Cookies help us deliver our services. By using our services, you agree to our use of cookies. More information

Deleye 2020 J Biol Chem

From Bioblast
The printable version is no longer supported and may have rendering errors. Please update your browser bookmarks and please use the default browser print function instead.
Publications in the MiPMap
Deleye Y, Cotte AK, Hannou SA, Hennuyer N, Bernard L, Derudas B, Caron S, Legry V, Vallez E, Dorchies E, Martin N, Lancel S, Annicotte JS, Bantubungi K, Pourtier A, Raverdy V, Pattou F, Lefebvre P, Abbadie C, Staels B, Haas JT, Paumelle R (2020) CDKN2A/p16INK4a suppresses hepatic fatty acid oxidation through the AMPKα2-SIRT1-PPARα signaling pathway. J Biol Chem 295:17310-22.

» PMID: 33037071 Open Access

Deleye Yann, Cotte Alexia Karen, Hannou Sarah Anissa, Nathalie Hennuyer, Bernard Lucie, Derudas Bruno, Caron Sandrine, Legry Vanessa, Vallez Emmanuelle, Dorchies Emilie, Martin Nathalie, Lancel Steve, Annicotte Jean-Sebastien, Bantubungi Kadiombo, Pourtier Albin, Raverdy Violeta, Pattou Francois, Lefebvre Philippe, Abbadie Corinne, Staels Bart, Haas Joel T, Paumelle Rejane (2020) J Biol Chem

Abstract: In addition to their well-known role in the control of cellular proliferation and cancer, cell cycle regulators are increasingly identified as important metabolic modulators. Several GWAS have identified SNPs near CDKN2A, the locus encoding for p16INK4a (p16), associated with elevated risk for cardiovascular diseases and type-2 diabetes development, two pathologies associated with impaired hepatic lipid metabolism. Although p16 was recently shown to control hepatic glucose homeostasis, it is unknown whether p16 also controls hepatic lipid metabolism. Using a combination of in vivo and in vitro approaches, we found that p16 modulates fasting-induced hepatic fatty acid oxidation (FAO) and lipid droplet accumulation. In primary hepatocytes, p16-deficiency was associated with elevated expression of genes involved in fatty acid catabolism. These transcriptional changes led to increased FAO and were associated with enhanced activation of PPARα through a mechanism requiring the catalytic AMPKα2 subunit and SIRT1, two known activators of PPARα. By contrast, p16 overexpression was associated with triglyceride accumulation and increased lipid droplet numbers in vitro, and decreased ketogenesis and hepatic mitochondrial activity in vivo. Finally, gene expression analysis of liver samples from obese patients revealed a negative correlation between CDKN2A expression and PPARA and its target genes. Our findings demonstrate that p16 represses hepatic lipid catabolism during fasting and may thus participate in the preservation of metabolic flexibility. Keywords: AMP-activated kinase (AMPK), Cell cycle, Fatty acid oxidation, Lipid metabolism, Liver, Peroxisome proliferator-activated receptor (PPAR) Bioblast editor: Plangger M O2k-Network Lab: FR Lille Duez H, FR Lille Lancel Steve


Labels: MiParea: Respiration, nDNA;cell genetics 


Organism: Mouse  Tissue;cell: Liver  Preparation: Homogenate 


Coupling state: LEAK, OXPHOS  Pathway: F, N  HRR: Oxygraph-2k 

2020-10