in influencing the brain's uptake of monoamine precursors, competing for a shared transporter. Beyond their involvement in protein synthesis, these amino acids contribute to the metabolic cycles of γ-aminobutyric acid and glutamate, as well as energy metabolism. Notably, they impact GABAergic neurons and the excitation/inhibition balance. The rhythmicity of branched-chain amino acids in plasma concentrations, observed over a 24-hour cycle and conserved in rodent models, is under circadian clock control. The mechanisms underlying those rhythms and the physiological consequences of their disruption are not fully understood. Disturbed sleep, obesity, diabetes, and cardiovascular diseases can elevate branched-chain amino acid concentrations or modify their oscillatory dynamics. The mechanisms driving these effects are currently the focal point of ongoing research efforts, since normalizing branched-chain amino acid levels has the ability to alleviate the severity of these pathologies. In this context, the Drosophila model, though underutilized, holds promise in shedding new light on these mechanisms. Initial findings indicate its potential to introduce novel concepts, particularly in elucidating the intricate connections between the circadian clock, sleep/wake, and metabolism. Consequently, the use and transport of branched-chain amino acids emerge as critical components and orchestrators in the web of interactions across multiple organs throughout the sleep/wake cycle. They could represent one of the so far elusive mechanisms connecting sleep patterns to metabolic and cardiovascular health, paving the way for potential therapeutic interventions.</p>","PeriodicalId":19113,"journal":{"name":"Neural Regeneration Research","volume":null,"pages":null},"PeriodicalIF":5.9000,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Neural Regeneration Research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.4103/NRR.NRR-D-23-02020","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/6/3 0:00:00","PubModel":"Epub","JCR":"Q2","JCRName":"CELL BIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

The sleep-wake cycle stands as an integrative process essential for sustaining optimal brain function and, either directly or indirectly, overall body health, encompassing metabolic and cardiovascular well-being. Given the heightened metabolic activity of the brain, there exists a considerable demand for nutrients in comparison to other organs. Among these, the branched-chain amino acids, comprising leucine, isoleucine, and valine, display distinctive significance, from their contribution to protein structure to their involvement in overall metabolism, especially in cerebral processes. Among the first amino acids that are released into circulation post-food intake, branched-chain amino acids assume a pivotal role in the regulation of protein synthesis, modulating insulin secretion and the amino acid sensing pathway of target of rapamycin. Branched-chain amino acids are key players in influencing the brain's uptake of monoamine precursors, competing for a shared transporter. Beyond their involvement in protein synthesis, these amino acids contribute to the metabolic cycles of γ-aminobutyric acid and glutamate, as well as energy metabolism. Notably, they impact GABAergic neurons and the excitation/inhibition balance. The rhythmicity of branched-chain amino acids in plasma concentrations, observed over a 24-hour cycle and conserved in rodent models, is under circadian clock control. The mechanisms underlying those rhythms and the physiological consequences of their disruption are not fully understood. Disturbed sleep, obesity, diabetes, and cardiovascular diseases can elevate branched-chain amino acid concentrations or modify their oscillatory dynamics. The mechanisms driving these effects are currently the focal point of ongoing research efforts, since normalizing branched-chain amino acid levels has the ability to alleviate the severity of these pathologies. In this context, the Drosophila model, though underutilized, holds promise in shedding new light on these mechanisms. Initial findings indicate its potential to introduce novel concepts, particularly in elucidating the intricate connections between the circadian clock, sleep/wake, and metabolism. Consequently, the use and transport of branched-chain amino acids emerge as critical components and orchestrators in the web of interactions across multiple organs throughout the sleep/wake cycle. They could represent one of the so far elusive mechanisms connecting sleep patterns to metabolic and cardiovascular health, paving the way for potential therapeutic interventions.

解码关系:支链氨基酸及其与睡眠、昼夜节律和心脏代谢健康的关系。
睡眠-觉醒周期是一个综合过程,对于维持大脑的最佳功能以及直接或间接地维持包括新陈代谢和心血管健康在内的全身健康至关重要。由于大脑的新陈代谢活动频繁,与其他器官相比,大脑对营养物质的需求量相当大。其中,由亮氨酸、异亮氨酸和缬氨酸组成的支链氨基酸,从对蛋白质结构的贡献到参与整体新陈代谢,尤其是大脑过程,都显示出独特的重要性。支链氨基酸是摄入食物后最先释放到血液循环中的氨基酸,在调节蛋白质合成、调节胰岛素分泌和雷帕霉素靶点的氨基酸传感途径中发挥着关键作用。支链氨基酸是影响大脑摄取单胺前体的关键因素,它们竞争共享一个转运体。除了参与蛋白质合成外,这些氨基酸还有助于γ-氨基丁酸和谷氨酸的代谢循环以及能量代谢。值得注意的是,它们会影响 GABA 能神经元和兴奋/抑制平衡。支链氨基酸在血浆中浓度的节律性受昼夜节律钟控制,这种节律性在 24 小时周期内均可观察到,并在啮齿动物模型中得到保留。目前还不完全清楚这些节律的内在机制及其被破坏后的生理后果。睡眠紊乱、肥胖、糖尿病和心血管疾病会使支链氨基酸浓度升高或改变其振荡动态。驱动这些影响的机制是目前研究工作的重点,因为使支链氨基酸水平正常化能够减轻这些病症的严重程度。在这种情况下,果蝇模型虽然利用率不高,但有望为这些机制带来新的启示。初步研究结果表明,果蝇模型具有引入新概念的潜力,特别是在阐明昼夜节律、睡眠/觉醒和新陈代谢之间错综复杂的联系方面。因此,支链氨基酸的使用和运输成为整个睡眠/觉醒周期中多个器官相互作用网络的关键组成部分和协调者。它们可能是迄今为止难以捉摸的连接睡眠模式与代谢和心血管健康的机制之一,为潜在的治疗干预铺平了道路。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Neural Regeneration Research
Neural Regeneration Research CELL BIOLOGY-NEUROSCIENCES
CiteScore
8.00
自引率
9.80%
发文量
515
审稿时长
1.0 months
期刊介绍: Neural Regeneration Research (NRR) is the Open Access journal specializing in neural regeneration and indexed by SCI-E and PubMed. The journal is committed to publishing articles on basic pathobiology of injury, repair and protection to the nervous system, while considering preclinical and clinical trials targeted at improving traumatically injuried patients and patients with neurodegenerative diseases.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信