p53/HMGB1 complexes regulate autophagy and apoptosis

Cancer Res. 2012 Apr 15;72(8):1996-2005. doi: 10.1158/0008-5472.CAN-11-2291. Epub 2012 Feb 16.

Abstract

The balance between apoptosis ("programmed cell death") and autophagy ("programmed cell survival") is important in tumor development and response to therapy. Here, we show that high mobility group box 1 (HMGB1) and p53 form a complex that regulates the balance between tumor cell death and survival. We show that knockout of p53 in HCT116 cells increases expression of cytosolic HMGB1 and induces autophagy. Conversely, knockout of HMGB1 in mouse embryonic fibroblasts increases p53 cytosolic localization and decreases autophagy. p53 is thus a negative regulator of the HMGB1/Beclin 1 complex, and HMGB1 promotes autophagy in the setting of diminished p53. HMGB1-mediated autophagy promotes tumor cell survival in the setting of p53-dependent processes. The HMGB1/p53 complex affects the cytoplasmic localization of the reciprocal binding partner, thereby regulating subsequent levels of autophagy and apoptosis. These insights provide a novel link between HMGB1 and p53 in the cross-regulation of apoptosis and autophagy in the setting of cell stress, providing insights into their reciprocal roles in carcinogenesis.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Apoptosis / physiology*
  • Autophagy / physiology*
  • Cell Transformation, Neoplastic / metabolism*
  • HCT116 Cells
  • HMGB1 Protein / metabolism*
  • Humans
  • Mice
  • Mice, Knockout
  • Tumor Suppressor Protein p53 / metabolism*

Substances

  • HMGB1 Protein
  • TP53 protein, human
  • Tumor Suppressor Protein p53