PT - JOURNAL ARTICLE AU - Ji Hye Yang AU - Nam Hee Kim AU - Jun Seop Yun AU - Eunae Sandra Cho AU - Yong Hoon Cha AU - Sue Bean Cho AU - Seon-Hyeong Lee AU - So Young Cha AU - Soo-Youl Kim AU - Jiwon Choi AU - Tin-Tin Manh Nguyen AU - Sunghyouk Park AU - Hyun Sil Kim AU - Jong In Yook TI - Snail augments fatty acid oxidation by suppression of mitochondrial ACC2 during cancer progression AID - 10.26508/lsa.202000683 DP - 2020 Jul 01 TA - Life Science Alliance PG - e202000683 VI - 3 IP - 7 4099 - https://www.life-science-alliance.org/content/3/7/e202000683.short 4100 - https://www.life-science-alliance.org/content/3/7/e202000683.full SO - Life Sci. Alliance2020 Jul 01; 3 AB - Despite the importance of mitochondrial fatty acid oxidation (FAO) in cancer metabolism, the biological mechanisms responsible for the FAO in cancer and therapeutic intervention based on catabolic metabolism are not well defined. In this study, we observe that Snail (SNAI1), a key transcriptional repressor of epithelial–mesenchymal transition, enhances catabolic FAO, allowing pro-survival of breast cancer cells in a starved environment. Mechanistically, Snail suppresses mitochondrial ACC2 (ACACB) by binding to a series of E-boxes located in its proximal promoter, resulting in decreased malonyl-CoA level. Malonyl-CoA being a well-known endogenous inhibitor of fatty acid transporter carnitine palmitoyltransferase 1 (CPT1), the suppression of ACC2 by Snail activates CPT1-dependent FAO, generating ATP and decreasing NADPH consumption. Importantly, combinatorial pharmacologic inhibition of pentose phosphate pathway and FAO with clinically available drugs efficiently reverts Snail-mediated metabolic reprogramming and suppresses in vivo metastatic progression of breast cancer cells. Our observations provide not only a mechanistic link between epithelial–mesenchymal transition and catabolic rewiring but also a novel catabolism-based therapeutic approach for inhibition of cancer progression.