4.7 Article

Acyl-CoA synthetase long-chain 3-mediated fatty acid oxidation is required for TGFβ1-induced epithelial-mesenchymal transition and metastasis of ecolorectal carcinoma

Journal

INTERNATIONAL JOURNAL OF BIOLOGICAL SCIENCES
Volume 18, Issue 6, Pages 2484-2496

Publisher

IVYSPRING INT PUBL
DOI: 10.7150/ijbs.69802

Keywords

Acyl-CoA synthetases 3; Fatty acid beta-oxidation; Epithelial-to-mesenchymal transition; Metastasis; Colorectal carcinoma

Funding

  1. National Natural Science Foundation of China [81874195, 82173144]
  2. Natural Science Foundation of Hunan Province [2020JJ4769]

Ask authors/readers for more resources

Cancer cells frequently undergo metabolic reprogramming, and alterations in fatty acid metabolism have received increasing concerns. Recent research shows that ACSL3 plays a role in energy metabolic reprogramming and promotes metastasis in colorectal carcinoma cells through FAO pathway.
Cancer cells frequently undergo metabolic reprogramming to support tumorigenicity and malignancy, which is recognized as a hallmark of cancer. In addition to glycolysis and glutaminolysis, alterations in fatty acid (FA) metabolism have received increasing concerns in the past few years. Recently, accumulating evidence has shown that fatty acid beta-oxidation (FAO) is abnormally activated in various tumors, which is associated with the machinery of proliferation, stemness, metastasis, and radiochemotherapeutic resistance of cancer cells. Acyl-CoA synthetases 3 (ACSL3) belongs to a family of enzymes responsible for converting free long-chain FAs into fatty acyl-CoA esters, which act as substrates both for lipid synthesis and FAO. Here, we demonstrate that transforming growth factor beta 1 (TGF beta 1) induces the up-regulation of ACSL3 through sterol regulatory element-binding protein 1 (SREBP1) signaling to promote energy metabolic reprogramming in colorectal carcinoma (CRC) cells. ACSL3 mediates the epithelial mesenchymal transition (EMT) and metastasis of CRC cells by activation of FAO pathway to produce ATP and reduced nicotinamide adenine dinucleotide phosphate (NADPH), which sustain redox homeostasis and fuel cancer cells for invasion and distal metastasis. Thus, targeting ACSL3 and FAO metabolic pathways might be exploited for therapeutic gain for CRC and other FAs- addicted cancers.

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