Cargando…

MIEF2 reprograms lipid metabolism to drive progression of ovarian cancer through ROS/AKT/mTOR signaling pathway

MIEF2 (mitochondrial elongation factor 2) is one of the key regulators of mitochondrial fission. Bioinformatics analysis indicated that high expression of MIEF2 predicted a poor prognosis in ovarian cancer patients. However, the relationship between MIEF2 and aberrant lipid metabolism in OC remains...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhao, Shuhua, Cheng, Lu, Shi, Yuan, Li, Jia, Yun, Qinghui, Yang, Hong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7791105/
https://www.ncbi.nlm.nih.gov/pubmed/33414447
http://dx.doi.org/10.1038/s41419-020-03336-6
_version_ 1783633539032416256
author Zhao, Shuhua
Cheng, Lu
Shi, Yuan
Li, Jia
Yun, Qinghui
Yang, Hong
author_facet Zhao, Shuhua
Cheng, Lu
Shi, Yuan
Li, Jia
Yun, Qinghui
Yang, Hong
author_sort Zhao, Shuhua
collection PubMed
description MIEF2 (mitochondrial elongation factor 2) is one of the key regulators of mitochondrial fission. Bioinformatics analysis indicated that high expression of MIEF2 predicted a poor prognosis in ovarian cancer patients. However, the relationship between MIEF2 and aberrant lipid metabolism in OC remains elusive. In this study, we demonstrated that MIEF2 significantly promoted lipid synthesis, while has no significant effect on fatty acid uptake and oxidation in OC cells. MIEF2 enhanced de novo fatty acid synthesis through up-regulating the expression of sterol regulatory element binding protein 1 (SREBP1) and its transcriptional target lipogenic genes ACC1, FASN and SCD1. Meanwhile, MIEF2-promoted cholesterol biosynthesis through up-regulating the expression of sterol regulatory element binding protein 2 (SREBP2) and its transcriptional target cholesterol biosynthesis genes HMGCS1 and HMGCR. Mechanistically, increased mitochondrial reactive oxygen species (ROS) production and subsequently activation of AKT/mTOR signaling pathway was found to be involved in the up-regulation of SREBP1 and SREBP2 in OC cells. Moreover, cell growth and metastasis assays indicated that MIEF2-regulated fatty acid synthesis and cholesterol biosynthesis played a critical role in the progression of OC. Taken together, our findings indicate that MIEF2 is a critical regulator of lipid synthesis in OC, which provides a strong line of evidence for this molecule to serve as a drug target in the treatment of this malignancy.
format Online
Article
Text
id pubmed-7791105
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-77911052021-01-15 MIEF2 reprograms lipid metabolism to drive progression of ovarian cancer through ROS/AKT/mTOR signaling pathway Zhao, Shuhua Cheng, Lu Shi, Yuan Li, Jia Yun, Qinghui Yang, Hong Cell Death Dis Article MIEF2 (mitochondrial elongation factor 2) is one of the key regulators of mitochondrial fission. Bioinformatics analysis indicated that high expression of MIEF2 predicted a poor prognosis in ovarian cancer patients. However, the relationship between MIEF2 and aberrant lipid metabolism in OC remains elusive. In this study, we demonstrated that MIEF2 significantly promoted lipid synthesis, while has no significant effect on fatty acid uptake and oxidation in OC cells. MIEF2 enhanced de novo fatty acid synthesis through up-regulating the expression of sterol regulatory element binding protein 1 (SREBP1) and its transcriptional target lipogenic genes ACC1, FASN and SCD1. Meanwhile, MIEF2-promoted cholesterol biosynthesis through up-regulating the expression of sterol regulatory element binding protein 2 (SREBP2) and its transcriptional target cholesterol biosynthesis genes HMGCS1 and HMGCR. Mechanistically, increased mitochondrial reactive oxygen species (ROS) production and subsequently activation of AKT/mTOR signaling pathway was found to be involved in the up-regulation of SREBP1 and SREBP2 in OC cells. Moreover, cell growth and metastasis assays indicated that MIEF2-regulated fatty acid synthesis and cholesterol biosynthesis played a critical role in the progression of OC. Taken together, our findings indicate that MIEF2 is a critical regulator of lipid synthesis in OC, which provides a strong line of evidence for this molecule to serve as a drug target in the treatment of this malignancy. Nature Publishing Group UK 2021-01-05 /pmc/articles/PMC7791105/ /pubmed/33414447 http://dx.doi.org/10.1038/s41419-020-03336-6 Text en © The Author(s) 2021 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
spellingShingle Article
Zhao, Shuhua
Cheng, Lu
Shi, Yuan
Li, Jia
Yun, Qinghui
Yang, Hong
MIEF2 reprograms lipid metabolism to drive progression of ovarian cancer through ROS/AKT/mTOR signaling pathway
title MIEF2 reprograms lipid metabolism to drive progression of ovarian cancer through ROS/AKT/mTOR signaling pathway
title_full MIEF2 reprograms lipid metabolism to drive progression of ovarian cancer through ROS/AKT/mTOR signaling pathway
title_fullStr MIEF2 reprograms lipid metabolism to drive progression of ovarian cancer through ROS/AKT/mTOR signaling pathway
title_full_unstemmed MIEF2 reprograms lipid metabolism to drive progression of ovarian cancer through ROS/AKT/mTOR signaling pathway
title_short MIEF2 reprograms lipid metabolism to drive progression of ovarian cancer through ROS/AKT/mTOR signaling pathway
title_sort mief2 reprograms lipid metabolism to drive progression of ovarian cancer through ros/akt/mtor signaling pathway
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7791105/
https://www.ncbi.nlm.nih.gov/pubmed/33414447
http://dx.doi.org/10.1038/s41419-020-03336-6
work_keys_str_mv AT zhaoshuhua mief2reprogramslipidmetabolismtodriveprogressionofovariancancerthroughrosaktmtorsignalingpathway
AT chenglu mief2reprogramslipidmetabolismtodriveprogressionofovariancancerthroughrosaktmtorsignalingpathway
AT shiyuan mief2reprogramslipidmetabolismtodriveprogressionofovariancancerthroughrosaktmtorsignalingpathway
AT lijia mief2reprogramslipidmetabolismtodriveprogressionofovariancancerthroughrosaktmtorsignalingpathway
AT yunqinghui mief2reprogramslipidmetabolismtodriveprogressionofovariancancerthroughrosaktmtorsignalingpathway
AT yanghong mief2reprogramslipidmetabolismtodriveprogressionofovariancancerthroughrosaktmtorsignalingpathway