Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 39(40): 6370-6386, 2020 10.
Article in English | MEDLINE | ID: mdl-32848212

ABSTRACT

For many tumor types chemotherapy still represents the therapy of choice and many standard treatments are based on the use of platinum (PT) drugs. However, de novo or acquired resistance to platinum is frequent and leads to disease progression. In Epithelial Ovarian Cancer (EOC) patients, PT-resistant recurrences are very common and improving the response to treatment still represents an unmet clinical need. To identify new modulators of PT-sensitivity, we performed a loss-of-function screening targeting 680 genes potentially involved in the response of EOC cells to platinum. We found that SGK2 (Serum-and Glucocorticoid-inducible kinase 2) plays a key role in PT-response. We show here that EOC cells relay on the induction of autophagy to escape PT-induced death and that SGK2 inhibition increases PT sensitivity inducing a block in the autophagy cascade due to the impairment of lysosomal acidification. Mechanistically we demonstrate that SGK2 controls autophagy in a kinase-dependent manner by binding and inhibiting the V-ATPase proton pump. Accordingly, SGK2 phosphorylates the subunit V1H (ATP6V1H) of V-ATPase and silencing or chemical inhibition of SGK2, affects the normal autophagic flux and sensitizes EOC cells to platinum. Hence, we identified a new pathway that links autophagy to the survival of cancer cells under platinum treatment in which the druggable kinase SGK2 plays a central role. Our data suggest that blocking autophagy via SGK2 inhibition could represent a novel therapeutic strategy to improve patients' response to platinum.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy/drug effects , Carcinoma, Ovarian Epithelial/drug therapy , Immediate-Early Proteins/metabolism , Ovarian Neoplasms/drug therapy , Protein Serine-Threonine Kinases/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Benzoates/pharmacology , Benzoates/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Carboplatin/pharmacology , Carboplatin/therapeutic use , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Female , Humans , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/metabolism , Vacuolar Proton-Translocating ATPases/metabolism
2.
J Mol Cell Biol ; 8(1): 17-30, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26450989

ABSTRACT

Exposure of normal and tumor-derived cells to TGFß results in different outcomes, depending on the regulation of key targets. The CDK inhibitor p27(Kip1) is one of these TGFß targets and is essential for the TGFß-induced cell cycle arrest. TGFß treatment inhibits p27(Kip1) degradation and induces its nuclear translocation, through mechanisms that are still unknown. Recent evidences suggest that SUMOylation, a post-translational modification able to modulate the stability and subcellular localization of target proteins, critically modifies members of the TGFß signaling pathway. Here, we demonstrate that p27(Kip1) is SUMOylated in response to TGFß treatment. Using different p27(Kip1) point mutants, we identified lysine 134 (K134) as the residue modified by small ubiquitin-like modifier 1 (SUMO1) in response to TGFß treatment. TGFß-induced K134 SUMOylation increased protein stability and nuclear localization of both endogenous and exogenously expressed p27(Kip1). We observed that SUMOylation regulated p27(Kip1) binding to CDK2, thereby governing its nuclear proteasomal degradation through the phosphorylation of threonine 187. Importantly, p27(Kip1) SUMOylation was necessary for proper cell cycle exit following TGFß treatment. These data indicate that SUMOylation is a novel regulatory mechanism that modulates p27(Kip1) function in response to TGFß stimulation. Given the involvement of TGFß signaling in cancer cell proliferation and invasion, our data may shed light on an important aspect of this pathway during tumor progression.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Sumoylation/physiology , Cell Division/genetics , Cell Division/physiology , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Mass Spectrometry , Phosphorylation/genetics , Phosphorylation/physiology , Protein Processing, Post-Translational , Protein Stability , Sumoylation/genetics , Transforming Growth Factor beta
SELECTION OF CITATIONS
SEARCH DETAIL
...