4.7 Article

DAPK3 inhibits gastric cancer progression via activation of ULK1-dependent autophagy

Journal

CELL DEATH AND DIFFERENTIATION
Volume 28, Issue 3, Pages 952-967

Publisher

SPRINGERNATURE
DOI: 10.1038/s41418-020-00627-5

Keywords

-

Funding

  1. National Natural Science Foundation of China [81072047, 81302079]
  2. Science and Technology Planning Project of Guangdong Province, China [2016A030303005, 2019A030317005]

Ask authors/readers for more resources

The study demonstrates that tumor-suppressive function of DAPK3 is dependent on autophagy process through direct phosphorylation of ULK1 at Ser556, enhancing ULK1 complex formation and VPS34 complex activation. Clinical samples also show that coordinated expression of DAPK3 with ULK1 Ser556 phosphorylation is associated with favorable survival outcomes in gastric cancer patients. This suggests that DAPK3 may serve as a promising prognostic autophagy-associated marker in gastric cancer.
Dysregulation of the balance between cell proliferation and cell death is a central feature of malignances. Death-associated protein kinase 3 (DAPK3) regulates programmed cell death including apoptosis and autophagy. Our previous study showed that DAPK3 downregulation was detected in more than half of gastric cancers (GCs), which was related to tumor invasion, metastasis, and poor prognosis. However, the precise molecular mechanism underlying DAPK3-mediated tumor suppression remains unclear. Here, we showed that the tumor suppressive function of DAPK3 was dependent on autophagy process. Mass spectrometry, in vitro kinase assay, and immunoprecipitation revealed that DAPK3 increased ULK1 activity by direct ULK1 phosphorylation at Ser556. ULK1 phosphorylation by DAPK3 facilitates the ULK1 complex formation, the VPS34 complex activation, and autophagy induction upon starvation. The kinase activity of DAPK3 and ULK1 Ser556 phosphorylation were required for DAPK3-modulated tumor suppression. The coordinate expression of DAPK3 with ULK1 Ser556 phosphorylation was confirmed in clinical GC samples, and this co-expression was correlated with favorable survival outcomes in patients. Collectively, these findings indicate that the tumor-suppressor roles of DAPK3 in GC are associated with autophagy and that DAPK3 is a novel autophagy regulator, which can directly phosphorylate ULK1 and activate ULK1. Thus, DAPK3 might be a promising prognostic autophagy-associated marker.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available