Cargando…

Phospholipid remodeling is critical for stem cell pluripotency by facilitating mesenchymal-to-epithelial transition

Metabolic reprogramming has emerged as a key regulator of cell fate decisions. Roles of glucose and amino acid metabolism have been extensively documented, whereas lipid metabolism in pluripotency remains largely unexplored. Using a high-coverage lipidomics approach, we reveal dynamic changes in pho...

Descripción completa

Detalles Bibliográficos
Autores principales: Wu, Yi, Chen, Keshi, Xing, Guangsuo, Li, Linpeng, Ma, Bochao, Hu, Zhijuan, Duan, Lifan, Liu, Xingguo
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Association for the Advancement of Science 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6881163/
https://www.ncbi.nlm.nih.gov/pubmed/31807705
http://dx.doi.org/10.1126/sciadv.aax7525
_version_ 1783473890980265984
author Wu, Yi
Chen, Keshi
Xing, Guangsuo
Li, Linpeng
Ma, Bochao
Hu, Zhijuan
Duan, Lifan
Liu, Xingguo
author_facet Wu, Yi
Chen, Keshi
Xing, Guangsuo
Li, Linpeng
Ma, Bochao
Hu, Zhijuan
Duan, Lifan
Liu, Xingguo
author_sort Wu, Yi
collection PubMed
description Metabolic reprogramming has emerged as a key regulator of cell fate decisions. Roles of glucose and amino acid metabolism have been extensively documented, whereas lipid metabolism in pluripotency remains largely unexplored. Using a high-coverage lipidomics approach, we reveal dynamic changes in phospholipids occurring during reprogramming and show that the CDP-ethanolamine (CDP-Etn) pathway for phosphatidylethanolamine (PE) synthesis is required at the early stage of reprogramming. Mechanistically, the CDP-Etn pathway inhibits NF-κB signaling and mesenchymal genes in a Pebp1-dependent manner, without affecting autophagy, resulting in accelerated mesenchymal-to-epithelial transition (MET) and enhanced reprogramming. Furthermore, PE binding to Pebp1 enhances the interaction of Pebp1 with IKKα/β and reduces the phosphorylation of IKKα/β. The CDP-Etn-Pebp1 axis is associated with EMT/MET in hepatocyte differentiation, indicating that Etn/PE is a broad-spectrum MET/EMT-regulating metabolite. Collectively, our study reveals an unforeseen connection between phospholipids, cell migration, and pluripotency and highlights the importance of phospholipids in cell fate transitions.
format Online
Article
Text
id pubmed-6881163
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher American Association for the Advancement of Science
record_format MEDLINE/PubMed
spelling pubmed-68811632019-12-05 Phospholipid remodeling is critical for stem cell pluripotency by facilitating mesenchymal-to-epithelial transition Wu, Yi Chen, Keshi Xing, Guangsuo Li, Linpeng Ma, Bochao Hu, Zhijuan Duan, Lifan Liu, Xingguo Sci Adv Research Articles Metabolic reprogramming has emerged as a key regulator of cell fate decisions. Roles of glucose and amino acid metabolism have been extensively documented, whereas lipid metabolism in pluripotency remains largely unexplored. Using a high-coverage lipidomics approach, we reveal dynamic changes in phospholipids occurring during reprogramming and show that the CDP-ethanolamine (CDP-Etn) pathway for phosphatidylethanolamine (PE) synthesis is required at the early stage of reprogramming. Mechanistically, the CDP-Etn pathway inhibits NF-κB signaling and mesenchymal genes in a Pebp1-dependent manner, without affecting autophagy, resulting in accelerated mesenchymal-to-epithelial transition (MET) and enhanced reprogramming. Furthermore, PE binding to Pebp1 enhances the interaction of Pebp1 with IKKα/β and reduces the phosphorylation of IKKα/β. The CDP-Etn-Pebp1 axis is associated with EMT/MET in hepatocyte differentiation, indicating that Etn/PE is a broad-spectrum MET/EMT-regulating metabolite. Collectively, our study reveals an unforeseen connection between phospholipids, cell migration, and pluripotency and highlights the importance of phospholipids in cell fate transitions. American Association for the Advancement of Science 2019-11-27 /pmc/articles/PMC6881163/ /pubmed/31807705 http://dx.doi.org/10.1126/sciadv.aax7525 Text en Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC). http://creativecommons.org/licenses/by-nc/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial license (http://creativecommons.org/licenses/by-nc/4.0/) , which permits use, distribution, and reproduction in any medium, so long as the resultant use is not for commercial advantage and provided the original work is properly cited.
spellingShingle Research Articles
Wu, Yi
Chen, Keshi
Xing, Guangsuo
Li, Linpeng
Ma, Bochao
Hu, Zhijuan
Duan, Lifan
Liu, Xingguo
Phospholipid remodeling is critical for stem cell pluripotency by facilitating mesenchymal-to-epithelial transition
title Phospholipid remodeling is critical for stem cell pluripotency by facilitating mesenchymal-to-epithelial transition
title_full Phospholipid remodeling is critical for stem cell pluripotency by facilitating mesenchymal-to-epithelial transition
title_fullStr Phospholipid remodeling is critical for stem cell pluripotency by facilitating mesenchymal-to-epithelial transition
title_full_unstemmed Phospholipid remodeling is critical for stem cell pluripotency by facilitating mesenchymal-to-epithelial transition
title_short Phospholipid remodeling is critical for stem cell pluripotency by facilitating mesenchymal-to-epithelial transition
title_sort phospholipid remodeling is critical for stem cell pluripotency by facilitating mesenchymal-to-epithelial transition
topic Research Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6881163/
https://www.ncbi.nlm.nih.gov/pubmed/31807705
http://dx.doi.org/10.1126/sciadv.aax7525
work_keys_str_mv AT wuyi phospholipidremodelingiscriticalforstemcellpluripotencybyfacilitatingmesenchymaltoepithelialtransition
AT chenkeshi phospholipidremodelingiscriticalforstemcellpluripotencybyfacilitatingmesenchymaltoepithelialtransition
AT xingguangsuo phospholipidremodelingiscriticalforstemcellpluripotencybyfacilitatingmesenchymaltoepithelialtransition
AT lilinpeng phospholipidremodelingiscriticalforstemcellpluripotencybyfacilitatingmesenchymaltoepithelialtransition
AT mabochao phospholipidremodelingiscriticalforstemcellpluripotencybyfacilitatingmesenchymaltoepithelialtransition
AT huzhijuan phospholipidremodelingiscriticalforstemcellpluripotencybyfacilitatingmesenchymaltoepithelialtransition
AT duanlifan phospholipidremodelingiscriticalforstemcellpluripotencybyfacilitatingmesenchymaltoepithelialtransition
AT liuxingguo phospholipidremodelingiscriticalforstemcellpluripotencybyfacilitatingmesenchymaltoepithelialtransition