2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Emodin down-regulates androgen receptor and inhibits prostate cancer cell growth.

      Cancer research
      Androgen Receptor Antagonists, Animals, COS Cells, Cell Growth Processes, drug effects, Cell Line, Tumor, Cell Nucleus, metabolism, Cercopithecus aethiops, Down-Regulation, Emodin, pharmacology, Gene Expression Regulation, Neoplastic, HSP90 Heat-Shock Proteins, Humans, Male, Mice, Mice, Transgenic, Nuclear Proteins, Prostatic Neoplasms, drug therapy, genetics, pathology, Proto-Oncogene Proteins, Proto-Oncogene Proteins c-mdm2, Receptors, Androgen, biosynthesis, Transcriptional Activation, Transfection, Ubiquitin, Xenograft Model Antitumor Assays

      Read this article at

      ScienceOpenPublisherPubMed
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Hormone-refractory relapse is an inevitable and lethal event for advanced prostate cancer patients after hormone deprivation. A growing body of evidence indicates that hormone deprivation may promote this aggressive prostate cancer phenotype. Notably, androgen receptor (AR) not only mediates the effect of androgen on the tumor initiation but also plays the major role in the relapse transition. This provides a strong rationale for searching new effective agents targeting the down-regulation of AR to treat or prevent advanced prostate cancer progression. Here, we show that emodin, a natural compound, can directly target AR to suppress prostate cancer cell growth in vitro and prolong the survival of C3(1)/SV40 transgenic mice in vivo. Emodin treatment resulted in repressing androgen-dependent transactivation of AR by inhibiting AR nuclear translocation. Emodin decreased the association of AR and heat shock protein 90 and increased the association of AR and MDM2, which in turn induces AR degradation through proteasome-mediated pathway in a ligand-independent manner. Our work indicates a new mechanism for the emodin-mediated anticancer effect and justifies further investigation of emodin as a therapeutic and preventive agent for prostate cancer.

          Related collections

          Author and article information

          Comments

          Comment on this article