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.
Mol Cancer ; 9: 258, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20863401

ABSTRACT

BACKGROUND: Prostate cancer (PCa) cells preferentially metastasize to bone at least in part by acquiring osteomimetic properties. Runx2, an osteoblast master transcription factor, is aberrantly expressed in PCa cells, and promotes their metastatic phenotype. The transcriptional programs regulated by Runx2 have been extensively studied during osteoblastogenesis, where it activates or represses target genes in a context-dependent manner. However, little is known about the gene regulatory networks influenced by Runx2 in PCa cells. We therefore investigated genome wide mRNA expression changes in PCa cells in response to Runx2. RESULTS: We engineered a C4-2B PCa sub-line called C4-2B/Rx2 dox, in which Doxycycline (Dox) treatment stimulates Runx2 expression from very low to levels observed in other PCa cells. Transcriptome profiling using whole genome expression array followed by in silico analysis indicated that Runx2 upregulated a multitude of genes with prominent cancer associated functions. They included secreted factors (CSF2, SDF-1), proteolytic enzymes (MMP9, CST7), cytoskeleton modulators (SDC2, Twinfilin, SH3PXD2A), intracellular signaling molecules (DUSP1, SPHK1, RASD1) and transcription factors (Sox9, SNAI2, SMAD3) functioning in epithelium to mesenchyme transition (EMT), tissue invasion, as well as homing and attachment to bone. Consistent with the gene expression data, induction of Runx2 in C4-2B cells enhanced their invasiveness. It also promoted cellular quiescence by blocking the G1/S phase transition during cell cycle progression. Furthermore, the cell cycle block was reversed as Runx2 levels declined after Dox withdrawal. CONCLUSIONS: The effects of Runx2 in C4-2B/Rx2 dox cells, as well as similar observations made by employing LNCaP, 22RV1 and PC3 cells, highlight multiple mechanisms by which Runx2 promotes the metastatic phenotype of PCa cells, including tissue invasion, homing to bone and induction of high bone turnover. Runx2 is therefore an attractive target for the development of novel diagnostic, prognostic and therapeutic approaches to PCa management. Targeting Runx2 may prove more effective than focusing on its individual downstream genes and pathways.


Subject(s)
Bone Neoplasms/secondary , Core Binding Factor Alpha 1 Subunit/metabolism , Prostatic Neoplasms/complications , Prostatic Neoplasms/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Apoptosis/genetics , Apoptosis/physiology , Biomarkers, Tumor/genetics , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation , Chemokine CXCL12/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Cystatins/genetics , Dual Specificity Phosphatase 1/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Male , Matrix Metalloproteinase 9/genetics , Microfilament Proteins/genetics , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/genetics , Protein-Tyrosine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Syndecan-2/genetics
2.
Blood ; 116(19): 3964-71, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20716776

ABSTRACT

Krox20/EGR2, one of the 4 early growth response genes, is a highly conserved transcription factor implicated in hindbrain development, peripheral nerve myelination, tumor suppression, and monocyte/macrophage cell fate determination. Here, we established a novel role for Krox20 in postnatal skeletal metabolism. Microcomputed tomographic analysis of 4- and 8-week-old mice revealed a low bone mass phenotype (LBM) in both the distal femur and the vertebra of Krox20(+/-) mice. This was attributable to accelerated bone resorption as demonstrated in vivo by increased osteoclast number and serum C-terminal telopeptides, a marker for collagen degradation. Krox20 haploinsufficiency did not reduce bone formation in vivo, nor did it compromise osteoblast differentiation in vitro. In contrast, growth and differentiation were significantly stimulated in preosteoclast cultures derived from Krox20(+/-) splenocytes, suggesting that the LBM is attributable to Krox20 haploinsufficiency in the monocytic lineage. Furthermore, Krox20 silencing in preosteoclasts increased cFms expression and response to macrophage colony-stimulating factor, leading to a cell-autonomous stimulation of cell-cycle progression. Our data indicate that the antimitogenic role of Krox20 in preosteoclasts is the predominant mechanism underlying the LBM phenotype of Krox20-deficient mice. Stimulation of Krox20 expression in preosteoclasts may present a viable therapeutic strategy for high-turnover osteoporosis.


Subject(s)
Bone and Bones/metabolism , Early Growth Response Protein 2/deficiency , Monocytes/cytology , Monocytes/metabolism , Osteoporosis/etiology , Animals , Base Sequence , Bone Resorption/etiology , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Cycle , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation , DNA Primers/genetics , Disease Models, Animal , Early Growth Response Protein 2/genetics , Female , Haploinsufficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , RNA, Small Interfering/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...