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.
J Dermatol Sci ; 81(3): 182-91, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26775629

ABSTRACT

BACKGROUND: The tumor suppressor p53 controls DNA repair, cell cycle, apoptosis, autophagy and numerous other cellular processes. Imiquimod (IMQ), a synthetic toll-like receptor (TLR) 7 ligand for the treatment of superficial basal cell carcinoma (BCC), eliminates cancer cells by activating cell-mediated immunity and directly inducing apoptosis and autophagy in cancer cells. OBJECTIVE: To evaluate the role of p53 in IMQ-induced cell death in skin cancer cells. METHODS: The expression, phosphorylation and subcellular localization of p53 were detected by real-time PCR, luciferase reporter assay, cycloheximide chase analysis, immunoblotting and immunocytochemistry. Using BCC/KMC1 cell line as a model, the upstream signaling of p53 activation was dissected by over-expression of TLR7/8, the addition of ROS scavenger, ATM/ATR inhibitors and pan-caspase inhibitor. The role of p53 in IMQ-induced apoptosis and autophagy was assessed by genetically silencing p53 and evaluated by a DNA content assay, immunoblotting, LC3 puncta detection and acridine orange staining. RESULTS: IMQ induced p53 mRNA expression and protein accumulation, increased Ser15 phosphorylation, promoted nuclear translocation and up-regulated its target genes in skin cancer cells in a TLR7/8-independent manner. In BCC/KMC1 cells, the induction of p53 by IMQ was achieved through increased ROS production to stimulate the ATM/ATR-Chk1/Chk2 axis but was not mediated by inducing DNA damage. The pharmacological inhibition of ATM/ATR significantly suppressed IMQ-induced p53 activation and apoptosis. Silencing of p53 significantly decreased the IMQ-induced caspase cascade activation and apoptosis but enhanced autophagy. Mutant p53 skin cancer cell lines were more resistant to IMQ-induced apoptosis than wildtype p53 skin cancer cell lines. CONCLUSION: IMQ induced ROS production to stimulate ATM/ATR pathways and contributed to p53-dependent apoptosis in a skin basal cell carcinoma cell line BCC/KMC1.


Subject(s)
Aminoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Basal Cell/drug therapy , Skin Neoplasms/drug therapy , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Autophagy/drug effects , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/metabolism , Carcinoma, Basal Cell/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , Humans , Imiquimod , Mutation , Phosphorylation , RNA Interference , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Time Factors , Transfection , Tumor Suppressor Protein p53/genetics
2.
Toxicol Appl Pharmacol ; 267(1): 113-24, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23274516

ABSTRACT

Compound C, a well-known inhibitor of the intracellular energy sensor AMP-activated protein kinase (AMPK), has been reported to cause apoptotic cell death in myeloma, breast cancer cells and glioma cells. In this study, we have demonstrated that compound C not only induced autophagy in all tested skin cancer cell lines but also caused more apoptosis in p53 wildtype skin cancer cells than in p53-mutant skin cancer cells. Compound C can induce upregulation, phosphorylation and nuclear translocalization of the p53 protein and upregulate expression of p53 target genes in wildtype p53-expressing skin basal cell carcinoma (BCC) cells. The changes of p53 status were dependent on DNA damage which was caused by compound C induced reactive oxygen species (ROS) generation and associated with activated ataxia-telangiectasia mutated (ATM) protein. Using the wildtype p53-expressing BCC cells versus stable p53-knockdown BCC sublines, we present evidence that p53-knockdown cancer cells were much less sensitive to compound C treatment with significant G2/M cell cycle arrest and attenuated the compound C-induced apoptosis but not autophagy. The compound C induced G2/M arrest in p53-knockdown BCC cells was associated with the sustained inactive Tyr15 phosphor-Cdc2 expression. Overall, our results established that compound C-induced apoptosis in skin cancer cells was dependent on the cell's p53 status.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Apoptosis/drug effects , Pyrazoles/antagonists & inhibitors , Pyrimidines/antagonists & inhibitors , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Suppressor Protein p53/physiology , AMP-Activated Protein Kinases/physiology , Apoptosis/physiology , Autophagy/drug effects , Autophagy/physiology , Cell Line, Tumor , Humans , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Skin Neoplasms/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...