Exercise training boosts eNOS-dependent mitochondrial biogenesis in mouse heart: role in adaptation of glucose metabolism

R Vettor, A Valerio, M Ragni… - American Journal …, 2014 - journals.physiology.org
R Vettor, A Valerio, M Ragni, E Trevellin, M Granzotto, M Olivieri, L Tedesco, C Ruocco…
American Journal of Physiology-Endocrinology and Metabolism, 2014journals.physiology.org
Endurance exercise training increases cardiac energy metabolism through poorly
understood mechanisms. Nitric oxide (NO) produced by endothelial NO synthase (eNOS) in
cardiomyocytes contributes to cardiac adaptation. Here we demonstrate that the NO donor
diethylenetriamine-NO (DETA-NO) activated mitochondrial biogenesis and function, as
assessed by upregulated peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-
1α), nuclear respiratory factor 1, and mitochondrial transcription factor A (Tfam) expression …
Endurance exercise training increases cardiac energy metabolism through poorly understood mechanisms. Nitric oxide (NO) produced by endothelial NO synthase (eNOS) in cardiomyocytes contributes to cardiac adaptation. Here we demonstrate that the NO donor diethylenetriamine-NO (DETA-NO) activated mitochondrial biogenesis and function, as assessed by upregulated peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), nuclear respiratory factor 1, and mitochondrial transcription factor A (Tfam) expression, and by increased mitochondrial DNA content and citrate synthase activity in primary mouse cardiomyocytes. DETA-NO also induced mitochondrial biogenesis and function and enhanced both basal and insulin-stimulated glucose uptake in HL-1 cardiomyocytes. The DETA-NO-mediated effects were suppressed by either PGC-1α or Tfam small-interference RNA in HL-1 cardiomyocytes. Wild-type and eNOS−/− mice were subjected to 6 wk graduated swim training. We found that eNOS expression, mitochondrial biogenesis, mitochondrial volume density and number, and both basal and insulin-stimulated glucose uptake were increased in left ventricles of swim-trained wild-type mice. On the contrary, the genetic deletion of eNOS prevented all these adaptive phenomena. Our findings demonstrate that exercise training promotes eNOS-dependent mitochondrial biogenesis in heart, which behaves as an essential step in cardiac glucose transport.
American Physiological Society