分享

夜宵怎么吃都是错的?夜间的限制饮食导致白天NADH升高和体温下降

 GCTA 2022-06-11 发布于贵州


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

NADH的SIRT1抑制在时间限制的进食过程中将能量状态与转录联系起来


|核心内容:

在哺乳动物中,昼夜节律被引导到光周期中,并驱动NAD+水平的每日振荡。NAD+是III类组蛋白去乙酰化酶sirtuin1(SIRT1)的共底物,与时钟转录因子相关。

虽然NAD+也参与氧化还原反应,但NAD(H)在多大程度上整合营养状态与昼夜转录周期尚不清楚。

在这里,我们发现,夜间动物仅在夜间限制热量限制饮食(TRF-CR),在夜间显示体温降低和白天肝脏NADH升高

通过转导短乳杆菌(LbNOX)形成水的NADH氧化酶,营养状态与NADH氧化还原状态的遗传解耦,增加了白天的体温以及血液和肝脏的酰基肉碱。

LbNOX在TRF-CR小鼠中的表达诱导大脑和肌肉arnt样蛋白1(BMAL1)和过氧化物酶体增殖物激活受体(PPARα)控制的氧化基因网络,并抑制氨基酸分解代谢途径。

酶学分析显示,NADH在体外抑制SIRT1,对应于体内TRF-CR过程中SIRT1底物去乙酰化的减少。

值得注意的是,即使NADH被LbNOX氧化,接受TRF-CR处理的Sirt1肝零合子动物也表现出持续的低温。

我们的研究结果表明,肝脏的NADH循环通过SIRT1的节律性调节,将营养状态与全身的能量学联系起来。

 Fig. 1 | Elevated NADH drives the daytime dip in body temperature during time-restricted calorie restriction.

Fig. 2 | Daytime NADH elevation regulates genome-wide transcription of fatty acid and amino acid metabolism genes during TRF-CR

 Fig. 3 | NADH inhibits SIRT1 in the morning during TRF-CR to regulate metabolism and body temperature.

 Fig. 4 | LbNOX redox state in liver drives energy conservation during nocturnal CR feeding.


 Although sirtuins have been associated with the beneficial effects  of CR across a range of organisms7,22,64,65, and CR promotes longevity  in yeast by increasing ySir2p activity and NAD+/NADH4,45, the situation is more complex in mammals, because CR exerts tissue-specific  effects on NAD+ and NADH22,66,67. 

Our findings using SAMDI-MS  indicate that NADH inhibits SIRT1 activity at physiologically relevant concentrations22,52–54, and our in vivo genetic analyses indicate  that hepatic NADH inhibition of SIRT1 during the daytime plays  an important role in energy-sparing in liver under a low-energy  state. 

Whether redox regulation of other sirtuin isoforms (SIRT2–7)  could also contribute to tissue-specific promotion of energy conservation during low-energy conditions remains to be explored. 

Future  studies are necessary to elucidate whether other energy-sensing  pathways influence the capacity of NADH to regulate the activity  of SIRT1 or other sirtuin isoforms during low-energy states in vivo.  

Our finding that the cytonuclear form of LbNOX drives SIRT1  activity in the nucleus to affect the transcriptional, metabolic and  physiological response to TRF-CR suggests that SIRT1 is sensitive  to changes in cytosolic NADH and/or to metabolic conditions that  cause export of mitochondrial NADH equivalents into the cytosol  as occurs during gluconeogenesis32–35. 

Although recent reports suggest that SIRT1 may interact with NADH in the nucleus68, continued development of methods that can accurately report subcellular  concentrations of reduced/oxidized NAD(H) in vivo will facilitate  greater understanding of how redox state across the day under different nutrient conditions may regulate SIRT1 and other sirtuin  isoforms16.
原文摘要:


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.


Metabolic homoeostasis in mammals is mediated by interlocked nutrient-sensing and temporal signals throughout  the 24-h light/dark cycle. 

The molecular clock network  drives oscillations of a broad range of transcripts and metabolites  that direct anabolic and catabolic metabolism in anticipation of  the fasting/feeding–sleep/wake cycle1–3 . 

Interactions between the  core molecular clock and nutrient-responsive transcription factors  (TFs) contribute to metabolic homoeostasis, yet how these pathways cooperate under long-term energy-deficient conditions has  remained obscure. 

In yeast, calorie restriction (CR) transcriptionally reprogrammes  metabolic gene expression to shift oxidative fuel preference and  maintain energetic homoeostasis of the cell4 . 

In mammals, CR  downregulates energy-intensive processes, such as thermogenesis  during sleep, and upregulates anabolic processes within the liver  that convert metabolite stores into energetic substrates for the  brain5,6 . 

NAD+ and the NAD+-dependent ySIR2p deacetylase are  required for the transcriptional response to CR in yeast7 , although  the role of NAD+ in the response to low-energy conditions in mammals remains less clear. 

Transcriptomic studies have revealed robust oscillations in the  expression of the rate-limiting enzyme involved in NAD+ biosynthesis across peripheral tissues, which in turn feedback to regulate  metabolic transcription cycles through SIRT1-mediated deacetylation and inhibition of the circadian repressor PER2 (refs. 8–13).  

Supplementation of NAD+ with the soluble precursor nicotinamide  riboside in ageing mice reverts senescence of the sleep/wake and  mitochondrial oxidative respiration cycles, suggesting that robustness of the NAD+–SIRT1-clock pathway may enhance fitness10.  

NAD+ also functions as an electron shuttle in oxidoreductase equilibrium reactions that vary according to both nutrient availability  and time of day14,15, yet whether metabolic control of rhythmic  transcription is modulated by cyclic changes in NAD(H) balance  remains unknown. 

To investigate whether NAD(H) balance provides a signal to regulate transcriptional and metabolic cycles in vivo, we examined the  effect of uncoupling nutrient state from NADH levels by tonically  inducing NADH oxidation during CR through hepatic transduction  of LbNOX16,17. 

When NADH levels were dissociated from energy state  using LbNOX, we observed transcriptional reprogramming of SIRT1-  and BMAL1-mediated gene networks that regulate fatty acid and  amino acid metabolism and thermogenesis. 

Our studies reveal that  NADH redox state drives energy conservation during sleep through  rhythmic inhibition of SIRT1 and downstream circadian processes.



参考文献:https:///10.1038/s42255-021-00498-1

 Data availability Data generated in this study are publicly available in the GEO repository  (GSE151281). We also utilized publicly accessible RNA-seq data from GEO  repositories GSE133989 and GSE118787. JASPAR databases are found at  http://jaspar./search?q=&collection=CORE&tax_group=vertebrates.  Correspondence and requests for materials should be addressed to Joseph Bass  (j-bass@northwestern.edu). Source data are provided with this paper. Received: 11 August 2021; Accepted: 28 October 2021;

--------------------------------------------------------------

--------------------------------------------------------------




    转藏 分享 献花(0

    0条评论

    发表

    请遵守用户 评论公约

    类似文章 更多