4.8 Article

NADH inhibition of SIRT1 links energy state to transcription during time-restricted feeding

Journal

NATURE METABOLISM
Volume 3, Issue 12, Pages 1621-+

Publisher

NATURE PORTFOLIO
DOI: 10.1038/s42255-021-00498-1

Keywords

-

Funding

  1. National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) [R01DK090625, R01DK100814, 1R01DK113011-01A1]
  2. Chicago Biomedical Consortium [S-007]
  3. National Institute on Aging (NIA) [P01AG011412]
  4. NIDDK [F32DK122675, 5K01DK105137-03, 1R03DK116012-01, R01DK108987]
  5. Swedish Research Council [2014-6888]
  6. Swedish Society for Medical Research
  7. Swedish Brain Research Foundation
  8. National Cancer Institute (NCI) [U54CA199091]
  9. NCI [R35CA197532-04]
  10. NIA [P01AG049665-05]
  11. National Institute of General Medical Sciences [T32GM008061]
  12. National Institute of Child Health and Human Development [R01HD089552]

Ask authors/readers for more resources

Through genetic uncoupling of nutrient state from NADH redox state, the study demonstrates that the hepatic NADH cycle regulates whole-body energetics by affecting SIRT1, thereby linking nutrient status with circadian transcriptional cycles.
In mammals, circadian rhythms are entrained to the light cycle and drive daily oscillations in levels of NAD(+), a cosubstrate of the class III histone deacetylase sirtuin 1 (SIRT1) that associates with clock transcription factors. Although NAD(+) also participates in redox reactions, the extent to which NAD(H) couples nutrient state with circadian transcriptional cycles remains unknown. Here we show that nocturnal animals subjected to time-restricted feeding of a calorie-restricted diet (TRF-CR) only during night-time display reduced body temperature and elevated hepatic NADH during daytime. Genetic uncoupling of nutrient state from NADH redox state through transduction of the water-forming NADH oxidase from Lactobacillus brevis (LbNOX) increases daytime body temperature and blood and liver acyl-carnitines. LbNOX expression in TRF-CR mice induces oxidative gene networks controlled by brain and muscle Arnt-like protein 1 (BMAL1) and peroxisome proliferator-activated receptor alpha (PPAR alpha) and suppresses amino acid catabolic pathways. Enzymatic analyses reveal that NADH inhibits SIRT1 in vitro, corresponding with reduced deacetylation of SIRT1 substrates during TRF-CR in vivo. Remarkably, Sirt1 liver nullizygous animals subjected to TRF-CR display persistent hypothermia even when NADH is oxidized by LbNOX. Our findings reveal that the hepatic NADH cycle links nutrient state to whole-body energetics through the rhythmic regulation of SIRT1. Using a model of time-restricted feeding in mice, Levine et al. show that the hepatic NADH cycle links nutrient state to whole-body energetics through the rhythmic regulation of SIRT1.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available