Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Prostate ; 71(13): 1429-40, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21360561

ABSTRACT

BACKGROUND: The transgenic adenocarcinoma of mouse prostate (TRAMP) model is by far the most practical transgenic model for preclinical prostate cancer chemoprevention studies. It is critical to characterize the prostate lobe-specificity of lesion lineages to consolidate the advantages of this model and minimize its limitations for chemoprevention studies. METHODS: We dissected dorsolateral (DLP), ventral (VP), and anterior prostate (AP) lobes, and macroscopic tumors from 90 male C57BL/6J TRAMP mice at 22-24 weeks of age (WOA) and analyzed lesions by histological, biochemical and proteomic approaches. To determine whether methylseleninic acid (MSeA) led to a deletion of initiated cells, we gave oral MSeA to TRAMP mice from 5 to 23 WOA or from 5 to 15 WOA and analyzed lesions at 23 WOA. RESULTS: All tumors (n = 18) were T-antigen(+), synaptophysin (SYP)(+), androgen-receptor(-), and E-cadherin(-) poorly differentiated neuroendocrine carcinomas (NE-Ca). They were traceable most frequently to VP (66.7%) and rarely to DLP (11.1%) and AP (5.6%) with an estimated life-time incidence of 1 out of 3 mice. In DLP, epithelial lesions ranged from mild-to-severe atypical hyperplasia, with T-antigen(+), SYP(-), androgen-receptor(+), and E-cadherin(+). Proteomic profiling revealed many molecular differences between VP and DLP. In MSeA experiment, 6 out of 19 (31.5%) mice developed NE-Ca in the control group, only 2 in each MSeA group of 17-18 mice (11.1-11.8%) bore a detectable NE-Ca. CONCLUSION: The C57BL/6J TRAMP mouse represents at least two lineages of prostate carcinogenesis. Chemoprevention studies should incorporate this knowledge for efficacy assessment and molecular target validations.


Subject(s)
Adenocarcinoma/prevention & control , Anticarcinogenic Agents/therapeutic use , Cysteine/analogs & derivatives , Organoselenium Compounds/therapeutic use , Prostatic Neoplasms/prevention & control , Adenocarcinoma/etiology , Adenocarcinoma/pathology , Animals , Cell Lineage , Cysteine/therapeutic use , Disease Models, Animal , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis , Prostatic Neoplasms/etiology , Prostatic Neoplasms/pathology , Proteomics , Selenocysteine/analogs & derivatives , Seminal Vesicles/pathology
2.
Cancer Prev Res (Phila) ; 3(7): 885-95, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20587701

ABSTRACT

Nonsteroidal anti-inflammatory drugs including sulindac are well documented to be highly effective for cancer chemoprevention. However, their cyclooxygenase (COX)-inhibitory activities cause severe gastrointestinal, renal, and cardiovascular toxicities, limiting their chronic use. Recent studies suggest that COX-independent mechanisms may be responsible for the chemopreventive benefits of nonsteroidal anti-inflammatory drugs and support the potential for the development of a novel generation of sulindac derivatives lacking COX inhibition for cancer chemoprevention. A prototypic sulindac derivative with a N,N-dimethylammonium substitution called sulindac sulfide amide (SSA) was recently identified to be devoid of COX-inhibitory activity yet displays much more potent tumor cell growth-inhibitory activity in vitro compared with sulindac sulfide. In this study, we investigated the androgen receptor (AR) signaling pathway as a potential target for its COX-independent antineoplastic mechanism and evaluated its chemopreventive efficacy against prostate carcinogenesis using the transgenic adenocarcinoma of mouse prostate model. The results showed that SSA significantly suppressed the growth of human and mouse prostate cancer cells expressing AR in strong association with G(1) arrest, and decreased AR level and AR-dependent transactivation. Dietary SSA consumption dramatically attenuated prostatic growth and suppressed AR-dependent glandular epithelial lesion progression through repressing cell proliferation in the transgenic adenocarcinoma of mouse prostate mice, whereas it did not significantly affect neuroendocrine carcinoma growth. Overall, the results suggest that SSA may be a chemopreventive candidate against prostate glandular epithelial carcinogenesis.


Subject(s)
Adenocarcinoma/prevention & control , Anticarcinogenic Agents/therapeutic use , Prostatic Neoplasms/prevention & control , Sulindac/analogs & derivatives , Adenocarcinoma/genetics , Animals , Anticarcinogenic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chemoprevention , Disease Models, Animal , Growth Inhibitors/chemistry , Growth Inhibitors/therapeutic use , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Androgen/metabolism , Sulindac/chemistry , Sulindac/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...