scholarly journals HIF-1-Mediated Suppression of Acyl-CoA Dehydrogenases and Fatty Acid Oxidation Is Critical for Cancer Progression

Cell Reports ◽  
2014 ◽  
Vol 8 (6) ◽  
pp. 1930-1942 ◽  
Author(s):  
De Huang ◽  
Tingting Li ◽  
Xinghua Li ◽  
Long Zhang ◽  
Linchong Sun ◽  
...  
2020 ◽  
Author(s):  
Benjamin Guy Bitler ◽  
Brandon Sawyer ◽  
Lubna Qamar ◽  
Jennifer K. Richer ◽  
Kian Behbakht ◽  
...  

2021 ◽  
Author(s):  
Bhuban Ruidas ◽  
Tapas Kumar Sur ◽  
Chitrangada Das Mukhopadhyay ◽  
Koel Sinha ◽  
Sutapa Som Chaudhury ◽  
...  

Abstract Recent evidence concreted that maximum energy in metastatic breast cancer progression is supplied by fatty acid oxidation (FAO) governed by a rate-limiting enzyme, carnitine palmitoyltransferase 1 (CPT1). Therefore, active limitation of FAO could be an emerging aspect to inhibit breast cancer progression. Herein, for the first time we have introduced Quercetin (QT) from a non-dietary source (Mikania micrantha Kunth) to seize the FAO in triple-negative breast cancer cells (TNBC) through an active targeting of CPT1. Apart from successive molecular quantification, QT has resulted a significant reduction in the intracellular mitochondrial respiration and glycolytic function limiting extensive ATP production. In turn, QT has elevated the reactive oxygen species (ROS) and depleted antioxidant level to induce anti-metastatic and cell apoptosis activities. Real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) investigated the FAO associated gene expression resulting significant depletion in FAO which were further confirmed through the successful in-silico molecular docking prediction for active binding potentiality of QT to CPT1. Subsequently, QT has shown an excellent in-vivo antitumor activities through the altered lipid profile and oxidative stress healing capabilities in female breast cancer BALB/c mice model. Therefore, all the obtained data significantly grounded the fact that QT could be a promising metabolism-targeted breast cancer therapeutics.


2020 ◽  
Vol 18 (7) ◽  
pp. 1088-1098 ◽  
Author(s):  
Brandon T. Sawyer ◽  
Lubna Qamar ◽  
Tomomi M. Yamamoto ◽  
Alexandra McMellen ◽  
Zachary L. Watson ◽  
...  

2020 ◽  
Vol 3 (7) ◽  
pp. e202000683 ◽  
Author(s):  
Ji Hye Yang ◽  
Nam Hee Kim ◽  
Jun Seop Yun ◽  
Eunae Sandra Cho ◽  
Yong Hoon Cha ◽  
...  

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.


2019 ◽  
Author(s):  
Helena Urquijo ◽  
Emma N Panting ◽  
Roderick N Carter ◽  
Emma J Agnew ◽  
Caitlin S Wyrwoll ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document