An NAD+-dependent metabolic checkpoint regulates hematopoietic stem cell activation and aging.

IF 17 Q1 CELL BIOLOGY Nature aging Pub Date : 2024-07-23 DOI:10.1038/s43587-024-00670-8
Zehan Song, Sang Hee Park, Wei-Chieh Mu, Yufan Feng, Chih-Ling Wang, Yifei Wang, Marine Barthez, Ayane Maruichi, Jiayue Guo, Fanghan Yang, Anita Wong Lin, Kartoosh Heydari, Claudia C S Chini, Eduardo N Chini, Cholsoon Jang, Danica Chen
{"title":"An NAD<sup>+</sup>-dependent metabolic checkpoint regulates hematopoietic stem cell activation and aging.","authors":"Zehan Song, Sang Hee Park, Wei-Chieh Mu, Yufan Feng, Chih-Ling Wang, Yifei Wang, Marine Barthez, Ayane Maruichi, Jiayue Guo, Fanghan Yang, Anita Wong Lin, Kartoosh Heydari, Claudia C S Chini, Eduardo N Chini, Cholsoon Jang, Danica Chen","doi":"10.1038/s43587-024-00670-8","DOIUrl":null,"url":null,"abstract":"<p><p>How hematopoietic stem cells (HSCs) maintain metabolic homeostasis to support tissue repair and regeneration throughout the lifespan is elusive. Here, we show that CD38, an NAD<sup>+</sup>-dependent metabolic enzyme, promotes HSC proliferation by inducing mitochondrial Ca<sup>2+</sup> influx and mitochondrial metabolism in young mice. Conversely, aberrant CD38 upregulation during aging is a driver of HSC deterioration in aged mice due to dysregulated NAD<sup>+</sup> metabolism and compromised mitochondrial stress management. The mitochondrial calcium uniporter, a mediator of mitochondrial Ca<sup>2+</sup> influx, also supports HSC proliferation in young mice yet drives HSC decline in aged mice. Pharmacological inactivation of CD38 reverses HSC aging and the pathophysiological changes of the aging hematopoietic system in aged mice. Together, our study highlights an NAD<sup>+</sup> metabolic checkpoint that balances mitochondrial activation to support HSC proliferation and mitochondrial stress management to enhance HSC self-renewal throughout the lifespan, and links aberrant Ca<sup>2+</sup> signaling to HSC aging.</p>","PeriodicalId":94150,"journal":{"name":"Nature aging","volume":null,"pages":null},"PeriodicalIF":17.0000,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Nature aging","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1038/s43587-024-00670-8","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CELL BIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

How hematopoietic stem cells (HSCs) maintain metabolic homeostasis to support tissue repair and regeneration throughout the lifespan is elusive. Here, we show that CD38, an NAD+-dependent metabolic enzyme, promotes HSC proliferation by inducing mitochondrial Ca2+ influx and mitochondrial metabolism in young mice. Conversely, aberrant CD38 upregulation during aging is a driver of HSC deterioration in aged mice due to dysregulated NAD+ metabolism and compromised mitochondrial stress management. The mitochondrial calcium uniporter, a mediator of mitochondrial Ca2+ influx, also supports HSC proliferation in young mice yet drives HSC decline in aged mice. Pharmacological inactivation of CD38 reverses HSC aging and the pathophysiological changes of the aging hematopoietic system in aged mice. Together, our study highlights an NAD+ metabolic checkpoint that balances mitochondrial activation to support HSC proliferation and mitochondrial stress management to enhance HSC self-renewal throughout the lifespan, and links aberrant Ca2+ signaling to HSC aging.

Abstract Image

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
依赖于 NAD+ 的代谢检查点调节造血干细胞的活化和衰老。
造血干细胞(HSCs)如何在整个生命周期内维持代谢平衡以支持组织修复和再生,目前尚无定论。在这里,我们发现,CD38是一种依赖于NAD+的代谢酶,在年轻小鼠体内通过诱导线粒体Ca2+流入和线粒体代谢促进造血干细胞增殖。相反,由于 NAD+ 代谢失调和线粒体应激管理受损,衰老过程中 CD38 的异常上调是老年小鼠造血干细胞退化的驱动因素。线粒体钙离子通道是线粒体 Ca2+ 流入的介质,它也支持年轻小鼠的造血干细胞增殖,但却驱动着老年小鼠造血干细胞的衰退。药物灭活 CD38 可逆转造血干细胞的衰老和衰老小鼠造血系统的病理生理变化。总之,我们的研究强调了一个 NAD+ 代谢检查点,它能平衡线粒体激活以支持造血干细胞增殖和线粒体应激管理以增强造血干细胞在整个生命周期的自我更新,并将异常 Ca2+ 信号传导与造血干细胞衰老联系起来。
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 去求助
来源期刊
CiteScore
14.70
自引率
0.00%
发文量
0
期刊最新文献
Rejuvenation of aged oocyte through exposure to young follicular microenvironment Stromal cell dysfunction contributes to thymic decline in aging DNA hydroxymethylation in aging Older age reduces mtDNA mutation inheritance Spermidine controls autophagy during fasting
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1