Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2021 Dec;3(12):1621-1632.
doi: 10.1038/s42255-021-00498-1. Epub 2021 Dec 13.

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

Affiliations

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

Daniel C Levine et al. Nat Metab. 2021 Dec.

Abstract

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 (PPARα) 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.

PubMed Disclaimer

Conflict of interest statement

The authors declare the following competing financial interest(s): M.M. is founder and chairman of SAMDI Tech Inc., which uses SAMDI-MS to perform high-throughput screening and assay chemistry to clients in the pharmaceutical industry. All other authors declare no competing financial interests.

Figures

Fig. 1
Fig. 1. Elevated NADH drives the daytime dip in body temperature during time-restricted calorie restriction.
a, Model for the TRF-CR diet using an automated feeder system in 4–6-month-old male C57BL/6J mice. Control mice received a 300 mg pellet of regular chow every 1.2 h throughout the dark period (TRF-Reg), whereas TRF-CR mice received a 300 mg pellet of a carbohydrate-depleted, nutrient-controlled chow every 2 h throughout the dark period, resulting in a 40% reduction in calories. b, Relative concentration of NADH and NAD+ by HPLC in liver of TRF-Reg (n = 12) and TRF-CR (n = 11) mice during the daytime (ZT4). c, log2(fold change) in daytime serum acyl-carnitine levels during TRF-CR compared with TRF-Reg mice (n = 3). d, Body temperature rhythms monitored non-invasively using subcutaneous probes in TRF-Reg (n = 12) and TRF-CR (n = 9) mice over 24 h (double plotted for clarity). e, Model depicting NADH-consuming reaction of LbNOX. Representative tissue-specific expression profile of cytonuclear LbNOX in null- and LbNOX-transduced mice relative to LbNOX-transduced liver. f, Relative concentration of NADH and NAD+ in liver of null- and LbNOX-transduced TRF-Reg (n = 6) or TRF-CR (n = 5 for NADH, n = 6 for NAD+) mice during the daytime. g, log2(fold change) in daytime serum acyl-carnitine levels in TRF-CR mice transduced with cytonuclear LbNOX compared with TRF-CR mice transduced with null virus (n = 3). h, Body temperature rhythms in LbNOX-expressing TRF-Reg (n = 12) or TRF-CR (n = 9) mice over 24 h (double plotted for clarity). Data are presented as mean ± s.e.m. Statistics were performed with unpaired, two-tailed Student’s t test unless otherwise noted in the figure. *P < 0.05, ***P < 0.001. ANOVA, analysis of variance; Cereb, cerebellum; DC, dicarboxylate; Gastroc, gastrocnemius muscle; gWAT, gonadal white adipose tissue; Hypoth, hypothalamus; i, isomer; iWAT, inguinal white adipose tissue; OH, hydroxy; Quad, quadriceps muscle.
Fig. 2
Fig. 2. Daytime NADH elevation regulates genome-wide transcription of fatty acid and amino acid metabolism genes during TRF-CR.
a, RNA-Seq in liver in the morning (ZT4) demonstrating the effect of TRF-CR in null-transduced (left) (n = 6) and LbNOX-transduced (middle) (n = 6 for TRF-Reg, n = 5 for TRF-CR) 4–6-month-old male C57BL/6J mice for genes differentially expressed (DESeq2 FDR-adjusted P < 0.05) by TRF-CR in null-transduced mice (930 genes). Venn diagram (right) displays overlap in differentially expressed genes by TRF-CR in null- and LbNOX-transduced mice. b, Quadrant plot comparing transcriptional responses between TRF-CR (x axis) and LbNOX in TRF-CR (y axis). Each point indicates a gene that is differentially expressed by TRF-CR in null-transduced mice (930 genes). Colouring indicates genes within quadrant 2 (green) and quadrant 4 (red), and the percentages within each quadrant are shown. c,d, For the genes within quadrants 2 and 4 from b, the top 15 (c) Kyoto Encyclopedia of Genes and Genomes (KEGG) terms enriched (P < 0.05) following gene ontology analysis and (d) JASPAR motifs enriched (P < 0.05) following HOMER DNA motif enrichment analysis are shown. e, Quadrant plots comparing the transcriptional response to TRF-CR in null-transduced mice (x axis) with that of genetic ablation of either Pparα (top) or Bmal1 (bottom) (y axis) in animals fed ad libitum. Each point indicates a gene that is differentially expressed by TRF-CR in null-transduced animals. Genes that have an absolute log2(fold change) > 0.5 for both comparisons are coloured blue or black, and the percentage of genes was determined by quadrant (n = 3). f, LC–MS metabolomics profiling of amino acids in liver during the daytime (ZT4). The log2(fold change) from TRF-CR (blue) (n = 5) and LbNOX in TRF-CR (white) (n = 6) is shown for select differential amino acids (two-tailed, unpaired Student’s t test with Benjamini and Hochberg adjustment for multiple measures FDR P < 0.05; see Supplementary Table 2 for full list of amino acids). Box and whisker plots depict the following: line, median; box limits, first and third quartiles; whiskers, 10th and 90th percentiles. g, Model depicting the interrelationship of NADH during TRF-CR to the activity of PPARα and BMAL1 and the transcription of downstream oxidative gene networks.
Fig. 3
Fig. 3. NADH inhibits SIRT1 in the morning during TRF-CR to regulate metabolism and body temperature.
a, Model to examine the role of SIRT1 in the NADH-dependent effects on RNAs, metabolites and body temperature during TRF-CR. b, Deacetylation rate for SIRT1 with increasing concentrations of NAD+ and NADH assayed by SAMDI-MS (n = 4). c, Model of pyruvate/lactate equilibrium shows that supplementation with pyruvate and lactate reduces and elevates NADH, respectively. Western blotting for SIRT1 targets, Ac-H3K9 and Ac-H4K16, in immortalized mouse embryonic fibroblasts treated with pyruvate or lactate to modulate NADH (n = 3). d,e, Western blotting for SIRT1 targets, Ac-p53 and Ac-FOXO1, in TRF-Reg or TRF-CR liver of 4–6-month-old male (d) control or liver-specific Sirt1/ mice or (e) null- or LbNOX-transduced mice. n.s., non-specific. Uncropped western blot scans labelled with molecular weight markers are presented in the Source Data files. f, BMAL1 ChIP-Seq in liver of TRF-Reg or TRF-CR mice. Peaks demonstrating an absolute log2(fold change) > 0.5 are coloured black. Box–whisker plots of BMAL1 ChIP-Seq demonstrating the effect of TRF-CR in null- (n = 6) and LbNOX-transduced (n = 3) liver on BMAL1 peaks identified in controls and with an absolute log2(fold change) > 0.5 in controls. Box and whisker plots depict the following: line, median; box limits, first and third quartiles; whiskers, 10th and 90th percentiles. g, Heatmap depicting log2(fold change) in gene expression (left), liver metabolite concentrations (middle) and serum acyl-carnitine levels (right) in indicated conditions/genotypes at ZT4. Heatmaps are subdivided into the genes, metabolites and acyl-carnitines that are regulated by LbNOX during TRF-CR through mechanisms requiring SIRT1 and sorted by effect of TRF-CR (RNA-Seq, n = 6; metabolomes, n = 5–6; acyl-carnitines n = 3). h, Body temperature rhythms over 24 h from subcutaneous probes implanted in null- (n = 6 for control, n = 5 for L-Sirt1/) or LbNOX-expressing (n = 5) control and liver-specific Sirt1/ mice on TRF-CR. Data are presented as mean values ± s.e.m. Source data
Fig. 4
Fig. 4. LbNOX redox state in liver drives energy conservation during nocturnal CR feeding.
TRF-CR generates rhythmic bouts of daytime torpor through increased levels of NADH in liver, inhibition of SIRT1 and downregulation of oxidative gene networks controlling acyl-carnitines and core body temperature. Reducing levels of NADH in the morning through the transduction of LbNOX in TRF-CR mice increases lipid oxidation through the activation of SIRT1, resulting in elevated daytime body temperature rhythms. These findings identify NAD(H) redox state as a link between TRF-CR and whole-body metabolism.
Extended Data Fig. 1
Extended Data Fig. 1. Circadian and metabolic profiling in TRF-CR mice.
a, (left) Representative wheel-running behaviour and (middle) quantification of average daily onset of activity for TRF-Reg (n = 5) and TRF-CR (n = 5) mice throughout the duration of the 4-wk intervention (double plotted for clarity). (right) Background-corrected PER2::LUCIFERASE readings from excised suprachiasmatic nucleus (SCN) of TRF-Reg (n = 6) and TRF-CR (n = 6) mice. The grey shading indicates mean values ± SEM. Experiments were performed in 4–6 mo old male C57BL6/J mice. b, Body weight of TRF-Reg (n = 15) and TRF-CR (n = 15) mice over the duration of the 4-wk study. c, Blood glucose and serum insulin during oral glucose tolerance testing performed during the daytime (at ZT4) in TRF-Reg (n = 6) and TRF-CR (n = 6) mice. d, GAP/DHAP mass isotopomer distribution determined by mass spectrometry of liver from TRF-Reg (n = 3) and TRF-CR (n = 2) mice 30 min following i.p. administration of a 10:1 mix of U13C-lactate:U13C-pyruvate (1 g/kg). e-f, (left) Relative concentration of (e) NADH and (f) NAD+ by HPLC in liver of TRF-CR compared to TRF-Reg mice during the day (ZT4) (n = 12 for TRF-Reg, n = 11 for TRF-CR,) and at night (ZT16) (n = 9 for each diet) for each time point. (right) Concentration of (e) NADH and (f) NAD+ in ad lib fed wild-type mouse liver by HPLC every 6 hrs for 24 hrs (n = 7). g-h, Log2-FC in daytime liver acyl-carnitine levels in (g) TRF-CR compared to TRF-Reg mice and (h) LbNOX- compared to null-overexpressing TRF-CR mice (n = 3). Data are presented as mean values ± SEM. Statistics were performed with unpaired, two-tailed student’s t test except as otherwise noted in the figure. *p < 0.5, ***p < 0.01.
Extended Data Fig. 2
Extended Data Fig. 2. Daytime NADH elevation regulates genome-wide transcription of fatty acid metabolism genes through BMAL1 during TRF-CR.
a, (left) RNA-seq reads per 10 million sequenced reads (RPM) that align to LbNOX in LbNOX- vs null-transduced TRF-Reg (n = 6 per genotype) and TRF-CR (n = 6 for null; n = 5 for LbNOX) liver. (right) Representative in situ immunohistochemistry against FLAG-LbNOX in liver of null- and LbNOX-transduced mice. Experiments were performed in 4–6 mo old male C57BL6/J mice unless otherwise noted. b, Heatmap depicting log2-FC in gene expression from TRF-CR and LbNOX in TRF-CR for genes within gluconeogenic and glycolytic gene ontology groups. c, Unbiased principal components analysis during the day (ZT4) or night (ZT16) of null- and LbNOX-expressing TRF-Reg or TRF-CR mice for genes that are differentially-expressed by TRF-CR in null mice at respective time points (DESeq2 p-adj<0.05) (n as in panel a). d, Log2-transformed FC in expression of select fatty acid metabolism genes during TRF-CR (blue) (n = 6), following LbNOX-overexpression during TRF-CR (white) (n = 5), and in Bmal1 knockout liver (purple) (n = 3). e, 24-hour fasted body temperatures in 4–6 mo old female liver-specific Bmal1fx/fx mice before and after retro-orbital administration of AAV8–TBG-iCre (n = 6) (left: ANOVA, p < 0.01; right: *p < 0.05 in paired, two-way student’s t test). f, Log2-transformed FC in expression of genes within the methionine pathway for conditions (n as in panel d). g, (top) Summary of methionine pathway and (bottom) metabolomics of effect of TRF-CR and LbNOX in TRF-CR on end-products of methionine metabolism in liver and serum performed during the daytime (ZT4) (n = 3). Data are presented as mean values ± SEM.
Extended Data Fig. 3
Extended Data Fig. 3. NADH inhibition of SIRT1 activity.
a, Lineweaver-Burk transformation of data from SAMDI-MS. b, Relative NADH quantified by HPLC following supplementation with pyruvate or lactate compared to controls (n = 3). c, Densitometric quantification of western blots from liver of 4–6 mo old male mice in indicated genotypes at ZT4 for p53-(K379)Ac (left) and FOXO1-(K242, K245, K262)Ac (right) relative to control TRF-Reg (from Fig. 3d) (n = 3). d, FOXO1 ChIP-seq from liver collected during the daytime comparing effect of FOXO1 binding in TRF-CR (x-axis) with the effect of hepatic SIRT1 ablation (L-Sirt1/) (y-axis). Each point indicates a FOXO1 peak in control liver. Peaks that have absolute log2(fold change) >0.5 for both comparisons are colored blue or black and counted by quadrant (n = 2). e, Densitometric quantification of western blots as in (c) relative to null-transduced TRF-Reg mice (from Fig. 3e) (Ac-p53: n = 4 for TRF-CR, LbNOX. n = 6 for all other conditions; Ac-FOXO1: n = 2 for TRF-CR, LbNOX. n = 3 for all other conditions). f, FOXO1 ChIP-seq from liver during the daytime comparing effect of TRF-CR (x-axis) (n = 3) with effect of (f) LbNOX in TRF-CR mice (y-axis) (n = 2) with coloring and counting as in panel d. (n = 2–3). g, Western blotting for Ac-FOXO1 during the nighttime (ZT16) in null- and LbNOX-transduced mice on TRF-Reg and TRF-CR (n.s. non-specific) and densitometric quantification (n = 3). h, BMAL1 ChIP-seq from liver during the daytime comparing the effect of TRF-CR and L-Sirt1/ as in panel d (n = 3). Uncropped Western blot scans labelled with molecular weight markers are presented in the Source Data Files. i, RNA-seq reads mapping to the exon of Sirt1 that is flanked by LoxP sites (Ex4) relative to exon 9 (Ex9) of Sirt1, and RNA-seq reads per 10 million sequenced reads (RPM) that align to iCre and LbNOX in null- or LbNOX-transduced TRF-CR mice co-transduced with iCre (n = 6). Null-expressing mice on TRF-CR (blue) (n = 5) are shown as reference. j, Quadrant plot comparing transcriptional responses to TRF-CR in null-transduced mice (x-axis) (n = 6) and LbNOX-expression in TRF-CR, L-Sirt1/ mice (y-axis) (n = 6). Each point indicates a gene that is DE by TRF-CR in null-transduced mice (930 genes). Percentages of genes within each quadrant are shown. k, Average 48-hour fasting body temperature in 4–6 mo old female liver-specific Sirt1/ mice (n = 5). Data are presented as mean values ± SEM. Statistics were performed with unpaired, two-tailed student’s t test except as otherwise noted in the figure. *p < 0.05. Source data

Comment in

References

    1. Bass J, Lazar MA. Circadian time signatures of fitness and disease. Science. 2016;354:994–999. - PubMed
    1. Koike N, et al. Transcriptional architecture and chromatin landscape of the core circadian clock in mammals. Science. 2012;338:349–354. - PMC - PubMed
    1. Zhang R, Lahens NF, Ballance HI, Hughes ME, Hogebesch JB. A circadian gene expression atlas in mammals: implications for biology and medicine. Proc. Natl Acad. Sci. USA. 2014;111:16219–16224. - PMC - PubMed
    1. Lin S-J, et al. Calorie restriction extends Saccharomyces cerevisiae lifespan by increasing respiration. Nature. 2002;418:344–348. - PubMed
    1. Weindruch R, Walford RL, Fligiel S, Guthrie D. The retardation of aging in mice by dietary restriction: longevity, cancer, immunity and lifetime energy intake. J. Nutr. 1986;116:641–654. - PubMed

Publication types