[HTML][HTML] ATP citrate lyase knockdown impacts cancer stem cells in vitro

JI Hanai, N Doro, P Seth, VP Sukhatme - Cell death & disease, 2013 - nature.com
JI Hanai, N Doro, P Seth, VP Sukhatme
Cell death & disease, 2013nature.com
ATP citrate lyase (ACL) knockdown (KD) causes tumor suppression and induces
differentiation. We have previously reported that ACL KD reverses epithelial–mesenchymal
transition (EMT) in lung cancer cells. Because EMT is often associated with processes that
induce stemness, we hypothesized that ACL KD impacts cancer stem cells. By assessing
tumorsphere formation and expression of stem cell markers, we showed this to be the case
in A549 cells, which harbor a Ras mutation, and in two other non-small-cell lung cancer cell …
Abstract
ATP citrate lyase (ACL) knockdown (KD) causes tumor suppression and induces differentiation. We have previously reported that ACL KD reverses epithelial–mesenchymal transition (EMT) in lung cancer cells. Because EMT is often associated with processes that induce stemness, we hypothesized that ACL KD impacts cancer stem cells. By assessing tumorsphere formation and expression of stem cell markers, we showed this to be the case in A549 cells, which harbor a Ras mutation, and in two other non-small-cell lung cancer cell lines, H1975 and H1650, driven by activating EGFR mutations. Inducible ACL KD had the same effect as stable ACL KD. Similar effects were noted in another well-characterized Ras-induced mammary model system (HMLER). Moreover, treatment with hydroxycitrate phenocopied the effects of ACL KD, suggesting that the enzymatic activity of ACL was critical. Indeed, acetate treatment reversed the ACL KD phenotype. Having previously established that ACL KD impacts signaling through the phosphatidylinositol 3-kinase (PI3K) pathway, not the Ras-mitogen-activated protein kinase (MAPK) pathway, and that EMT can be reversed by PI3K inhibitors, we were surprised to find that stemness in these systems was maintained through Ras-MAPK signaling, and not via PI3K signaling. Snail is a downstream transcription factor impacted by Ras-MAPK signaling and known to promote EMT and stemness. We found that snail expression was reduced by ACL KD. In tumorigenic HMLER cells, ACL overexpression increased snail expression and stemness, both of which were reduced by ACL KD. Furthermore, ACL could not initiate either tumorigenesis or stemness by itself. ACL and snail proteins interacted and ACL expression regulated the transcriptional activity of snail. Finally, ACL KD counteracted stem cell characteristics induced in diverse cell systems driven by activation of pathways outside of Ras-MAPK signaling. Our findings unveil a novel aspect of ACL function, namely its impact on cancer stemness in a broad range of genetically diverse cell types.
nature.com