4.7 Article

Hypoxia-driven metabolic reprogramming of adipocytes fuels cancer cell proliferation

Journal

FRONTIERS IN ENDOCRINOLOGY
Volume 13, Issue -, Pages -

Publisher

FRONTIERS MEDIA SA
DOI: 10.3389/fendo.2022.989523

Keywords

hypoxia; adipocytes; cancer cells; metabolites; lipids; obesity

Funding

  1. Wellcome Trust ISSF3
  2. British Heart Foundation REA2 [RE/13/3/30183]
  3. Chief Scientist Office [SCAF/17/02]
  4. Wellcome Trust [101067/Z/13/Z]
  5. MRC Centre grant [MR/N022556/1]
  6. CRUK programme grant [C17950/A26783]

Ask authors/readers for more resources

This study found that hypoxia in the adipose-tumour microenvironment promotes lipid uptake in cancer cells, leading to increased proliferation. Additionally, hypoxia alters the metabolome of adipocytes and drives proliferation of non-malignant cells.
ObjectiveObesity 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. MethodsHuman 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. ResultsHypoxic 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 HIF1 alpha expression in adipocytes, as mouse adipocytes lacking HIF1 alpha 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. ConclusionHere, 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.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available