ApoE expression in macrophages communicates immunometabolic signaling that controls hyperlipidemia‐driven hematopoiesis & inflammation via extracellular vesicles

Author:

Phu Tuan Anh12,Ng Martin12,Vu Ngan K.12,Gao Alex S.12,Raffai Robert L.123ORCID

Affiliation:

1. Department of Veterans Affairs Surgical Service (112G) San Francisco VA Medical Center San Francisco California USA

2. Northern California Institute for Research and Education San Francisco California USA

3. Department of Surgery Division of Endovascular and Vascular Surgery University of California San Francisco California USA

Abstract

AbstractWhile apolipoprotein E (apoE) expression by myeloid cells is recognized to control inflammation, whether such benefits can be communicated via extracellular vesicles is not known. Through the study of extracellular vesicles produced by macrophages derived from the bone marrow of Wildtype (WT‐BMDM‐EV) and ApoE deficient (EKO‐BMDM‐EV) mice, we uncovered a critical role for apoE expression in regulating their cell signaling properties. WT‐BMDM‐EV communicated anti‐inflammatory properties to recipient myeloid cells by increasing cellular levels of apoE and miR‐146a‐5p, that reduced NF‐κB signalling. They also downregulated cellular levels of miR‐142a‐3p, resulting in increased levels of its target carnitine palmitoyl transferase 1A (CPT1A) which improved fatty acid oxidation (FAO) and oxidative phosphorylation (OxPHOS) in recipient cells. Such favorable metabolic polarization enhanced cell‐surface MerTK levels and the phagocytic uptake of apoptotic cells. In contrast, EKO‐BMDM‐EV exerted opposite effects by reducing cellular levels of apoE and miR‐146a‐5p, which increased NF‐κB−driven GLUT1‐mediated glucose uptake, aerobic glycolysis, and oxidative stress. Furthermore, EKO‐BMDM‐EV increased cellular miR‐142a‐3p levels, which reduced CPT1A levels and impaired FAO and OxPHOS in recipient myeloid cells. When cultured with naïve CD4+ T lymphocytes, EKO‐BMDM‐EV drove their activation and proliferation, and fostered their transition to a Th1 phenotype. While infusions of WT‐BMDM‐EV into hyperlipidemic mice resolved inflammation, infusions of EKO‐BMDM‐EV increased hematopoiesis and drove inflammatory responses in myeloid cells and T lymphocytes. ApoE‐dependent immunometabolic signaling by macrophage extracellular vesicles was dependent on transcriptional axes controlled by miR‐146a‐5p and miR‐142a‐3p that could be reproduced by infusing miR‐146a mimics & miR‐142a antagonists into hyperlipidemic apoE‐deficient mice. Together, our findings unveil a novel property for apoE expression in macrophages that modulates the immunometabolic regulatory properties of their secreted extracellular vesicles.

Funder

U.S. Department of Veterans Affairs

National Institutes of Health

Publisher

Wiley

Subject

Cell Biology,Histology

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

同舟云学术

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

"同舟云学术"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,当前同舟云学术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 © 2019-2024 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