Cells (Jan 2020)

Mitochondrial Fusion Via OPA1 and MFN1 Supports Liver Tumor Cell Metabolism and Growth

  • Meng Li,
  • Ling Wang,
  • Yijin Wang,
  • Shaoshi Zhang,
  • Guoying Zhou,
  • Ruby Lieshout,
  • Buyun Ma,
  • Jiaye Liu,
  • Changbo Qu,
  • Monique M. A. Verstegen,
  • Dave Sprengers,
  • Jaap Kwekkeboom,
  • Luc J. W. van der Laan,
  • Wanlu Cao,
  • Maikel P. Peppelenbosch,
  • Qiuwei Pan

DOI
https://doi.org/10.3390/cells9010121
Journal volume & issue
Vol. 9, no. 1
p. 121

Abstract

Read online

Metabolic reprogramming universally occurs in cancer. Mitochondria act as the hubs of bioenergetics and metabolism. The morphodynamics of mitochondria, comprised of fusion and fission processes, are closely associated with mitochondrial functions and are often dysregulated in cancer. In this study, we aim to investigate the mitochondrial morphodynamics and its functional consequences in human liver cancer. We observed excessive activation of mitochondrial fusion in tumor tissues from hepatocellular carcinoma (HCC) patients and in vitro cultured tumor organoids from cholangiocarcinoma (CCA). The knockdown of the fusion regulator genes, OPA1 (Optic atrophy 1) or MFN1 (Mitofusin 1), inhibited the fusion process in HCC cell lines and CCA tumor organoids. This resulted in inhibition of cell growth in vitro and tumor formation in vivo, after tumor cell engraftment in mice. This inhibitory effect is associated with the induction of cell apoptosis, but not related to cell cycle arrest. Genome-wide transcriptomic profiling revealed that the inhibition of fusion predominately affected cellular metabolic pathways. This was further confirmed by the blocking of mitochondrial fusion which attenuated oxygen consumption and cellular ATP production of tumor cells. In conclusion, increased mitochondrial fusion in liver cancer alters metabolism and fuels tumor cell growth.

Keywords