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.
Oncotarget ; 9(4): 5016-5031, 2018 Jan 12.
Article in English | MEDLINE | ID: mdl-29435159

ABSTRACT

The MYC transcription factor coordinates, via different RNA polymerases, the transcription of both ribosomal RNA (rRNA) and protein genes necessary for nucleolar as well as mitochondrial ribogenesis. In this study we tested if MYC-coordination of rRNA transcription in the nucleolus and in the mitochondrion drives (cancer) cell proliferation. Here we show that the anti-proliferative effect of CX-5461, a Pol I inhibitor of rRNA transcription, in ovarian (cancer) cell contexts characterized by MYC overexpression is enhanced either by 2'-C-Methyl Adenosine (2'-C-MeA), a ribonucleoside that inhibits POLRMT mitochondrial rRNA (mt-rRNA) transcription and doxycycline, a tetracycline known to affect mitochondrial translation. Thus, hindering not only mt-rRNA transcription, but also mitoribosome function in MYC-overexpressing ovarian (cancer) cells, potentiates the antiproliferative effect of CX-5461. Targeting MYC-regulated rRNA transcription and ribogenesis in both the nucleolus and mitochondrion seems to be a novel approach worth of consideration for treating MYC-driven cancer.

2.
Cell Cycle ; 15(18): 2515-26, 2016 Sep 16.
Article in English | MEDLINE | ID: mdl-27485818

ABSTRACT

Upregulation of RNA Polymerase (Pol I)-mediated transcription of rRNA and increased ribogenesis are hallmarks of breast cancer. According to several datasets, including The Cancer Genome Atlas (TCGA), amplification/upregulation of genes encoding for basal components of the Pol I transcriptional machinery is frequent at different breast cancer stages. Here we show that knock down of the RNA polymerase I-specific transcription initiation factor RRN3 (TIF-IA) in breast cancer cells is sufficient to reduce rRNA synthesis and inhibit cell proliferation, and second that stable ectopic expression of RRN3 in human mammary epithelial (HME1) cells, by increasing rRNA transcription, confers increased sensitivity to the anti-proliferative effects of a selective Pol I inhibitor. Further, RRN3-overexpressing HME1 cells, when grown in in vitro 3-dimensional (3D) culture, develop into morphologically aberrant acinar structures lacking a lumen and filled with proliferative cells, thus acquiring a morphology resembling in situ ductal breast cancer lesions (DCIS). Consequently, interference with RRN3 control of Pol I transcription seems capable of both compromising mammary epithelial morphogenetic processes at early breast cancer stages, and driving breast cancer progression by fostering proliferation.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Epithelial Cells/pathology , Mammary Glands, Human/pathology , Morphogenesis/genetics , RNA Polymerase I/genetics , Transcription, Genetic , Benzothiazoles/pharmacology , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Gene Knockdown Techniques , Genome, Human , Humans , MCF-7 Cells , Morphogenesis/drug effects , Naphthyridines/pharmacology , Neoplasm Invasiveness , Pol1 Transcription Initiation Complex Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/metabolism , Protein Stability/drug effects , RNA, Ribosomal/genetics , Transcription, Genetic/drug effects , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...