Formation of Tankyrase Inhibitor-Induced Degradasomes Requires Proteasome Activity

PLoS One. 2016 Aug 2;11(8):e0160507. doi: 10.1371/journal.pone.0160507. eCollection 2016.

Abstract

In canonical Wnt signaling, the protein levels of the key signaling mediator β-catenin are under tight regulation by the multimeric destruction complex that mediates proteasomal degradation of β-catenin. In colorectal cancer, destruction complex activity is often compromised due to mutations in the multifunctional scaffolding protein Adenomatous Polyposis Coli (APC), leading to a stabilization of β-catenin. Recently, tankyrase inhibitors (TNKSi), a novel class of small molecule inhibitors, were shown to re-establish a functional destruction complex in APC-mutant cancer cell lines by stabilizing AXIN1/2, whose protein levels are usually kept low via poly(ADP-ribosyl)ation by the tankyrase enzymes (TNKS1/2). Surprisingly, we found that for the formation of the morphological correlates of destruction complexes, called degradasomes, functional proteasomes are required. In addition we found that AXIN2 is strongly upregulated after 6 h of TNKS inhibition. The proteasome inhibitor MG132 counteracted TNKSi-induced degradasome formation and AXIN2 stabilization, and this was accompanied by reduced transcription of AXIN2. Mechanistically we could implicate the transcription factor FoxM1 in this process, which was recently shown to be a transcriptional activator of AXIN2. We observed a substantial reduction in TNKSi-induced stabilization of AXIN2 after siRNA-mediated depletion of FoxM1 and found that proteasome inhibition reduced the active (phosphorylated) fraction of FoxM1. This can explain the decreased protein levels of AXIN2 after MG132 treatment. Our findings have implications for the design of in vitro studies on the destruction complex and for clinical applications of TNKSi.

MeSH terms

  • Adenomatous Polyposis Coli Protein / genetics
  • Adenomatous Polyposis Coli Protein / metabolism
  • Axin Protein / genetics
  • Axin Protein / metabolism
  • Caco-2 Cells
  • Cell Line, Tumor
  • Enzyme Inhibitors / pharmacology
  • Forkhead Box Protein M1 / antagonists & inhibitors
  • Forkhead Box Protein M1 / genetics*
  • Forkhead Box Protein M1 / metabolism
  • Gene Expression Regulation, Neoplastic*
  • Humans
  • Leupeptins / pharmacology
  • Phosphorylation / drug effects
  • Proteasome Endopeptidase Complex / drug effects
  • Proteasome Endopeptidase Complex / metabolism*
  • Protein Stability
  • Proteolysis / drug effects
  • RNA, Small Interfering / genetics
  • RNA, Small Interfering / metabolism
  • Tankyrases / antagonists & inhibitors
  • Tankyrases / genetics*
  • Tankyrases / metabolism
  • Wnt Signaling Pathway
  • beta Catenin / genetics
  • beta Catenin / metabolism

Substances

  • APC protein, human
  • AXIN1 protein, human
  • AXIN2 protein, human
  • Adenomatous Polyposis Coli Protein
  • Axin Protein
  • CTNNB1 protein, human
  • Enzyme Inhibitors
  • FOXM1 protein, human
  • Forkhead Box Protein M1
  • Leupeptins
  • RNA, Small Interfering
  • beta Catenin
  • TNKS2 protein, human
  • Tankyrases
  • TNKS protein, human
  • Proteasome Endopeptidase Complex
  • benzyloxycarbonylleucyl-leucyl-leucine aldehyde

Grants and funding

NMP is a postdoctoral fellow of the Norwegian Cancer Society (Grant number 4541899145315, https://kreftforeningen.no/en/). TET is a PhD student of the South-Eastern Norway Regional Health Authority (http://www.helse-sorost.no/). SWS is a research fellow of the Norwegian Cancer Society (Grant number 6789795-2015). EMW is a senior research fellow of the South-Eastern Norway Regional Health Authority (Grant number 2015014). HS has been supported by the Norwegian Cancer Society. This work was partly supported by the Research Council of Norway through its Centres of Excellence funding scheme, project number 179571 (http://www.forskningsradet.no/). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.