当前位置: X-MOL 学术Cell Death Differ. › 论文详情
Our official English website, www.x-mol.net, welcomes your feedback! (Note: you will need to create a separate account there.)
DAPK3 inhibits gastric cancer progression via activation of ULK1-dependent autophagy
Cell Death and Differentiation ( IF 13.7 ) Pub Date : 2020-10-09 , DOI: 10.1038/s41418-020-00627-5
Guan-Man Li 1, 2 , Lei Li 3 , Meng-Qing Li 4 , Xu Chen 5 , Qiao Su 6 , Zhi-Juan Deng 1, 7 , Hai-Bo Liu 8 , Bin Li 9 , Wen-Hui Zhang 9 , Yong-Xu Jia 10, 11 , Wen-Jian Wang 1 , Jie-Yi Ma 1 , Hai-Liang Zhang 4 , Dan Xie 4 , Xiao-Feng Zhu 4 , Yu-Long He 12, 13 , Xin-Yuan Guan 3, 4 , Jiong Bi 1
Affiliation  

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.



中文翻译:

DAPK3通过激活ULK1依赖性自噬抑制胃癌进展

细胞增殖和细胞死亡之间平衡的失调是恶性肿瘤的核心特征。死亡相关蛋白激酶 3 (DAPK3) 调节程序性细胞死亡,包括细胞凋亡和自噬。我们之前的研究表明,一半以上的胃癌(GCs)中检测到 DAPK3 下调,这与肿瘤侵袭、转移和预后不良有关。然而,DAPK3 介导的肿瘤抑制的确切分子机制仍不清楚。在这里,我们发现 DAPK3 的肿瘤抑制功能依赖于自噬过程。质谱、体外激酶测定和免疫沉淀表明 DAPK3 通过在 Ser556 位点直接磷酸化 ULK1 增加 ULK1 活性。DAPK3 对 ULK1 的磷酸化促进了 ULK1 复合物的形成,VPS34 复合物的激活,和饥饿时的自噬诱导。DAPK3 的激酶活性和 ULK1 Ser556 磷酸化是 DAPK3 调节的肿瘤抑制所必需的。在临床 GC 样本中证实了 DAPK3 与 ULK1 Ser556 磷酸化的协同表达,并且这种共表达与患者的良好生存结果相关。总的来说,这些发现表明 DAPK3 在 GC 中的抑癌作用与自噬有关,并且 DAPK3 是一种新型的自噬调节剂,可以直接磷酸化 ULK1 并激活 ULK1。因此,DAPK3 可能是一个有前途的预后自噬相关标志物。在临床 GC 样本中证实了 DAPK3 与 ULK1 Ser556 磷酸化的协同表达,并且这种共表达与患者的良好生存结果相关。总的来说,这些发现表明 DAPK3 在 GC 中的抑癌作用与自噬有关,并且 DAPK3 是一种新型的自噬调节剂,可以直接磷酸化 ULK1 并激活 ULK1。因此,DAPK3 可能是一个有前途的预后自噬相关标志物。在临床 GC 样本中证实了 DAPK3 与 ULK1 Ser556 磷酸化的协同表达,并且这种共表达与患者的良好生存结果相关。总的来说,这些发现表明 DAPK3 在 GC 中的抑癌作用与自噬有关,并且 DAPK3 是一种新型的自噬调节剂,可以直接磷酸化 ULK1 并激活 ULK1。因此,DAPK3 可能是一个有前途的预后自噬相关标志物。

更新日期:2020-10-11
down
wechat
bug