Mice deficient in the Shmt2 gene have mitochondrial respiration defects and are embryonic lethal

Accumulation of somatic mutations in mitochondrial DNA (mtDNA) has been proposed to be responsible for human aging and age-associated mitochondrial respiration defects. However, our previous findings suggested an alternative hypothesis of human aging—that epigenetic changes but not mutations regulat...

Full description

Saved in:
Bibliographic Details
Published inScientific reports Vol. 8; no. 1; p. 425
Main Authors Tani, Haruna, Ohnishi, Sakiko, Shitara, Hiroshi, Mito, Takayuki, Yamaguchi, Midori, Yonekawa, Hiromichi, Hashizume, Osamu, Ishikawa, Kaori, Nakada, Kazuto, Hayashi, Jun-Ichi
Format Journal Article
LanguageEnglish
Published London Nature Publishing Group UK 11.01.2018
Nature Publishing Group
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Accumulation of somatic mutations in mitochondrial DNA (mtDNA) has been proposed to be responsible for human aging and age-associated mitochondrial respiration defects. However, our previous findings suggested an alternative hypothesis of human aging—that epigenetic changes but not mutations regulate age-associated mitochondrial respiration defects, and that epigenetic downregulation of nuclear-coded genes responsible for mitochondrial translation [e.g., glycine C-acetyltransferase ( GCAT ), serine hydroxymethyltransferase 2 ( SHMT2 )] is related to age-associated respiration defects. To examine our hypothesis, here we generated mice deficient in Gcat or Shmt2 and investigated whether they have respiration defects and premature aging phenotypes. Gcat -deficient mice showed no macroscopic abnormalities including premature aging phenotypes for up to 9 months after birth. In contrast, Shmt2 -deficient mice showed embryonic lethality after 13.5 days post coitum (dpc), and fibroblasts obtained from 12.5-dpc Shmt2 -deficient embryos had respiration defects and retardation of cell growth. Because Shmt2 substantially controls production of N-formylmethionine-tRNA (fMet-tRNA) in mitochondria, its suppression would reduce mitochondrial translation, resulting in expression of the respiration defects in fibroblasts from Shmt2 -deficient embryos. These findings support our hypothesis that age-associated respiration defects in fibroblasts of elderly humans are caused not by mtDNA mutations but by epigenetic regulation of nuclear genes including SHMT2 .
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:2045-2322
2045-2322
DOI:10.1038/s41598-017-18828-3