figshare
Browse
martin-asystematicfluxanalysis-2019.pdf (2.23 MB)

A systematic flux analysis approach to identify metabolic vulnerabilities in human breast cancer cell lines

Download (2.23 MB)
Version 3 2024-06-18, 18:49
Version 2 2024-06-03, 15:17
Version 1 2020-06-01, 14:13
journal contribution
posted on 2024-06-18, 18:49 authored by Sheree D Martin, Sean McgeeSean Mcgee
AbstractBackgroundIncreased flux through both glycolytic and oxidative metabolic pathways is a hallmark of breast cancer cells and is critical for their growth and survival. As such, targeting this metabolic reprograming has received much attention as a potential treatment approach. However, the heterogeneity of breast cancer cell metabolism, even within classifications, suggests a necessity for an individualised approach to treatment in breast cancer patients.MethodsThe metabolic phenotypes of a diverse panel of human breast cancer cell lines representing the major breast cancer classifications were assessed using real-time metabolic flux analysis. Flux linked to ATP production, pathway reserve capacities and specific macromolecule oxidation rates were quantified. Suspected metabolic vulnerabilities were targeted with specific pathway inhibitors, and relative cell viability was assessed using the crystal violet assay. Measures of AMPK and mTORC1 activity were analysed through immunoblotting.ResultsBreast cancer cells displayed heterogeneous energy requirements and utilisation of non-oxidative and oxidative energy-producing pathways. Quantification of basal glycolytic and oxidative reserve capacities identified cell lines that were highly dependent on individual pathways, while assessment of substrate oxidation relative to total oxidative capacity revealed cell lines that were highly dependent on individual macromolecules. Based on these findings, mild mitochondrial inhibition in ESH-172 cells, including with the anti-diabetic drug metformin, and mild glycolytic inhibition in Hs578T cells reduced relative viability, which did not occur in non-transformed MCF10a cells. The effects on viability were associated with AMPK activation and inhibition of mTORC1 signalling. Hs578T were also found to be highly dependent on glutamine oxidation and inhibition of this process also impacted viability.ConclusionsTogether, these data highlight that systematic flux analysis in breast cancer cells can identify targetable metabolic vulnerabilities, despite heterogeneity in metabolic profiles between individual cancer cell lines.

History

Journal

CANCER & METABOLISM

Volume

7

Article number

ARTN 12

Pagination

1 - 14

Location

England

Open access

  • Yes

ISSN

2049-3002

eISSN

2049-3002

Language

English

Publication classification

C1 Refereed article in a scholarly journal

Issue

1

Publisher

BMC