PUBLICATION

The von Hippel-Lindau tumor suppressor regulates programmed cell death 5-mediated degradation of Mdm2

Authors
Essers, P.B., Klasson, T.D., Pereboom, T.C., Mans, D.A., Nicastro, M., Boldt, K., Giles, R.H., and MacInnes, A.W.
ID
ZDB-PUB-140402-5
Date
2015
Source
Oncogene   146(3): 789-800.e8 (Journal)
Registered Authors
Pereboom, Tamara
Keywords
none
MeSH Terms
  • Animals
  • Apoptosis Regulatory Proteins/genetics*
  • Apoptosis Regulatory Proteins/metabolism
  • Carcinoma, Renal Cell/genetics*
  • Carcinoma, Renal Cell/pathology
  • Cell Nucleus/genetics
  • DNA Damage/genetics
  • Humans
  • Inhibitor of Apoptosis Proteins/biosynthesis
  • Neoplasm Proteins/genetics*
  • Neoplasm Proteins/metabolism
  • Proteolysis
  • Proto-Oncogene Proteins c-mdm2/metabolism*
  • Tumor Suppressor Protein p53/genetics*
  • Ubiquitin-Protein Ligases/genetics
  • Ubiquitin-Protein Ligases/metabolism
  • Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors
  • Von Hippel-Lindau Tumor Suppressor Protein/biosynthesis
  • Von Hippel-Lindau Tumor Suppressor Protein/genetics*
  • Zebrafish
  • Zebrafish Proteins/biosynthesis
PubMed
24469044 Full text @ Oncogene
Abstract

Functional loss of the von Hippel–Lindau (VHL) tumor suppressor protein (pVHL), which is part of an E3-ubiquitin ligase complex, initiates most inherited and sporadic clear-cell renal cell carcinomas (ccRCC). Genetic inactivation of the TP53 gene in ccRCC is rare, suggesting that an alternate mechanism alleviates the selective pressure for TP53 mutations in ccRCC. Here we use a zebrafish model to describe the functional consequences of pVHL loss on the p53/Mdm2 pathway. We show that p53 is stabilized in the absence of pVHL and becomes hyperstabilized upon DNA damage, which we propose is because of a novel in vivo interaction revealed between human pVHL and a negative regulator of Mdm2, the programmed cell death 5 (PDCD5) protein. PDCD5 is normally localized at the plasma membrane and in the cytoplasm. However, upon hypoxia or loss of pVHL, PDCD5 relocalizes to the nucleus, an event that is coupled to the degradation of Mdm2. Despite the subsequent hyperstabilization and normal transcriptional activity of p53, we find that zebrafish vhl/ cells are still as highly resistant to DNA damage-induced cell cycle arrest and apoptosis as human ccRCC cells. We suggest this is because of a marked increase in expression of birc5a, the zebrafish homolog of Survivin. Accordingly, when we knock down Survivin in human ccRCC cells we are able to restore caspase activity in response to DNA damage. Taken together, our study describes a new mechanism for p53 stabilization through PDCD5 upon hypoxia or pVHL loss, and reveals new clinical potential for the treatment of pathobiological disorders linked to hypoxic stress.

Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping