4.7 Article

Metabolomics reveals that CAF-derived lipids promote colorectal cancer peritoneal metastasis by enhancing membrane fluidity

期刊

INTERNATIONAL JOURNAL OF BIOLOGICAL SCIENCES
卷 18, 期 5, 页码 1912-1932

出版社

IVYSPRING INT PUBL
DOI: 10.7150/ijbs.68484

关键词

Lipidomics; colorectal cancer peritoneal metastasis; cancer-associated fibroblasts (CAFs); glucose metabolism; C16:0; metabolomics

资金

  1. Natural Science Foundation of Guangdong Province [2021A1515010639, 2020A151501613, 2018A0303130303, 2018A030313567]
  2. National Natural Science Foundation of China [81972245, 81902877, 31900505]
  3. Natural Science Fund for Distinguished Young Scholars of Guangdong Province [2016A030306002]
  4. Young Teacher Foundation of Sun Yat-sen University (CN) [19ykpy03]
  5. Sun Yat-Sen University Clinical Research 5010 Program [2018026L]
  6. Five FiveTalent Team Construction Project of the Sixth Affiliated Hospital Of Sun Yat-Sen University [P20150227202010251]

向作者/读者索取更多资源

This study investigates the role of cancer-associated fibroblasts (CAFs) in peritoneal metastasis (PM) of colorectal cancer (CRC) and identifies a potential therapeutic target for the treatment of PM-CRC. The study shows that the abundance of phosphatidylcholine (PC) with unsaturated acyl chains is increased in clinical PM-CRC specimens. It also reveals that CAFs are present at a higher relative abundance in primary PM-CRC tumors and that incubation with CAF-conditioned medium (CM) increases the membrane fluidity in CRC cells. The study suggests that elevated membrane fluidity enhances glucose uptake and metabolism in CRC cells.
Patients with peritoneal metastasis (PM) of colorectal cancer (CRC) have poorer overall survival outcomes than those without PM. Cancer-associated fibroblasts (CAFs) are a major component of the tumor microenvironment and mediate CRC progression and PM. It is imperative to identify and develop novel therapeutic targets for PM-CRC driven by CAFs. Using lipidomics, we reveal that the abundance of phosphatidylcholine (PC) with unsaturated acyl chains was increased in clinical PM-CRC specimens. Additionally, we found that CAFs were present at a higher relative abundance in primary PM-CRC tumors and that membrane fluidity in CRC cells was increased after incubation with CAF-conditioned medium (CM) through three independent methods: lipidomics, fluorescence recovery after photobleaching (FRAP), and generalized polarization. Then, we found that increased membrane fluidity can enhance glucose uptake and metabolism, as supported by real-time bioenergetics analysis and U-C-13 glucose labeling. Interestingly, stearoyl-CoA desaturase 1 (SCD), the rate-limiting enzyme in the biosynthesis of unsaturated fatty acids (uS-FAs), was expressed at low levels in PM and associated with poor prognosis in CRC patients. Importantly, by untargeted metabolomics analysis and fatty acid ([U-C-1(3)]-stearic acid) tracing analyses, we found that CRC cells take up lipids and lipid-like metabolites secreted from CAFs, which may compensate for low SCD expression. Both in vitro and in vivo experiments demonstrated that sodium palmitate (C16:0) treatment could decrease the CAF-induced change in cell membrane fluidity, limit glucose metabolism, suppress cell invasiveness, and impair tumor growth and intraperitoneal dissemination. An increased C16:0 concentration was shown to induce apoptosis linked to lipotoxicity. Furthermore, C16:0 effectively enhanced the antitumor activity of 5-fluorouracil (5-FU) in vitro and was well tolerated in vivo. Taken together, these findings suggest that adding the saturated fatty acid (S-FA) C16:0 to neoadjuvant chemotherapy may open new opportunities for treating PM-CRC in the future.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据