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
1.
Oncogene ; 36(46): 6508, 2017 11 16.
Article in English | MEDLINE | ID: mdl-28925393

ABSTRACT

This corrects the article DOI: 10.1038/onc.2016.320.

2.
Oncogene ; 36(11): 1537-1545, 2017 03.
Article in English | MEDLINE | ID: mdl-27694900

ABSTRACT

Ovarian cancer is the leading cause of death among all gynecological malignancies due to the development of acquired chemoresistance and disease relapse. Although the role of cancer stem cells (CSCs), a subset of tumor cells with the self-renewal and differentiation capabilities, in therapeutic resistance is beginning to be better understood, the significance of epigenetic regulatory mechanisms responsible for integrating the stemness with drug resistance remain poorly understood. Here we identified that lysine demethylase KDM3A as a critical regulator of ovarian cancer stemness and cisplatin resistance by inducing the expressions of pluripotent molecules Sox2 and Nanog and anti-apoptotic B-cell lymphoma 2 (Bcl-2), respectively. In addition, KDM3A induces ovarian cancer growth while antagonizing cellular senescence by repressing the expression of cyclin-dependent kinase inhibitor, p21Waf1/Cip1. The underlying mechanism of the noted biological processes include KDM3A-mediated stimulation of Sox2 expression, and demethylating p53 protein and consequently, modulating its target genes such as Bcl-2 and p21Waf1/Cip1 expression. Consistently, KDM3A depletion inhibited the growth of subcutaneously implanted cisplatin-resistant human ovarian cancer cells in athymic nude mice. Moreover, KDM3A is abundantly expressed and positively correlated with Sox2 expression in human ovarian cancer tissues. In brief, our findings reveal a novel mechanism by which KDM3A promotes ovarian CSCs, proliferation and chemoresistance and thus, highlights the significance of KDM3A as a novel therapeutic target for resistant ovarian cancer.


Subject(s)
Drug Resistance, Neoplasm/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine/metabolism , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cisplatin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Models, Animal , Epigenesis, Genetic , Female , Gene Expression , Gene Knockdown Techniques , Heterografts , Humans , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism
3.
Oncogene ; 36(1): 47-59, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27270439

ABSTRACT

Invasive growth and apoptosis resistance of breast cancer cells are associated with metastasis and disease relapse. Here we identified that the lysine-specific demethylase KDM3A played a dual role in breast cancer cell invasion and apoptosis by demethylating histone and the non-histone protein p53, respectively. While inducing pro-invasive genes by erasing repressive histone H3 lysine 9 methylation, KDM3A promotes chemoresistance by demethylating p53. KDM3A suppressed pro-apoptotic functions of p53 by erasing p53-K372me1, as this methylation is crucial for the stability of chromatin-bound p53. Unexpectedly, depletion of KDM3A was capable of reactivating mutated p53 to induce the expression of pro-apoptotic genes in breast cancer with mutant p53. Moreover, KDM3A knockdown also potently inhibited tumorigenic potentials of breast cancer stem-like cells and rendered them sensitive to apoptosis induced by chemotherapeutic drugs. Taken together, our results suggest that KDM3A might be a potential therapeutic target for human breast cancer treatment and prevention.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic/drug effects , Female , Gene Expression , Gene Knockdown Techniques , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Lysine/metabolism , Methylation , Mice , Mutation , Neoplastic Stem Cells/metabolism , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...