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.
PLoS One ; 8(11): e79542, 2013.
Article in English | MEDLINE | ID: mdl-24260246

ABSTRACT

ELL-associated factor 2 (EAF2) is an androgen-responsive tumor suppressor frequently deleted in advanced prostate cancer that functions as a transcription elongation factor of RNA Pol II through interaction with the ELL family proteins. EAF2 knockout mice on a 129P2/OLA-C57BL/6J background developed late-onset lung adenocarcinoma, hepatocellular carcinoma, B-cell lymphoma and high-grade prostatic intraepithelial neoplasia. In order to further characterize the role of EAF2 in the development of prostatic defects, the effects of EAF2 loss were compared in different murine strains. In the current study, aged EAF2(-/-) mice on both the C57BL/6J and FVB/NJ backgrounds exhibited mPIN lesions as previously reported on a 129P2/OLA-C57BL/6J background. In contrast to the 129P2/OLA-C57BL/6J mixed genetic background, the mPIN lesions in C57BL/6J and FVB/NJ EAF2(-/-) mice were associated with stromal defects characteristic of a reactive stroma and a statistically significant increase in prostate microvessel density. Stromal inflammation and increased microvessel density was evident in EAF2-deficient mice on a pure C57BL/6J background at an early age and preceded the development of the histologic epithelial hyperplasia and neoplasia found in the prostates of older EAF2(-/-) animals. Mice deficient in EAF2 had an increased recovery rate and a decreased overall response to the effects of androgen deprivation. EAF2 expression in human cancer was significantly down-regulated and microvessel density was significantly increased compared to matched normal prostate tissue; furthermore EAF2 expression was negatively correlated with microvessel density. These results suggest that the EAF2 knockout mouse on the C57BL/6J and FVB/NJ genetic backgrounds provides a model of PIN lesions associated with an altered prostate microvasculature and reactive stromal compartment corresponding to that reported in human prostate tumors.


Subject(s)
Nuclear Proteins/metabolism , Prostatic Intraepithelial Neoplasia/metabolism , Trans-Activators/metabolism , Animals , Cell Line, Tumor , Humans , Immunohistochemistry , In Situ Hybridization , Laser Capture Microdissection , Mice , Mice, Knockout , Mice, Mutant Strains , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Prostatic Intraepithelial Neoplasia/genetics , Trans-Activators/deficiency , Trans-Activators/genetics
2.
Endocrinology ; 154(7): 2296-307, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23671262

ABSTRACT

Androgen deprivation therapy (ADT) is the standard treatment for patients with prostate-specific antigen progression after treatment for localized prostate cancer. An alternative to continuous ADT is intermittent ADT (IADT), which allows recovery of testosterone during off-cycles to stimulate regrowth and differentiation of the regressed prostate tumor. IADT offers patients a reduction in side effects associated with ADT, improved quality of life, and reduced cost with no difference in overall survival. Our previous studies showed that IADT coupled with 5α-reductase inhibitor (5ARI), which blocks testosterone conversion to DHT could prolong survival of animals bearing androgen-sensitive prostate tumors when off-cycle duration was fixed. To further investigate this clinically relevant observation, we measured the time course of testosterone-induced regrowth of regressed LuCaP35 and LNCaP xenograft tumors in the presence or absence of a 5ARI. 5α-Reductase inhibitors suppressed the initial regrowth of regressed prostate tumors. However, tumors resumed growth and were no longer responsive to 5α-reductase inhibition several days after testosterone replacement. This finding was substantiated by bromodeoxyuridine and Ki67 staining of LuCaP35 tumors, which showed inhibition of prostate tumor cell proliferation by 5ARI on day 2, but not day 14, after testosterone replacement. 5α-Reductase inhibitors also suppressed testosterone-stimulated proliferation of LNCaP cells precultured in androgen-free media, suggesting that blocking testosterone conversion to DHT can inhibit prostate tumor cell proliferation via an intracrine mechanism. These results suggest that short off-cycle coupled with 5α-reductase inhibition could maximize suppression of prostate tumor growth and, thus, improve potential survival benefit achieved in combination with IADT.


Subject(s)
5-alpha Reductase Inhibitors/therapeutic use , Prostatic Neoplasms/chemically induced , Prostatic Neoplasms/drug therapy , Testosterone/adverse effects , Animals , Azasteroids/pharmacology , Azasteroids/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Dutasteride , Finasteride/pharmacology , Finasteride/therapeutic use , Humans , Immunohistochemistry , Letrozole , Male , Mice , Mice, Nude , Mice, SCID , Nitriles/pharmacology , Nitriles/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Triazoles/pharmacology , Triazoles/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...