SPOP suppresses prostate cancer through regulation of CYCLIN E1 stability

Cell Death Differ. 2019 Jun;26(6):1156-1168. doi: 10.1038/s41418-018-0198-0. Epub 2018 Sep 20.

Abstract

SPOP is one of the important subunits for CUL3/SPOP/RBX1 complex tightly connected with tumorigenesis. However, its exact roles in different cancers remain debatable. Here, we identify CYCLIN E1, as a novel substrate for SPOP. SPOP directly interacts with CYCLIN E1 and specific regulates its stability in prostate cancer cell lines. SPOP/CUL3/RBX1 complex regulates CYCLIN E1 stability through poly-ubiquitination. CDK2 competes with SPOP for CYCLIN E1 interaction, suggesting that SPOP probably regulates the stability of CDK2-free CYCLIN E1. CYCLIN E1 expression rescued proliferation, migration, and tumor formation of prostate cancer cell suppressed by SPOP. Furthermore, we found SPOP selectively regulates the substrates' stability and signaling pathways in prostate cancer and CCRC cell lines, suggesting that complicated mechanisms exist for SPOP to regulate substrate specificity. Altogether, we have revealed a novel mechanism for SPOP in suppressing prostate cancer and provided evidence to show SPOP has dual functions in prostate cancer and CCRC.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Cell Line
  • Cell Movement
  • Cell Proliferation
  • Cyclin E / genetics
  • Cyclin E / metabolism*
  • Humans
  • Male
  • Nuclear Proteins / metabolism*
  • Oncogene Proteins / genetics
  • Oncogene Proteins / metabolism*
  • Prostatic Neoplasms / metabolism*
  • Prostatic Neoplasms / pathology
  • Protein Stability
  • Repressor Proteins / metabolism*
  • Signal Transduction

Substances

  • CCNE1 protein, human
  • Cyclin E
  • Nuclear Proteins
  • Oncogene Proteins
  • Repressor Proteins
  • SPOP protein, human