1. Academic Validation
  2. Cullin3-KLHL25 ubiquitin ligase targets ACLY for degradation to inhibit lipid synthesis and tumor progression

Cullin3-KLHL25 ubiquitin ligase targets ACLY for degradation to inhibit lipid synthesis and tumor progression

  • Genes Dev. 2016 Sep 1;30(17):1956-70. doi: 10.1101/gad.283283.116.
Cen Zhang 1 Juan Liu 1 Grace Huang 2 Yuhan Zhao 1 Xuetian Yue 1 Hao Wu 1 Jun Li 1 Junlan Zhu 2 Zhiyuan Shen 1 Bruce G Haffty 1 Wenwei Hu 1 Zhaohui Feng 3
Affiliations

Affiliations

  • 1 Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey 08903, USA;
  • 2 Nelson Institute of Environmental Medicine, New York University School of Medicine, New York University, Tuxedo, New Jersey 10987, USA;
  • 3 Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey 08903, USA; Department of Pharmacology, Rutgers University, The State University of New Jersey, Piscataway, New Jersey 08854, USA.
Abstract

Increased lipid synthesis is a key characteristic of many cancers that is critical for Cancer progression. ATP-citrate lyase (ACLY), a key Enzyme for lipid synthesis, is frequently overexpressed or activated in Cancer to promote lipid synthesis and tumor progression. Cullin3 (CUL3), a core protein for the CUL3-RING ubiquitin ligase complex, has been reported to be a tumor suppressor and frequently down-regulated in lung Cancer. Here, we found that CUL3 interacts with ACLY through its adaptor protein, KLHL25 (Kelch-like family member 25), to ubiquitinate and degrade ACLY in cells. Through negative regulation of ACLY, CUL3 inhibits lipid synthesis, cell proliferation, and xenograft tumor growth of lung Cancer cells. Furthermore, ACLY inhibitor SB-204990 greatly abolishes the promoting effect of CUL3 down-regulation on lipid synthesis, cell proliferation, and tumor growth. Importantly, low CUL3 expression is associated with high ACLY expression and poor prognosis in human lung Cancer. In summary, our results identify CUL3-KLHL25 ubiquitin ligase as a novel negative regulator for ACLY and lipid synthesis and demonstrate that decreased CUL3 expression is an important mechanism for increased ACLY expression and lipid synthesis in lung Cancer. These results also reveal that negative regulation of ACLY and lipid synthesis is a novel and critical mechanism for CUL3 in tumor suppression.

Keywords

ACLY; CUL3; KLHL25; lipid synthesis; tumor progression; ubiquitination.

Figures