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.
Cell Death Differ ; 22(2): 311-22, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25257171

ABSTRACT

The transcription factor adenovirus E2 promoter-binding factor (E2F)-1 normally enhances cell-cycle progression, but it also induces apoptosis under certain conditions, including DNA damage and serum deprivation. Although DNA damage facilitates the phosphorylation and stabilization of E2F1 to trigger apoptosis, how serum starvation renders cells vulnerable to E2F1-induced apoptosis remains unclear. Because poly(ADP-ribose) polymerase 1 (PARP1), a nuclear enzyme essential for genomic stability and chromatin remodeling, interacts directly with E2F1, we investigated the effects of PARP1 on E2F1-mediated functions in the presence and absence of serum. PARP1 attenuation, which increased E2F1 transactivation, induced G2/M cell-cycle arrest under normal growth conditions, but enhanced E2F1-induced apoptosis in serum-starved cells. Interestingly, basal PARP1 activity was sufficient to modify E2F1 by poly(ADP-ribosyl)ation, which stabilized the interaction between E2F1 and the BIN1 tumor suppressor in the nucleus. Accordingly, BIN1 acted as an RB1-independent E2F1 corepressor. Because E2F1 directly activates the BIN1 gene promoter, BIN1 curbed E2F1 activity through a negative-feedback mechanism. Conversely, when the BIN1-E2F1 interaction was abolished by PARP1 suppression, E2F1 continuously increased BIN1 levels. This is functionally germane, as PARP1-depletion-associated G2/M arrest was reversed by the transfection of BIN1 siRNA. Moreover, PARP-inhibitor-associated anti-transformation activity was compromised by the coexpression of dominant-negative BIN1. Because serum starvation massively reduced the E2F1 poly(ADP-ribosyl)ation, we conclude that the release of BIN1 from hypo-poly(ADP-ribosyl)ated E2F1 is a mechanism by which serum starvation promotes E2F1-induced apoptosis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenosine Diphosphate/metabolism , Apoptosis , E2F1 Transcription Factor/metabolism , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Tumor Suppressor Proteins/metabolism , 3T3 Cells , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Division , Cell Line , DNA Damage , E2F1 Transcription Factor/genetics , Humans , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Promoter Regions, Genetic , Tumor Suppressor Proteins/genetics
2.
Br J Cancer ; 110(3): 656-67, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24281001

ABSTRACT

BACKGROUND: Histone deacetylase inhibitors (HDACi) are promising antineoplastic agents, but their precise mechanisms of actions are not well understood. In particular, the relevance of p53 for HDACi-induced effects has not been fully elucidated. We investigated the anticancer effects of four structurally distinct HDACi, vorinostat, entinostat, apicidin and valproic acid, using isogenic HCT-116 colon cancer cell lines differing in p53 status. METHODS: Effects were assessed by MTT assay, flow-cytometric analyses of propidium iodide uptake, mitochondrial depolarisation and cell-cycle distribution, as well as by gene expression profiling. RESULTS: Vorinostat was equally effective in p53 wild-type and null cells, whereas entinostat was less effective in p53 null cells. Histone deacetylase inhibitors treatment suppressed the expression of MDM2 and increased the abundance of p53. Combination treatments showed that vorinostat enhanced the cytotoxic activity of TRAIL and bortezomib, independent of the cellular p53 status. Investigations into the effects of an inhibitor of the sirtuin class of HDAC, tenovin-1, revealed that tenovin-1-mediated cell death hinged on p53. CONCLUSION: These results demonstrate that vorinostat activates p53, but does not require p53 for inducing its anticancer action. Yet they also demonstrate that entinostat-induced cytotoxic effects partially depend on p53, indicating that different HDACi have a different requirement for p53.


Subject(s)
Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Histone Deacetylase Inhibitors/administration & dosage , Tumor Suppressor Protein p53/metabolism , Benzamides/administration & dosage , Colonic Neoplasms/pathology , Flow Cytometry , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Hydroxamic Acids/administration & dosage , Peptides, Cyclic/administration & dosage , Proto-Oncogene Proteins c-mdm2 , Pyridines/administration & dosage , Tumor Suppressor Protein p53/genetics , Valproic Acid/administration & dosage , Vorinostat
SELECTION OF CITATIONS
SEARCH DETAIL
...