Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
2.
Oncotarget ; 7(28): 44142-44160, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27283898

ABSTRACT

Oncogenic Ras signalling occurs frequently in many human cancers. However, no effective targeted therapies are currently available to treat patients suffering from Ras-driven tumours. Therefore, it is imperative to identify downstream effectors of Ras signalling that potentially represent promising new therapeutic options. Particularly, considering that autophagy inhibition can impair the survival of Ras-transformed cells in tissue culture and mouse models, an understanding of factors regulating the balance between autophagy and apoptosis in Ras-transformed human cells is needed. Here, we report critical roles of the STK38 protein kinase in oncogenic Ras transformation. STK38 knockdown impaired anoikis resistance, anchorage-independent soft agar growth, and in vivo xenograft growth of Ras-transformed human cells. Mechanistically, STK38 supports Ras-driven transformation through promoting detachment-induced autophagy. Even more importantly, upon cell detachment STK38 is required to sustain the removal of damaged mitochondria by mitophagy, a selective autophagic process, to prevent excessive mitochondrial reactive oxygen species production that can negatively affect cancer cell survival. Significantly, knockdown of PINK1 or Parkin, two positive regulators of mitophagy, also impaired anoikis resistance and anchorage-independent growth of Ras-transformed human cells, while knockdown of USP30, a negative regulator of PINK1/Parkin-mediated mitophagy, restored anchorage-independent growth of STK38-depleted Ras-transformed human cells. Therefore, our findings collectively reveal novel molecular players that determine whether Ras-transformed human cells die or survive upon cell detachment, which potentially could be exploited for the development of novel strategies to target Ras-transformed cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , Mitophagy/genetics , Protein Serine-Threonine Kinases/genetics , ras Proteins/genetics , Animals , Anoikis/genetics , Apoptosis/genetics , Autophagy/genetics , Cell Line , Cell Line, Tumor , HCT116 Cells , HEK293 Cells , Humans , Mice, Nude , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Transplantation, Heterologous , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , ras Proteins/metabolism
3.
Curr Biol ; 25(19): 2479-92, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26387716

ABSTRACT

Autophagy plays key roles in development, oncogenesis, cardiovascular, metabolic, and neurodegenerative diseases. Hence, understanding how autophagy is regulated can reveal opportunities to modify autophagy in a disease-relevant manner. Ideally, one would want to functionally define autophagy regulators whose enzymatic activity can potentially be modulated. Here, we describe the STK38 protein kinase (also termed NDR1) as a conserved regulator of autophagy. Using STK38 as bait in yeast-two-hybrid screens, we discovered STK38 as a novel binding partner of Beclin1, a key regulator of autophagy. By combining molecular, cell biological, and genetic approaches, we show that STK38 promotes autophagosome formation in human cells and in Drosophila. Upon autophagy induction, STK38-depleted cells display impaired LC3B-II conversion; reduced ATG14L, ATG12, and WIPI-1 puncta formation; and significantly decreased Vps34 activity, as judged by PI3P formation. Furthermore, we observed that STK38 supports the interaction of the exocyst component Exo84 with Beclin1 and RalB, which is required to initiate autophagosome formation. Upon studying the activation of STK38 during autophagy induction, we found that STK38 is stimulated in a MOB1- and exocyst-dependent manner. In contrast, RalB depletion triggers hyperactivation of STK38, resulting in STK38-dependent apoptosis under prolonged autophagy conditions. Together, our data establish STK38 as a conserved regulator of autophagy in human cells and flies. We also provide evidence demonstrating that STK38 and RalB assist the coordination between autophagic and apoptotic events upon autophagy induction, hence further proposing a role for STK38 in determining cellular fate in response to autophagic conditions.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis/physiology , Beclin-1 , Cell Line, Tumor , Cells, Cultured , Drosophila , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , Protein Binding , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...