Cargando…

Tumor Cells Switch to Mitochondrial Oxidative Phosphorylation under Radiation via mTOR-Mediated Hexokinase II Inhibition - A Warburg-Reversing Effect

A unique feature of cancer cells is to convert glucose into lactate to produce cellular energy, even under the presence of oxygen. Called aerobic glycolysis [The Warburg Effect] it has been extensively studied and the concept of aerobic glycolysis in tumor cells is generally accepted. However, it is...

Descripción completa

Detalles Bibliográficos
Autores principales: Lu, Chung-Ling, Qin, Lili, Liu, Hsin-Chen, Candas, Demet, Fan, Ming, Li, Jian Jian
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4373728/
https://www.ncbi.nlm.nih.gov/pubmed/25807077
http://dx.doi.org/10.1371/journal.pone.0121046
_version_ 1782363372307611648
author Lu, Chung-Ling
Qin, Lili
Liu, Hsin-Chen
Candas, Demet
Fan, Ming
Li, Jian Jian
author_facet Lu, Chung-Ling
Qin, Lili
Liu, Hsin-Chen
Candas, Demet
Fan, Ming
Li, Jian Jian
author_sort Lu, Chung-Ling
collection PubMed
description A unique feature of cancer cells is to convert glucose into lactate to produce cellular energy, even under the presence of oxygen. Called aerobic glycolysis [The Warburg Effect] it has been extensively studied and the concept of aerobic glycolysis in tumor cells is generally accepted. However, it is not clear if aerobic glycolysis in tumor cells is fixed, or can be reversed, especially under therapeutic stress conditions. Here, we report that mTOR, a critical regulator in cell proliferation, can be relocated to mitochondria, and as a result, enhances oxidative phosphorylation and reduces glycolysis. Three tumor cell lines (breast cancer MCF-7, colon cancer HCT116 and glioblastoma U87) showed a quick relocation of mTOR to mitochondria after irradiation with a single dose 5 Gy, which was companied with decreased lactate production, increased mitochondrial ATP generation and oxygen consumption. Inhibition of mTOR by rapamycin blocked radiation-induced mTOR mitochondrial relocation and the shift of glycolysis to mitochondrial respiration, and reduced the clonogenic survival. In irradiated cells, mTOR formed a complex with Hexokinase II [HK II], a key mitochondrial protein in regulation of glycolysis, causing reduced HK II enzymatic activity. These results support a novel mechanism by which tumor cells can quickly adapt to genotoxic conditions via mTOR-mediated reprogramming of bioenergetics from predominantly aerobic glycolysis to mitochondrial oxidative phosphorylation. Such a “waking-up” pathway for mitochondrial bioenergetics demonstrates a flexible feature in the energy metabolism of cancer cells, and may be required for additional cellular energy consumption for damage repair and survival. Thus, the reversible cellular energy metabolisms should be considered in blocking tumor metabolism and may be targeted to sensitize them in anti-cancer therapy.
format Online
Article
Text
id pubmed-4373728
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-43737282015-03-27 Tumor Cells Switch to Mitochondrial Oxidative Phosphorylation under Radiation via mTOR-Mediated Hexokinase II Inhibition - A Warburg-Reversing Effect Lu, Chung-Ling Qin, Lili Liu, Hsin-Chen Candas, Demet Fan, Ming Li, Jian Jian PLoS One Research Article A unique feature of cancer cells is to convert glucose into lactate to produce cellular energy, even under the presence of oxygen. Called aerobic glycolysis [The Warburg Effect] it has been extensively studied and the concept of aerobic glycolysis in tumor cells is generally accepted. However, it is not clear if aerobic glycolysis in tumor cells is fixed, or can be reversed, especially under therapeutic stress conditions. Here, we report that mTOR, a critical regulator in cell proliferation, can be relocated to mitochondria, and as a result, enhances oxidative phosphorylation and reduces glycolysis. Three tumor cell lines (breast cancer MCF-7, colon cancer HCT116 and glioblastoma U87) showed a quick relocation of mTOR to mitochondria after irradiation with a single dose 5 Gy, which was companied with decreased lactate production, increased mitochondrial ATP generation and oxygen consumption. Inhibition of mTOR by rapamycin blocked radiation-induced mTOR mitochondrial relocation and the shift of glycolysis to mitochondrial respiration, and reduced the clonogenic survival. In irradiated cells, mTOR formed a complex with Hexokinase II [HK II], a key mitochondrial protein in regulation of glycolysis, causing reduced HK II enzymatic activity. These results support a novel mechanism by which tumor cells can quickly adapt to genotoxic conditions via mTOR-mediated reprogramming of bioenergetics from predominantly aerobic glycolysis to mitochondrial oxidative phosphorylation. Such a “waking-up” pathway for mitochondrial bioenergetics demonstrates a flexible feature in the energy metabolism of cancer cells, and may be required for additional cellular energy consumption for damage repair and survival. Thus, the reversible cellular energy metabolisms should be considered in blocking tumor metabolism and may be targeted to sensitize them in anti-cancer therapy. Public Library of Science 2015-03-25 /pmc/articles/PMC4373728/ /pubmed/25807077 http://dx.doi.org/10.1371/journal.pone.0121046 Text en © 2015 Lu et al http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Lu, Chung-Ling
Qin, Lili
Liu, Hsin-Chen
Candas, Demet
Fan, Ming
Li, Jian Jian
Tumor Cells Switch to Mitochondrial Oxidative Phosphorylation under Radiation via mTOR-Mediated Hexokinase II Inhibition - A Warburg-Reversing Effect
title Tumor Cells Switch to Mitochondrial Oxidative Phosphorylation under Radiation via mTOR-Mediated Hexokinase II Inhibition - A Warburg-Reversing Effect
title_full Tumor Cells Switch to Mitochondrial Oxidative Phosphorylation under Radiation via mTOR-Mediated Hexokinase II Inhibition - A Warburg-Reversing Effect
title_fullStr Tumor Cells Switch to Mitochondrial Oxidative Phosphorylation under Radiation via mTOR-Mediated Hexokinase II Inhibition - A Warburg-Reversing Effect
title_full_unstemmed Tumor Cells Switch to Mitochondrial Oxidative Phosphorylation under Radiation via mTOR-Mediated Hexokinase II Inhibition - A Warburg-Reversing Effect
title_short Tumor Cells Switch to Mitochondrial Oxidative Phosphorylation under Radiation via mTOR-Mediated Hexokinase II Inhibition - A Warburg-Reversing Effect
title_sort tumor cells switch to mitochondrial oxidative phosphorylation under radiation via mtor-mediated hexokinase ii inhibition - a warburg-reversing effect
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4373728/
https://www.ncbi.nlm.nih.gov/pubmed/25807077
http://dx.doi.org/10.1371/journal.pone.0121046
work_keys_str_mv AT luchungling tumorcellsswitchtomitochondrialoxidativephosphorylationunderradiationviamtormediatedhexokinaseiiinhibitionawarburgreversingeffect
AT qinlili tumorcellsswitchtomitochondrialoxidativephosphorylationunderradiationviamtormediatedhexokinaseiiinhibitionawarburgreversingeffect
AT liuhsinchen tumorcellsswitchtomitochondrialoxidativephosphorylationunderradiationviamtormediatedhexokinaseiiinhibitionawarburgreversingeffect
AT candasdemet tumorcellsswitchtomitochondrialoxidativephosphorylationunderradiationviamtormediatedhexokinaseiiinhibitionawarburgreversingeffect
AT fanming tumorcellsswitchtomitochondrialoxidativephosphorylationunderradiationviamtormediatedhexokinaseiiinhibitionawarburgreversingeffect
AT lijianjian tumorcellsswitchtomitochondrialoxidativephosphorylationunderradiationviamtormediatedhexokinaseiiinhibitionawarburgreversingeffect