Snail1 is stabilized by O‐GlcNAc modification in hyperglycaemic condition

SY Park, HS Kim, NH Kim, S Ji, SY Cha, JG Kang… - The EMBO …, 2010 - embopress.org
SY Park, HS Kim, NH Kim, S Ji, SY Cha, JG Kang, I Ota, K Shimada, N Konishi, HW Nam
The EMBO journal, 2010embopress.org
Protein O‐phosphorylation often occurs reciprocally with O‐GlcNAc modification and
represents a regulatory principle for proteins. O‐phosphorylation of serine by glycogen
synthase kinase‐3β on Snail1, a transcriptional repressor of E‐cadherin and a key regulator
of the epithelial–mesenchymal transition (EMT) programme, results in its proteasomal
degradation. We show that by suppressing O‐phosphorylation‐mediated degradation, O‐
GlcNAc at serine112 stabilizes Snail1 and thus increases its repressor function, which in …
Protein O‐phosphorylation often occurs reciprocally with O‐GlcNAc modification and represents a regulatory principle for proteins. O‐phosphorylation of serine by glycogen synthase kinase‐3β on Snail1, a transcriptional repressor of E‐cadherin and a key regulator of the epithelial–mesenchymal transition (EMT) programme, results in its proteasomal degradation. We show that by suppressing O‐phosphorylation‐mediated degradation, O‐GlcNAc at serine112 stabilizes Snail1 and thus increases its repressor function, which in turn attenuates E‐cadherin mRNA expression. Hyperglycaemic condition enhances O‐GlcNAc modification and initiates EMT by transcriptional suppression of E‐cadherin through Snail1. Thus, dynamic reciprocal O‐phosphorylation and O‐GlcNAc modification of Snail1 constitute a molecular link between cellular glucose metabolism and the control of EMT.
embopress.org