1. Academic Validation
  2. PPARα inhibition modulates multiple reprogrammed metabolic pathways in kidney cancer and attenuates tumor growth

PPARα inhibition modulates multiple reprogrammed metabolic pathways in kidney cancer and attenuates tumor growth

  • Am J Physiol Cell Physiol. 2015 Jun 1;308(11):C890-8. doi: 10.1152/ajpcell.00322.2014.
Omran Abu Aboud 1 Dallas Donohoe 2 Scott Bultman 3 Mark Fitch 4 Tim Riiff 5 Marc Hellerstein 4 Robert H Weiss 6
Affiliations

Affiliations

  • 1 Graduate Group in Comparative Pathology, University of California, Davis, California; Division of Nephrology, Department of Internal Medicine, University of California, Davis, California;
  • 2 Department of Nutrition, University of Tennessee, Knoxville, Tennessee;
  • 3 Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina;
  • 4 Department of Nutritional Science and Toxicology, University of California, Berkeley, California;
  • 5 KineMed, Inc., Emeryville, California; and.
  • 6 Graduate Group in Comparative Pathology, University of California, Davis, California; Division of Nephrology, Department of Internal Medicine, University of California, Davis, California; Cancer Center, University of California, Davis, California; Medical Service, Sacramento Veterans Affairs Medical Center, Sacramento, California [email protected].
Abstract

Kidney Cancer [renal cell carcinoma (RCC)] is the sixth-most-common Cancer in the United States, and its incidence is increasing. The current progression-free survival for patients with advanced RCC rarely extends beyond 1-2 yr due to the development of therapeutic resistance. We previously identified peroxisome proliferator-activating receptor-α (PPARα) as a potential therapeutic target for this disease and showed that a specific PPARα Antagonist, GW6471, induced Apoptosis and cell cycle arrest at G0/G1 in RCC cell lines associated with attenuation of cell cycle regulatory proteins. We now extend that work and show that PPARα inhibition attenuates components of RCC metabolic reprogramming, capitalizing on the Warburg effect. The specific PPARα Inhibitor GW6471, as well as a siRNA specific to PPARα, attenuates the enhanced fatty acid oxidation and oxidative phosphorylation associated with glycolysis inhibition, and PPARα antagonism also blocks the enhanced glycolysis that has been observed in RCC cells; this effect did not occur in normal human kidney epithelial cells. Such cell type-specific inhibition of glycolysis corresponds with changes in protein levels of the oncogene c-Myc and has promising clinical implications. Furthermore, we show that treatment with GW6471 results in RCC tumor growth attenuation in a xenograft mouse model, with minimal obvious toxicity, a finding associated with the expected on-target effects on c-Myc. These studies demonstrate that several pivotal cancer-relevant metabolic pathways are inhibited by PPARα antagonism. Our data support the concept that targeting PPARα, with or without concurrent inhibition of glycolysis, is a potential novel and effective therapeutic approach for RCC that targets metabolic reprogramming in this tumor.

Keywords

kidney cancer; metabolomics; peroxisome proliferator-activating receptor-α; reprogramming.

Figures
Products
  • Cat. No.
    Product Name
    Description
    Target
    Research Area
  • HY-15372
    98.81%, PPAR Antagonist