p53/HMGB1 complexes regulate autophagy and apoptosis

Kristen M. Livesey, Rui Kang, Philip Vernon, William Buchser, Patricia Loughran, Simon C. Watkins, Lin Zhang, James J. Manfredi, Herbert J. Zeh, Luyuan Li, Michael T. Lotze, Daolin Tang

Research output: Contribution to journalArticlepeer-review

219 Scopus citations

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.Weshow 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.

Original languageEnglish (US)
Pages (from-to)1996-2005
Number of pages10
JournalCancer research
Volume72
Issue number8
DOIs
StatePublished - Apr 15 2012
Externally publishedYes

ASJC Scopus subject areas

  • Oncology
  • Cancer Research

Fingerprint

Dive into the research topics of 'p53/HMGB1 complexes regulate autophagy and apoptosis'. Together they form a unique fingerprint.

Cite this