hMOB3 modulates MST1 apoptotic signaling and supports tumor growth in glioblastoma multiforme

Cancer Res. 2014 Jul 15;74(14):3779-89. doi: 10.1158/0008-5472.CAN-13-3430. Epub 2014 May 28.

Abstract

New therapeutic targets are needed that circumvent inherent therapeutic resistance of glioblastoma multiforme (GBM). Here, we report such a candidate target in the uncharacterized adaptor protein hMOB3, which we show is upregulated in GBM. In a search for its biochemical function, we found that hMOB3 specifically interacts with MST1 kinase in response to apoptotic stimuli and cell-cell contact. Moreover, hMOB3 negatively regulated apoptotic signaling by MST1 in GBM cells by inhibiting the MST1 cleavage-based activation process. Physical interaction between hMOB3 and MST1 was essential for this process. In vivo investigations established that hMOB3 sustains GBM cell growth at high cell density and promotes tumorigenesis. Our results suggest hMOB3 as a candidate therapeutic target for the treatment of malignant gliomas.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Apoptosis*
  • Cell Line, Tumor
  • Cell Proliferation
  • Disease Models, Animal
  • Gene Expression
  • Glioblastoma / genetics
  • Glioblastoma / metabolism*
  • Glioblastoma / pathology*
  • Hepatocyte Growth Factor / metabolism*
  • Heterografts
  • Humans
  • Immunohistochemistry
  • Microtubule-Associated Proteins / genetics
  • Microtubule-Associated Proteins / metabolism*
  • Protein Binding
  • Proteolysis
  • Proto-Oncogene Proteins / metabolism*
  • Signal Transduction*
  • Tumor Burden

Substances

  • MAP1B protein, human
  • Microtubule-Associated Proteins
  • Proto-Oncogene Proteins
  • macrophage stimulating protein
  • Hepatocyte Growth Factor