Your browser doesn't support javascript.
loading
PHDs/CPT1B/VDAC1 axis regulates long-chain fatty acid oxidation in cardiomyocytes.
Angelini, Aude; Saha, Pradip K; Jain, Antrix; Jung, Sung Yun; Mynatt, Randall L; Pi, Xinchun; Xie, Liang.
Affiliation
  • Angelini A; Department of Medicine, Section of Athero & Lipo, Baylor College of Medicine, Houston, TX 77030, USA; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA.
  • Saha PK; Department of Medicine, Division of Diabetes, Endocrinology & Metabolism, Diabetes Research Center, Baylor College of Medicine, Houston, TX 77030, USA.
  • Jain A; Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX 77030, USA.
  • Jung SY; Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX 77030, USA.
  • Mynatt RL; Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
  • Pi X; Department of Medicine, Section of Athero & Lipo, Baylor College of Medicine, Houston, TX 77030, USA; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA.
  • Xie L; Department of Medicine, Section of Athero & Lipo, Baylor College of Medicine, Houston, TX 77030, USA; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA. Electronic address: liangx@bcm.edu.
Cell Rep ; 37(1): 109767, 2021 10 05.
Article in En | MEDLINE | ID: mdl-34610308
ABSTRACT
Cardiac metabolism is a high-oxygen-consuming process, showing a preference for long-chain fatty acid (LCFA) as the fuel source under physiological conditions. However, a metabolic switch (favoring glucose instead of LCFA) is commonly reported in ischemic or late-stage failing hearts. The mechanism regulating this metabolic switch remains poorly understood. Here, we report that loss of PHD2/3, the cellular oxygen sensors, blocks LCFA mitochondria uptake and ß-oxidation in cardiomyocytes. In high-fat-fed mice, PHD2/3 deficiency improves glucose metabolism but exacerbates the cardiac defects. Mechanistically, we find that PHD2/3 bind to CPT1B, a key enzyme of mitochondrial LCFA uptake, promoting CPT1B-P295 hydroxylation. Further, we show that CPT1B-P295 hydroxylation is indispensable for its interaction with VDAC1 and LCFA ß-oxidation. Finally, we demonstrate that a CPT1B-P295A mutant constitutively binds to VDAC1 and rescues LCFA metabolism in PHD2/3-deficient cardiomyocytes. Together, our data identify an oxygen-sensitive regulatory axis involved in cardiac metabolism.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Carnitine O-Palmitoyltransferase / Procollagen-Proline Dioxygenase / Voltage-Dependent Anion Channel 1 / Fatty Acids / Hypoxia-Inducible Factor-Proline Dioxygenases Type of study: Prognostic_studies Limits: Animals Language: En Journal: Cell Rep Year: 2021 Document type: Article Affiliation country:

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Carnitine O-Palmitoyltransferase / Procollagen-Proline Dioxygenase / Voltage-Dependent Anion Channel 1 / Fatty Acids / Hypoxia-Inducible Factor-Proline Dioxygenases Type of study: Prognostic_studies Limits: Animals Language: En Journal: Cell Rep Year: 2021 Document type: Article Affiliation country: