4.6 Article

Epigenetic Silencing of miR-33b Promotes Peritoneal Metastases of Ovarian Cancer by Modulating the TAK1/FASN/CPT1A/NF-κB Axis

Journal

CANCERS
Volume 13, Issue 19, Pages -

Publisher

MDPI
DOI: 10.3390/cancers13194795

Keywords

ovarian cancer; omental metastases; miR-33b; DNA methylation; lipid metabolism

Categories

Funding

  1. University-Industry Collaboration Programme, the Innovation and Technology Commission (Hong Kong) [UIM367]

Ask authors/readers for more resources

The study reveals that miR-33b is frequently silenced by DNA hypermethylation in metastatic ovarian cancer cells, and its restoration can impair lipid metabolic activities and reduce oncogenic properties, potentially serving as a therapeutic target for ovarian cancer metastatic progression.
Simple Summary Omental metastasis and peritoneal dissemination are frequently observed in ovarian cancer peritoneal metastases and are associated with high mortality and poor prognosis. The tumor microenvironment is known to influence cancer epigenomics, which plays an essential role in promoting tumor development and metastatic progression. Therefore, investigation of the epigenetic mechanisms underlying the growth of ovarian cancer cells in the omental metastatic microenvironment is of great importance. Here, we report that miR-33b is significantly silenced by DNA hypermethylation in metastatic ovarian cancer cells to adapt to a lipid-rich microenvironment. Restoration of miR-33b was shown to impair lipid metabolic activities and reduce the oncogenic properties of ovarian cancer cells by negatively regulating the TAK1/FASN/CPT1A/NF-kappa B pathway, indicating that targeting this signaling cascade may be a molecular therapeutic choice for ovarian cancer metastatic progression. Peritoneal metastases are frequently found in high-grade serous carcinoma (HGSOC) patients and are commonly associated with a poor prognosis. The tumor microenvironment (TME) is a complex milieu that plays a critical role in epigenetic alterations driving tumor development and metastatic progression. However, the impact of epigenetic alterations on metastatic ovarian cancer cells in the harsh peritoneal microenvironment remains incompletely understood. Here, we identified that miR-33b is frequently silenced by promoter hypermethylation in HGSOC cells derived from metastatic omental tumor tissues. Enforced expression of miR-33b abrogates the oncogenic properties of ovarian cancer cells cocultured in omental conditioned medium (OCM), which mimics the ascites microenvironment, and in vivo tumor growth. Of note, restoration of miR-33b inhibited OCM-upregulated de novo lipogenesis and fatty acid beta-oxidation in ovarian cancer cells, indicating that miR-33b may play a novel tumor suppressor role in the lipid-mediated oncogenic properties of metastatic ovarian cancer cells found in the omentum. Mechanistic studies demonstrated that miR-33b directly targets transforming growth factor beta-activated kinase 1 (TAK1), thereby suppressing the activities of fatty acid synthase (FASN) and carnitine palmitoyltransferase 1A (CPT1A) in modulating lipid metabolic activities and simultaneously inhibiting the phosphorylation of NF-kappa B signaling to govern the oncogenic behaviors of ovarian cancer cells. Thus, our data suggest that a lipid-rich microenvironment may cause epigenetic silencing of miR-33b, which negatively modulates ovarian cancer peritoneal metastases, at least in part, by suppressing TAK1/FASN/CPT1A/NF-kappa B signaling.

Authors

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

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available