Hypoxia-driven metabolic reprogramming of adipocytes fuels cancer cell proliferation

Obesity increases the risk of certain cancers, especially tumours that reside close to adipose tissue (breast and ovarian metastasis in the omentum). The obesogenic and tumour micro-environment share a common pathogenic feature, oxygen deprivation (hypoxia). Here we test how hypoxia changes the meta...

Full description

Saved in:
Bibliographic Details
Published inFrontiers in endocrinology (Lausanne) Vol. 13; p. 989523
Main Authors Aird, R, Wills, J, Roby, K F, Bénézech, C, Stimson, R H, Wabitsch, M, Pollard, J W, Finch, A, Michailidou, Z
Format Journal Article
LanguageEnglish
Published Switzerland Frontiers Media S.A 18.10.2022
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Obesity increases the risk of certain cancers, especially tumours that reside close to adipose tissue (breast and ovarian metastasis in the omentum). The obesogenic and tumour micro-environment share a common pathogenic feature, oxygen deprivation (hypoxia). Here we test how hypoxia changes the metabolome of adipocytes to assist cancer cell growth. Human and mouse breast and ovarian cancer cell lines were co-cultured with human and mouse adipocytes respectively under normoxia or hypoxia. Proliferation and lipid uptake in cancer cells were measured by commercial assays. Metabolite changes under normoxia or hypoxia were measured in the media of human adipocytes by targeted LC/MS. Hypoxic cancer-conditioned media increased lipolysis in both human and mouse adipocytes. This led to increased transfer of lipids to cancer cells and consequent increased proliferation under hypoxia. These effects were dependent on HIF1α expression in adipocytes, as mouse adipocytes lacking HIF1α showed blunted responses under hypoxic conditions. Targeted metabolomics of the human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes media revealed that culture with hypoxic-conditioned media from non-malignant mammary epithelial cells (MCF10A) can alter the adipocyte metabolome and drive proliferation of the non-malignant cells. Here, we show that hypoxia in the adipose-tumour microenvironment is the driving force of the lipid uptake in both mammary and ovarian cancer cells. Hypoxia can modify the adipocyte metabolome towards accelerated lipolysis, glucose deprivation and reduced ketosis. These metabolic shifts in adipocytes could assist both mammary epithelial and cancer cells to bypass the inhibitory effects of hypoxia on proliferation and thrive.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
Reviewed by: Gerald V. Denis, Boston University, United States; Kang Liu, China Pharmaceutical University, China
Edited by: Nabil Rabhi, Boston University, United States
This article was submitted to Cellular Endocrinology, a section of the journal Frontiers in Endocrinology
ISSN:1664-2392
1664-2392
DOI:10.3389/fendo.2022.989523