The long non-coding RNA Cerox1 is a post transcriptional regulator of mitochondrial complex I catalytic activity

Elife. 2019 May 2:8:e45051. doi: 10.7554/eLife.45051.

Abstract

To generate energy efficiently, the cell is uniquely challenged to co-ordinate the abundance of electron transport chain protein subunits expressed from both nuclear and mitochondrial genomes. How an effective stoichiometry of this many constituent subunits is co-ordinated post-transcriptionally remains poorly understood. Here we show that Cerox1, an unusually abundant cytoplasmic long noncoding RNA (lncRNA), modulates the levels of mitochondrial complex I subunit transcripts in a manner that requires binding to microRNA-488-3p. Increased abundance of Cerox1 cooperatively elevates complex I subunit protein abundance and enzymatic activity, decreases reactive oxygen species production, and protects against the complex I inhibitor rotenone. Cerox1 function is conserved across placental mammals: human and mouse orthologues effectively modulate complex I enzymatic activity in mouse and human cells, respectively. Cerox1 is the first lncRNA demonstrated, to our knowledge, to regulate mitochondrial oxidative phosphorylation and, with miR-488-3p, represent novel targets for the modulation of complex I activity.

Keywords: biochemistry; chemical biology; energy metabolism; genetics; genomics; human; long noncoding RNA; miRNA; mitochondria; mouse; post-transcriptional regulation.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Cell Line
  • Electron Transport Complex I / genetics
  • Electron Transport Complex I / metabolism*
  • Gene Expression Profiling
  • Gene Expression Regulation*
  • Humans
  • Mice
  • MicroRNAs / metabolism
  • Mitochondria / enzymology*
  • RNA, Long Noncoding / metabolism*

Substances

  • MIRN488 microRNA, human
  • MicroRNAs
  • RNA, Long Noncoding
  • Electron Transport Complex I