The ketone body β‐hydroxybutyrate shifts microglial metabolism and suppresses amyloid‐β oligomer‐induced inflammation in human microglia

Author:

Jin Lee‐Way123ORCID,Di Lucente Jacopo1ORCID,Ruiz Mendiola Ulises1ORCID,Suthprasertporn Nopparat1ORCID,Tomilov Alexey4ORCID,Cortopassi Gino4ORCID,Kim Kyoungmi25ORCID,Ramsey Jon J.4ORCID,Maezawa Izumi123ORCID

Affiliation:

1. Department of Pathology and Laboratory Medicine and Medical Investigation of Neurodevelopmental Disorders University of California Davis Medical Center Sacramento California USA

2. Medical Investigation of Neurodevelopmental Disorders Institute University of California Davis Medical Center Sacramento California USA

3. Alzheimer's Disease Research Center University of California Davis Medical Center Sacramento California USA

4. Department of Molecular Biosciences University of California, Davis Davis California USA

5. Department of Public Health Sciences University of California, Davis Davis California USA

Abstract

AbstractFatty acids are metabolized by β‐oxidation within the “mitochondrial ketogenic pathway” (MKP) to generate β‐hydroxybutyrate (BHB), a ketone body. BHB can be generated by most cells but largely by hepatocytes following exercise, fasting, or ketogenic diet consumption. BHB has been shown to modulate systemic and brain inflammation; however, its direct effects on microglia have been little studied. We investigated the impact of BHB on Aβ oligomer (AβO)‐stimulated human iPS‐derived microglia (hiMG), a model relevant to the pathogenesis of Alzheimer's disease (AD). HiMG responded to AβO with proinflammatory activation, which was mitigated by BHB at physiological concentrations of 0.1–2 mM. AβO stimulated glycolytic transcripts, suppressed genes in the β‐oxidation pathway, and induced over‐expression of AD‐relevant p46Shc, an endogenous inhibitor of thiolase, actions that are expected to suppress MKP. AβO also triggered mitochondrial Ca2+ increase, mitochondrial reactive oxygen species production, and activation of the mitochondrial permeability transition pore. BHB potently ameliorated all the above mitochondrial changes and rectified the MKP, resulting in reduced inflammasome activation and recovery of the phagocytotic function impaired by AβO. These results indicate that microglia MKP can be induced to modulate microglia immunometabolism, and that BHB can remedy “keto‐deficiency” resulting from MKP suppression and shift microglia away from proinflammatory mitochondrial metabolism. These effects of BHB may contribute to the beneficial effects of ketogenic diet intervention in aged mice and in human subjects with mild AD.

Funder

National Institute of Neurological Disorders and Stroke

National Institute on Aging

Publisher

Wiley

Subject

Genetics,Molecular Biology,Biochemistry,Biotechnology

Cited by 2 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

全球学者库

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"全球学者库"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前全球学者库共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2023 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3