Regulation of anoikis by deleted in breast cancer-1 (DBC1) through NF-κB

Apoptosis. 2013 Aug;18(8):949-62. doi: 10.1007/s10495-013-0847-1.

Abstract

Anoikis-resistance of tumor cells is critical for anchorage-independent growth and metastasis. The inflammatory-response transcription factor NF-κB contributes to anoikis-resistance and tumor progression through mechanisms that are understood incompletely. Deleted in breast cancer-1 (DBC1) protein (KIAA1967) is over-expressed in several tumor types, and correlates with a poorer prognosis in some cases. We report here that DBC1 suppressed anoikis in normal epithelial and breast cancer cell lines. DBC1 interacted with IKK-β, stimulating its kinase activity, promoting NF-κB transcriptional activity through the phosphorylation of relA serine-536 and enhancing the expression of the NF-κB target genes, c-FLIP and bcl-xl. Our results indicate that DBC1 is an important co-factor for the control of the IKK-β-NF-κB signaling pathway that regulates anoikis.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Adaptor Proteins, Signal Transducing*
  • Anoikis*
  • Breast Neoplasms / genetics
  • Breast Neoplasms / metabolism*
  • Breast Neoplasms / physiopathology
  • Cell Line, Tumor
  • Female
  • Humans
  • I-kappa B Kinase / genetics
  • I-kappa B Kinase / metabolism
  • NF-kappa B / genetics*
  • NF-kappa B / metabolism
  • Phosphorylation
  • Protein Binding
  • Signal Transduction
  • Transcription Factor RelA / genetics
  • Transcription Factor RelA / metabolism
  • Transcriptional Activation

Substances

  • Adaptor Proteins, Signal Transducing
  • CCAR2 protein, human
  • NF-kappa B
  • Transcription Factor RelA
  • I-kappa B Kinase