Raloxifene-induced myeloma cell apoptosis: a study of nuclear factor-kappaB inhibition and gene expression signature.

Details

Serval ID
serval:BIB_B76BB4F6C402
Type
Article: article from journal or magazin.
Collection
Publications
Title
Raloxifene-induced myeloma cell apoptosis: a study of nuclear factor-kappaB inhibition and gene expression signature.
Journal
Molecular Pharmacology
Author(s)
Olivier S., Close P., Castermans E., de Leval L., Tabruyn S., Chariot A., Malaise M., Merville M.P., Bours V., Franchimont N.
ISSN
0026-895X[print], 0026-895X[linking]
Publication state
Published
Issued date
2006
Volume
69
Number
5
Pages
1615-1623
Language
english
Abstract
Because multiple myeloma remains associated with a poor prognosis, novel drugs targeting specific signaling pathways are needed. The efficacy of selective estrogen receptor modulators for the treatment of multiple myeloma is not well documented. In the present report, we studied the antitumor activity of raloxifene, a selective estrogen receptor modulator, on multiple myeloma cell lines. Raloxifene effects were assessed by tetrazolium salt reduction assay, cell cycle analysis, and Western blotting. Mobility shift assay, immunoprecipitation, chromatin immunoprecipitation assay, and gene expression profiling were performed to characterize the mechanisms of raloxifene-induced activity. Indeed, raloxifene, as well as tamoxifen, decreased JJN-3 and U266 myeloma cell viability and induced caspase-dependent apoptosis. Raloxifene and tamoxifen also increased the cytotoxic response to vincristine and arsenic trioxide. Moreover, raloxifene inhibited constitutive nuclear factor-kappaB (NF-kappaB) activity in myeloma cells by removing p65 from its binding sites through estrogen receptor alpha interaction with p65. It is noteworthy that microarray analysis showed that raloxifene treatment decreased the expression of known NF-kappaB-regulated genes involved in myeloma cell survival and myeloma-induced bone lesions (e.g., c-myc, mip-1alpha, hgf, pac1,...) and induced the expression of a subset of genes regulating cellular cycle (e.g., p21, gadd34, cyclin G2,...). In conclusion, raloxifene induces myeloma cell cycle arrest and apoptosis partly through NF-kappaB-dependent mechanisms. These findings also provide a transcriptional profile of raloxifene treatment on multiple myeloma cells, offering the framework for future studies of selective estrogen receptor modulators therapy in multiple myeloma.
Keywords
Apoptosis/drug effects, Cell Cycle/drug effects, Cell Line, Tumor, Estrogen Receptor alpha/drug effects, Estrogen Receptor alpha/genetics, Estrogen Receptor beta/drug effects, Estrogen Receptor beta/genetics, Gene Expression Regulation, Neoplastic/drug effects, Humans, Multiple Myeloma, NF-kappa B/antagonists & inhibitors, NF-kappa B/metabolism, Polymerase Chain Reaction, Raloxifene/pharmacology, Selective Estrogen Receptor Modulators/pharmacology
Pubmed
Create date
27/10/2010 10:06
Last modification date
20/08/2019 16:25
Usage data