PLoS ONE (Jan 2020)

Mitochondrial dysfunction underlying sporadic inclusion body myositis is ameliorated by the mitochondrial homing drug MA-5.

  • Yoshitsugu Oikawa,
  • Rumiko Izumi,
  • Masashi Koide,
  • Yoshihiro Hagiwara,
  • Makoto Kanzaki,
  • Naoki Suzuki,
  • Koichi Kikuchi,
  • Tetsuro Matsuhashi,
  • Yukako Akiyama,
  • Mariko Ichijo,
  • Shun Watanabe,
  • Takafumi Toyohara,
  • Takehiro Suzuki,
  • Eikan Mishima,
  • Yasutoshi Akiyama,
  • Yoshiaki Ogata,
  • Chitose Suzuki,
  • Hironori Hayashi,
  • Eiichi N Kodama,
  • Ken-Ichiro Hayashi,
  • Eiji Itoi,
  • Masashi Aoki,
  • Shigeo Kure,
  • Takaaki Abe

DOI
https://doi.org/10.1371/journal.pone.0231064
Journal volume & issue
Vol. 15, no. 12
p. e0231064

Abstract

Read online

Sporadic inclusion body myositis (sIBM) is the most common idiopathic inflammatory myopathy, and several reports have suggested that mitochondrial abnormalities are involved in its etiology. We recruited 9 sIBM patients and found significant histological changes and an elevation of growth differential factor 15 (GDF15), a marker of mitochondrial disease, strongly suggesting the involvement of mitochondrial dysfunction. Bioenergetic analysis of sIBM patient myoblasts revealed impaired mitochondrial function. Decreased ATP production, reduced mitochondrial size and reduced mitochondrial dynamics were also observed in sIBM myoblasts. Cell vulnerability to oxidative stress also suggested the existence of mitochondrial dysfunction. Mitochonic acid-5 (MA-5) increased the cellular ATP level, reduced mitochondrial ROS, and provided protection against sIBM myoblast death. MA-5 also improved the survival of sIBM skin fibroblasts as well as mitochondrial morphology and dynamics in these cells. The reduction in the gene expression levels of Opa1 and Drp1 was also reversed by MA-5, suggesting the modification of the fusion/fission process. These data suggest that MA-5 may provide an alternative therapeutic strategy for treating not only mitochondrial diseases but also sIBM.