Scientific Reports (Oct 2023)

Empagliflozin induces the transcriptional program for nutrient homeostasis in skeletal muscle in normal mice

  • Ryo Kawakami,
  • Hiroki Matsui,
  • Miki Matsui,
  • Tatsuya Iso,
  • Tomoyuki Yokoyama,
  • Hideki Ishii,
  • Masahiko Kurabayashi

DOI
https://doi.org/10.1038/s41598-023-45390-y
Journal volume & issue
Vol. 13, no. 1
pp. 1 – 11

Abstract

Read online

Abstract Sodium-glucose cotransporter 2 inhibitors (SGLT2i) improve heart failure (HF) outcomes across a range of patient characteristics. A hypothesis that SGLT2i induce metabolic change similar to fasting has recently been proposed to explain their profound clinical benefits. However, it remains unclear whether SGLT2i primarily induce this change in physiological settings. Here, we demonstrate that empagliflozin administration under ad libitum feeding did not cause weight loss but did increase transcripts of the key nutrient sensors, AMP-activated protein kinase and nicotinamide phosphoribosyltransferase, and the master regulator of mitochondrial gene expression, PGC-1α, in quadriceps muscle in healthy mice. Expression of these genes correlated with that of PPARα and PPARδ target genes related to mitochondrial metabolism and oxidative stress response, and also correlated with serum ketone body β-hydroxybutyrate. These results were not observed in the heart. Collectively, this study revealed that empagliflozin activates transcriptional programs critical for sensing and adaptation to nutrient availability intrinsic to skeletal muscle rather than the heart even in normocaloric condition. As activation of PGC-1α is sufficient for metabolic switch from fatigable, glycolytic metabolism toward fatigue-resistant, oxidative mechanism in skeletal muscle myofibers, our findings may partly explain the improvement of exercise tolerance in patients with HF receiving empagliflozin.