Your browser doesn't support javascript.
loading
Lipid accumulation facilitates mitotic slippage-induced adaptation to anti-mitotic drug treatment.
Wong, Alex; Chen, Sixun; Yang, Lay Kien; Kanagasundaram, Yoganathan; Crasta, Karen.
Afiliación
  • Wong A; 1Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
  • Chen S; 2Institute for Health Technologies, Interdisciplinary Graduate School, Nanyang Technological University, Singapore, Singapore.
  • Yang LK; 1Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
  • Kanagasundaram Y; 3Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research, Singapore, Singapore.
  • Crasta K; 4Bioinformatics Institute, Agency for Science, Technology, and Research, Singapore, Singapore.
Cell Death Discov ; 4: 109, 2018.
Article en En | MEDLINE | ID: mdl-30510774
ABSTRACT
Aberrant lipid accumulation is a hallmark of cancer known to contribute to its aggressiveness and malignancy. Emerging studies have demonstrated context-dependent changes in lipid metabolism during chemotherapy. However, there is little known regarding the mechanisms linking lipid metabolism to chemotherapy-induced cell fates. Here, we describe lipid accumulation in cells following antimitotic drug treatment. Cells arrested in mitosis, as well as cells that escaped mitotic arrest and underwent mitotic slippage, showed elevated cytoplasmic lipid droplets. Interestingly, we found that TOFA, a lipid biosynthesis inhibitor that targets acetyl-CoA carboxylase (ACC) and blocks lipid accumulation, promoted early slippage, reduced cellular stress and enhanced survival of antimitotic-treated cells. Our work previously revealed that cells that survive after mitotic slippage can become senescent and confer pro-tumourigenic effects through paracrine signalling. Modulating lipid biosynthesis in cells post slippage by TOFA amplified their inflammatory secretion profiles and accelerated the development of tumourigenic behaviour, particularly cell migration and invasion, in a paracrine-dependent manner. In contrast to TOFA, inhibition of lipid accumulation by C75, a drug targeting fatty acid synthase (FASN), significantly reduced the production of pro-tumourigenic factors and associated phenotypic effects. This suggests that discrete lipid biosynthesis pathways could contribute differentially to the regulation of pro-tumourigenic inflammation. The divergent effects of TOFA and C75 may be attributed to the opposing regulation of Malonyl-CoA, an intermediate in fatty acid synthesis that serves as a mediator of fatty acid oxidation. Taken together, our data reveal a previously unappreciated role for lipid accumulation in the cellular adaptation to antimitotic drug treatment. Targeting lipid biosynthesis in cells post slippage may reprogramme its secretory profile such that it not only negates tumour-promoting effects, but may also promote anti-tumour inflammation for clearance of post-slippage senescent cells.

Texto completo: 1 Bases de datos: MEDLINE Idioma: En Revista: Cell Death Discov Año: 2018 Tipo del documento: Article País de afiliación: Singapur

Texto completo: 1 Bases de datos: MEDLINE Idioma: En Revista: Cell Death Discov Año: 2018 Tipo del documento: Article País de afiliación: Singapur