Frontiers in Oncology (Mar 2022)

Tumor Suppressor Par-4 Regulates Complement Factor C3 and Obesity

  • Nathalia Araujo,
  • James Sledziona,
  • Sunil K. Noothi,
  • Ravshan Burikhanov,
  • Nikhil Hebbar,
  • Saptadwipa Ganguly,
  • Tripti Shrestha-Bhattarai,
  • Beibei Zhu,
  • Beibei Zhu,
  • Wendy S. Katz,
  • Wendy S. Katz,
  • Yi Zhang,
  • Barry S. Taylor,
  • Jinze Liu,
  • Li Chen,
  • Li Chen,
  • Heidi L. Weiss,
  • Heidi L. Weiss,
  • Daheng He,
  • Chi Wang,
  • Chi Wang,
  • Andrew J. Morris,
  • Andrew J. Morris,
  • Lisa A. Cassis,
  • Lisa A. Cassis,
  • Mariana Nikolova-Karakashian,
  • Mariana Nikolova-Karakashian,
  • Prabhakar R. Nagareddy,
  • Olle Melander,
  • Olle Melander,
  • B. Mark Evers,
  • B. Mark Evers,
  • Philip A. Kern,
  • Philip A. Kern,
  • Vivek M. Rangnekar,
  • Vivek M. Rangnekar,
  • Vivek M. Rangnekar,
  • Vivek M. Rangnekar

DOI
https://doi.org/10.3389/fonc.2022.860446
Journal volume & issue
Vol. 12

Abstract

Read online

Prostate apoptosis response-4 (Par-4) is a tumor suppressor that induces apoptosis in cancer cells. However, the physiological function of Par-4 remains unknown. Here we show that conventional Par-4 knockout (Par-4-/-) mice and adipocyte-specific Par-4 knockout (AKO) mice, but not hepatocyte-specific Par-4 knockout mice, are obese with standard chow diet. Par-4-/- and AKO mice exhibit increased absorption and storage of fat in adipocytes. Mechanistically, Par-4 loss is associated with mdm2 downregulation and activation of p53. We identified complement factor c3 as a p53-regulated gene linked to fat storage in adipocytes. Par-4 re-expression in adipocytes or c3 deletion reversed the obese mouse phenotype. Moreover, obese human subjects showed lower expression of Par-4 relative to lean subjects, and in longitudinal studies, low baseline Par-4 levels denoted an increased risk of developing obesity later in life. These findings indicate that Par-4 suppresses p53 and its target c3 to regulate obesity.

Keywords