EGF receptor stimulation shifts breast cancer cell glucose metabolism toward glycolytic flux through PI3 kinase signaling.

Breast cancers that express epidermal growth factor (EGF) receptors (EGFRs) are associated with poor prognosis. Our group recently showed in breast cancer patients that EGFR expression is strongly correlated with high tumor uptake of the glucose analogue, 18F-fluorodeoxyglucose (FDG). Here, we explo...

Full description

Bibliographic Details
Main Authors: Kyung-Ho Jung, Eun Jeong Lee, Jin Won Park, Jin Hee Lee, Seung Hwan Moon, Young Seok Cho, Kyung-Han Lee
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2019-01-01
Series:PLoS ONE
Online Access:https://doi.org/10.1371/journal.pone.0221294
id doaj-afc2c9140c994e1b828dd55f63819733
record_format Article
spelling doaj-afc2c9140c994e1b828dd55f638197332021-03-03T21:07:45ZengPublic Library of Science (PLoS)PLoS ONE1932-62032019-01-01149e022129410.1371/journal.pone.0221294EGF receptor stimulation shifts breast cancer cell glucose metabolism toward glycolytic flux through PI3 kinase signaling.Kyung-Ho JungEun Jeong LeeJin Won ParkJin Hee LeeSeung Hwan MoonYoung Seok ChoKyung-Han LeeBreast cancers that express epidermal growth factor (EGF) receptors (EGFRs) are associated with poor prognosis. Our group recently showed in breast cancer patients that EGFR expression is strongly correlated with high tumor uptake of the glucose analogue, 18F-fluorodeoxyglucose (FDG). Here, we explored the cellular mechanism and signaling pathways that can explain the relation between EGFR and breast cancer cell glucose metabolism. FDG uptake, lactate production and hexokinase (HK) activity were measured, and proliferation assays and western blots were performed. EGF stimulated an increase of FDG uptake in EGFR-positive T47D and MDA-MB-468 cells, but not in MCF-7 cells. In T47D cells, the effect was dose-dependent and was accompanied by increased lactate production, indicating a shift toward glycolytic flux. This metabolic response occurred through enhanced HK activity and upregulated glucose transporter 1 (GLUT1) expression. EGFR stimulation also increased T47D cell proliferation. Blocking EGFR activation with BIBX1382 or gefitinib completely abolished both FDG uptake and proliferation effects. EGFR stimulation induced MAP kinase (MAPK) and PI3 kinase (PI3K) activation. Increased cell proliferation by EGFR stimulation was completely abolished by MAPK inhibition with PD98059 or by PI3K inhibition with LY294002. Increased FDG uptake was also completely abrogated by PI3K inhibition but was uninfluenced by MAPK inhibition. These findings suggest that the association between breast tumor EGFR expression and high FDG uptake might be contributed by stimulation of the PI3K pathway downstream of EGFR activation. This was in contrast to EGFR-mediated cell proliferation that required MAPK as well as PI3K signaling.https://doi.org/10.1371/journal.pone.0221294
collection DOAJ
language English
format Article
sources DOAJ
author Kyung-Ho Jung
Eun Jeong Lee
Jin Won Park
Jin Hee Lee
Seung Hwan Moon
Young Seok Cho
Kyung-Han Lee
spellingShingle Kyung-Ho Jung
Eun Jeong Lee
Jin Won Park
Jin Hee Lee
Seung Hwan Moon
Young Seok Cho
Kyung-Han Lee
EGF receptor stimulation shifts breast cancer cell glucose metabolism toward glycolytic flux through PI3 kinase signaling.
PLoS ONE
author_facet Kyung-Ho Jung
Eun Jeong Lee
Jin Won Park
Jin Hee Lee
Seung Hwan Moon
Young Seok Cho
Kyung-Han Lee
author_sort Kyung-Ho Jung
title EGF receptor stimulation shifts breast cancer cell glucose metabolism toward glycolytic flux through PI3 kinase signaling.
title_short EGF receptor stimulation shifts breast cancer cell glucose metabolism toward glycolytic flux through PI3 kinase signaling.
title_full EGF receptor stimulation shifts breast cancer cell glucose metabolism toward glycolytic flux through PI3 kinase signaling.
title_fullStr EGF receptor stimulation shifts breast cancer cell glucose metabolism toward glycolytic flux through PI3 kinase signaling.
title_full_unstemmed EGF receptor stimulation shifts breast cancer cell glucose metabolism toward glycolytic flux through PI3 kinase signaling.
title_sort egf receptor stimulation shifts breast cancer cell glucose metabolism toward glycolytic flux through pi3 kinase signaling.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2019-01-01
description Breast cancers that express epidermal growth factor (EGF) receptors (EGFRs) are associated with poor prognosis. Our group recently showed in breast cancer patients that EGFR expression is strongly correlated with high tumor uptake of the glucose analogue, 18F-fluorodeoxyglucose (FDG). Here, we explored the cellular mechanism and signaling pathways that can explain the relation between EGFR and breast cancer cell glucose metabolism. FDG uptake, lactate production and hexokinase (HK) activity were measured, and proliferation assays and western blots were performed. EGF stimulated an increase of FDG uptake in EGFR-positive T47D and MDA-MB-468 cells, but not in MCF-7 cells. In T47D cells, the effect was dose-dependent and was accompanied by increased lactate production, indicating a shift toward glycolytic flux. This metabolic response occurred through enhanced HK activity and upregulated glucose transporter 1 (GLUT1) expression. EGFR stimulation also increased T47D cell proliferation. Blocking EGFR activation with BIBX1382 or gefitinib completely abolished both FDG uptake and proliferation effects. EGFR stimulation induced MAP kinase (MAPK) and PI3 kinase (PI3K) activation. Increased cell proliferation by EGFR stimulation was completely abolished by MAPK inhibition with PD98059 or by PI3K inhibition with LY294002. Increased FDG uptake was also completely abrogated by PI3K inhibition but was uninfluenced by MAPK inhibition. These findings suggest that the association between breast tumor EGFR expression and high FDG uptake might be contributed by stimulation of the PI3K pathway downstream of EGFR activation. This was in contrast to EGFR-mediated cell proliferation that required MAPK as well as PI3K signaling.
url https://doi.org/10.1371/journal.pone.0221294
work_keys_str_mv AT kyunghojung egfreceptorstimulationshiftsbreastcancercellglucosemetabolismtowardglycolyticfluxthroughpi3kinasesignaling
AT eunjeonglee egfreceptorstimulationshiftsbreastcancercellglucosemetabolismtowardglycolyticfluxthroughpi3kinasesignaling
AT jinwonpark egfreceptorstimulationshiftsbreastcancercellglucosemetabolismtowardglycolyticfluxthroughpi3kinasesignaling
AT jinheelee egfreceptorstimulationshiftsbreastcancercellglucosemetabolismtowardglycolyticfluxthroughpi3kinasesignaling
AT seunghwanmoon egfreceptorstimulationshiftsbreastcancercellglucosemetabolismtowardglycolyticfluxthroughpi3kinasesignaling
AT youngseokcho egfreceptorstimulationshiftsbreastcancercellglucosemetabolismtowardglycolyticfluxthroughpi3kinasesignaling
AT kyunghanlee egfreceptorstimulationshiftsbreastcancercellglucosemetabolismtowardglycolyticfluxthroughpi3kinasesignaling
_version_ 1714818700767395840