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








Language
Year range
1.
Experimental & Molecular Medicine ; : 236-240, 2012.
Article in English | WPRIM | ID: wpr-194080

ABSTRACT

We previously reported that the p53 tumor suppressor protein plays an essential role in the induction of tetraploid G1 arrest in response to perturbation of the actin cytoskeleton, termed actin damage. In this study, we investigated the role of p53, ataxia telangiectasia mutated protein (ATM), and catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) in tetraploid G1 arrest induced by actin damage. Treatment with actin-damaging agents including pectenotoxin-2 (PTX-2) increases phosphorylation of Ser-15 and Ser-37 residues of p53, but not Ser-20 residue. Knockdown of ATM and DNA-PKcs do not affect p53 phosphorylation induced by actin damage. However, while ATM knockdown does not affect tetraploid G1 arrest, knockdown of DNA-PKcs not only perturbs tetraploid G1 arrest, but also results in formation of polyploidy and induction of apoptosis. These results indicate that DNA-PKcs is essential for the maintenance of actin damage induced-tetraploid G1 arrest in a p53-independent manner. Furthermore, actin damage-induced p53 expression is not observed in cells synchronized at G1/S of the cell cycle, implying that p53 induction is due to actin damage-induced tetraploidy rather than perturbation of actin cytoskeleton. Therefore, these results suggest that p53 and DNA-PKcs independently function for tetraploid G1 arrest and preventing polyploidy formation.


Subject(s)
Humans , Actins/metabolism , Apoptosis , Catalytic Domain , Cell Cycle Proteins/genetics , Cell Line , Cell Line, Tumor , DNA-Activated Protein Kinase/chemistry , DNA-Binding Proteins/genetics , Furans/pharmacology , G1 Phase , Gene Knockdown Techniques , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Pyrans/pharmacology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
2.
Experimental & Molecular Medicine ; : 389-392, 2011.
Article in English | WPRIM | ID: wpr-102685

ABSTRACT

Cellular senescence is a tumor-suppressive process instigated by proliferation in the absence of telomere replication, by cellular stresses such as oncogene activation, or by activation of the tumor suppressor proteins, such as Rb or p53. This process is characterized by an irreversible cell cycle exit, a unique morphology, and expression of senescence-associated-beta-galactosidase (SA-beta-gal). Despite the potential biological importance of cellular senescence, little is known of the mechanisms leading to the senescent phenotype. p41-Arc has been known to be a putative regulatory component of the mammalian Arp2/3 complex, which is required for the formation of branched networks of actin filaments at the cell cortex. In this study, we demonstrate that p41-Arc can induce senescent phenotypes when it is overexpressed in human tumor cell line, SaOs-2, which is deficient in p53 and Rb tumor suppressor genes, implying that p41 can induce senescence in a p53-independent way. p41-Arc overexpression causes a change in actin filaments, accumulating actin filaments in nuclei. Therefore, these results imply that a change in actin filament can trigger an intrinsic senescence program in the absence of p53 and Rb tumor suppressor genes.


Subject(s)
Humans , Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Cellular Senescence , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Fibroblasts/physiology , Recombinant Proteins/genetics , Retinoblastoma Protein/deficiency , Tumor Suppressor Protein p53/deficiency
3.
Cancer Research and Treatment ; : 224-228, 2006.
Article in English | WPRIM | ID: wpr-115205

ABSTRACT

PURPOSE: Recent studies have suggested that p53 regulates the G2 checkpoint in the cell cycle and this function is required for the maintenance of genomic integrity. In this study, we addressed a role of p53 in escaping from cell cycle G2 arrest following DNA damage. MATERIALS AND METHODS: Cell cycle checkpoint arrest in the human colon cancer cell line HCT116 and its derivatives carry p53 or p21 deletions, were examined by FACS analysis, immunoprecipitation, Western blot and IP-kinase assay. RESULTS: While the cells with functional p53 were arrested at both the G1 and G2 checkpoints, the p53-deficient cells failed to arrest at G1, but they were arrested at G2. However, the p53-deficient cells failed to sustain G2 checkpoint arrest and they entered mitosis earlier than did the p53-positive cells and so this resulted in extensive cell death. Cdc2 kinase becomes reactivated in p53-deficient cells in association with entry into mitosis, but not in the p53-positive cells. Upon DNA damage, the p21-deficient cells, like the p53-negative cells, not only failed to repress cdk2- dependent NF-Y phosphorylation, but they also failed to repress the expression of such cell cycle G2-regulatory genes as cdc2, cyclin B, RNR-R2 and cdc25C, which have all been previously reported as targets of NF-Y transcription factor. CONCLUSION: p53 is essential to prevent immature escaping from cell cycle G2 checkpoint arrest through p21-mediated cdk2 inactivation, and this leads to inhibition of cdk2-dependent NF-Y phosphorylation and NF-Y dependent transcription of the cell cycle G2-rgulatory genes, including cdc2 and cyclin B.


Subject(s)
Humans , Blotting, Western , CCAAT-Binding Factor , CDC2 Protein Kinase , Cell Cycle Checkpoints , Cell Cycle , Cell Death , Cell Line , Colonic Neoplasms , Cyclin B , DNA Damage , G2 Phase , Immunoprecipitation , Mitosis , Phosphorylation , Phosphotransferases , Transcription Factors , Tumor Suppressor Protein p53 , United Nations
SELECTION OF CITATIONS
SEARCH DETAIL