Abstract

Simple SummaryOmental 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-κB 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 β-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-κB 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-κB signaling.

Highlights

  • Ovarian cancer is one of the most common and lethal cancer types in women worldwide [1]

  • fatty acid synthase (FASN) and carnitine palmitoyltransferase 1A (CPT1A) expression in ES-2 cells compared with coculture in 1% fetal bovine serum (FBS)-negative control medium, whereas the omental conditioned medium (OCM)-mediated upregulation of FASN and CPT1A was attenuated upon overexpression of miR-33b in ovarian cancer cells (Figure 5A). These results suggest that overexpression of miR-33b mitigates FASN and CPT1A regulation and that de novo lipogenesis and fatty acid degradation in ovarian cancer cells are inhibited as a result

  • The results showed that miR-33b overexpression reduced transforming growth factor beta-activated kinase 1 (TAK1), FASN, CPT1A, P-IKKα/β, P-IκBα, and Ki67 protein expression in xenograft tumor tissues (Figure 6F)

Read more

Summary

Introduction

Ovarian cancer is one of the most common and lethal cancer types in women worldwide [1]. Similar to other human solid cancers, metastases are a critical factor leading to death and recurrence in ovarian cancer patients [2,3]. Accounts for over 90% of ovarian cancer cases, and the deadliest and most frequent histological subtype is high-grade serous carcinoma (HGSOC), representing the largest subgroup (>70%) of EOC [2,3]. Due to the lack of early clinical symptoms, HGSOC often presents with “silent symptoms,” and the majority of cases are diagnosed at an advanced stage, accompanied by peritoneal metastases, which contribute to most ovarian cancer-related deaths and a typically abysmal prognosis [1,4,5]. Diffuse peritoneal dissemination of EOC, as well as a favorable microenvironment for tumor development and progression, contributes to acquired platinum resistance and disease recurrence in ovarian cancer patients [7,8]. A better understanding of the pathophysiology of ovarian cancer is urgently needed to explore alternative and novel antitumor therapies

Methods
Findings
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call