Subcellular localization of glycogen synthase kinase 3β controls embryonic stem cell self-renewal

M Bechard, S Dalton - Molecular and cellular biology, 2009 - Taylor & Francis
M Bechard, S Dalton
Molecular and cellular biology, 2009Taylor & Francis
Phosphatidylinositol 3-kinase (PI3K), protein kinase B (AKT1), and c-myc have well-
established roles in promoting the maintenance of murine embryonic stem cells (mESCs). In
contrast, the activity of glycogen synthase kinase 3β (GSK3β), a negatively regulated target
of AKT1 signaling, antagonizes self-renewal. Here, we show that PI3K/AKT1 signaling
promotes self-renewal by suppressing GSK3β activity and restricting its access to nuclear
substrates such as c-myc. GSK3β shuttles between the cytoplasm and nucleus in mESCs …
Phosphatidylinositol 3-kinase (PI3K), protein kinase B (AKT1), and c-myc have well-established roles in promoting the maintenance of murine embryonic stem cells (mESCs). In contrast, the activity of glycogen synthase kinase 3β (GSK3β), a negatively regulated target of AKT1 signaling, antagonizes self-renewal. Here, we show that PI3K/AKT1 signaling promotes self-renewal by suppressing GSK3β activity and restricting its access to nuclear substrates such as c-myc. GSK3β shuttles between the cytoplasm and nucleus in mESCs but accumulates in the cytoplasm in an inactive form due to AKT1-dependent nuclear export and inhibitory phosphorylation. When PI3K/AKT1 signaling declines following leukemia inhibitory factor withdrawal, active GSK3β accumulates in the nucleus, where it targets c-myc through phosphorylation on threonine 58 (T58), promoting its degradation. Ectopic nuclear localization of active GSK3β promotes differentiation, but this process is blocked by a mutant form of c-myc (T58A) that evades phosphorylation by GSK3β. This novel mechanism explains how AKT1 promotes self-renewal by regulating the activity and localization of GSK3β. This pathway converges on c-myc, a key regulator of mESC self-renewal.
Taylor & Francis Online