Trends in Cell Biology
Volume 24, Issue 8, August 2014, Pages 464-471
Journal home page for Trends in Cell Biology

Review
Special Focus – Metabolism
NAD+ and sirtuins in aging and disease

https://doi.org/10.1016/j.tcb.2014.04.002Get rights and content

Highlights

  • NAD+ plays a key role in regulating metabolism and circadian rhythm through sirtuins.

  • NAD+ becomes limiting during aging, affecting sirtuins’ activities.

  • NAD+ decline is likely to be due to a NAD+ biosynthesis defect and increased depletion.

  • Supplementing key NAD+ intermediates can restore NAD+ levels and ameliorate age-associated pathophysiologies.

Nicotinamide adenine dinucleotide (NAD+) is a classical coenzyme mediating many redox reactions. NAD+ also plays an important role in the regulation of NAD+-consuming enzymes, including sirtuins, poly-ADP-ribose polymerases (PARPs), and CD38/157 ectoenzymes. NAD+ biosynthesis, particularly mediated by nicotinamide phosphoribosyltransferase (NAMPT), and SIRT1 function together to regulate metabolism and circadian rhythm. NAD+ levels decline during the aging process and may be an Achilles’ heel, causing defects in nuclear and mitochondrial functions and resulting in many age-associated pathologies. Restoring NAD+ by supplementing NAD+ intermediates can dramatically ameliorate these age-associated functional defects, counteracting many diseases of aging, including neurodegenerative diseases. Thus, the combination of sirtuin activation and NAD+ intermediate supplementation may be an effective antiaging intervention, providing hope to aging societies worldwide.

Section snippets

NAD+ as an essential compound for many enzymatic processes

NAD+ was discovered more than a century ago by Sir Arthur Harden, as a low molecular weight substance present in a boiled yeast extract that could stimulate fermentation and alcohol production in vitro [1]. Subsequent studies over the next several decades determined that the structure of NAD+ comprised two covalently joined mononucleotides [nicotinamide mononucleotide (NMN) and AMP] and identified the keystone function of NAD+ and NADH as enzyme cofactors mediating hydrogen transfer in

NAD+ plays a key role in regulating metabolism and circadian rhythm

The canonical role of NAD+, mentioned above, is to facilitate hydrogen transfer in key metabolic pathways (Figure 1A). For example, NAD+ is converted to NADH in the glyceraldehyde 3-phosphate dehydrogenase step of glycolysis, a pathway in which glucose is converted to pyruvate. Conversion of NAD+ to NADH is also important in mitochondrial metabolism. In that compartment, NAD+ is converted to NADH in four steps of the mitochondrial tricarboxylic acid (TCA) cycle, in which acetyl-coenzyme A (CoA)

NAD+ declines with aging and can be restored by supplementation with NAD+ precursors

Several studies have reported that the activity of sirtuins decays with aging 26, 35, 36. The mammalian Sir2 ortholog SIRT1 can be regulated by many mechanisms, including transcriptionally, post-translationally by changes in stability, phosphorylation, and SIRT1-binding proteins, and by changes in NAD+ levels [14]. Of these mechanisms regulating SIRT1, a systemic decline in NAD+ has emerged as a likely explanation for why aging affects sirtuins. The decline in NAD+ was first noticed in

Possible mechanisms for how NAD+ levels decline in aging

Why do NAD+ levels decline with aging? One possibility is that one or more of the NAD+ biosynthetic pathways decline. There is some evidence that levels of NAMPT decline during aging [37], whereas exercise training has the opposite effect, at least in skeletal muscle [42]. Moreover, as discussed above, NAMPT is a major output of the circadian transcription factors BMAL and CLOCK. If the activity of the circadian machinery systemically declined with aging, as appears to be the case in the SCN

Mitochondria as a common target of aging-induced NAD+ decline

It is now clear that aging-induced inactivation of SIRT1 has a direct and deleterious effect on mitochondria, as first suggested by the important associations between SIRT1 and PGC-1α [48] and SIRT1 and TFAM [35]. A reduction in SIRT1 activity downregulates mitochondrial biogenesis, oxidative metabolism, and associated antioxidant defense pathways, leading to damage to complex I of the electron transport chain and a decline in mitochondrial function (Figure 4, right). A similar effect could

Prospects for treating neurodegenerative diseases?

Transgenic mice overexpressing SIRT1 throughout the body have been shown to counteract detrimental effects of energy-dense diet and aging and also mimic some physiological phenotypes induced by DR [11]. Furthermore, SIRT1 transgenic mice overexpressing this protein in the brain are protected in mouse models of Alzheimer's disease 53, 54, Parkinson's disease [55], and Huntington's disease 56, 57. In another mouse model, Wallerian degeneration slow (WldS) mice owe their heightened protection

Concluding remarks

Recent studies have indicated that NAD+ decline may drive aging through decreased sirtuin activities in the nucleus and mitochondria. NAD+ decline might be caused by the defect in NAMPT-mediated NAD+ biosynthesis and the PARP-mediated depletion of NAD+, both of which appear to occur during the aging process and perhaps in age-associated diseases, including neurodegenerative diseases. Supplementation of key NAD+ intermediates, such as NMN and NR, can ameliorate various age-associated

Disclaimer statement

S.I. had a sponsored research agreement with Oriental Yeast Co., Japan and is a cofounder of Metro Midwest Biotech. L.G. consults for GlaxoSmithKline, Chronos, Segterra, and Elysium Health.

Acknowledgments

The authors apologize to those whose work is not cited due to space limitations. They thank members of the Imai laboratory and the Guarente laboratory for critical discussions and suggestions. S.I. is supported by grants from the National Institute on Aging (AG024150, AG037457). L.G. is supported by the Glenn Foundation for Medical Research and grants from the National Institutes of Health.

References (71)

  • H.C. Chang et al.

    SIRT1 mediates central circadian control in the SCN by a mechanism that decays with aging

    Cell

    (2013)
  • S. Imai

    “Clocks” in the NAD world: NAD as a metabolic oscillator for the regulation of metabolism and aging

    Biochim. Biophys. Acta

    (2010)
  • P. Aksoy

    Regulation of intracellular levels of NAD: a novel role for CD38

    Biochem. Biophys. Res. Commun.

    (2006)
  • G.S. Young

    Decreased cADPR and increased NAD+ in the Cd38−/− mouse

    Biochem. Biophys. Res. Commun.

    (2006)
  • H.C. Lee

    Cyclic ADP-ribose and nicotinic acid adenine dinucleotide phosphate (NAADP) as messengers for calcium mobilization

    J. Biol. Chem.

    (2012)
  • A.P. Gomes

    Declining NAD+ induces a pseudohypoxic state disrupting nuclear–mitochondrial communication during aging

    Cell

    (2013)
  • J. Yoshino

    Nicotinamide mononucleotide, a key NAD+ intermediate, treats the pathophysiology of diet- and age-induced diabetes in mice

    Cell Metab.

    (2011)
  • P. Belenky

    Nicotinamide riboside promotes Sir2 silencing and extends lifespan via Nrk and Urh1/Pnp1/Meu1 pathways to NAD+

    Cell

    (2007)
  • P. Bieganowski et al.

    Discoveries of nicotinamide riboside as a nutrient and conserved NRK genes establish a Preiss–Handler independent route to NAD+ in fungi and humans

    Cell

    (2004)
  • C. Canto

    The NAD+ precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity

    Cell Metab.

    (2012)
  • G. Karamanlidis

    Mitochondrial complex I deficiency increases protein acetylation and accelerates heart failure

    Cell Metab.

    (2013)
  • T. Singh et al.

    Inflammatory markers in population studies of aging

    Ageing Res. Rev.

    (2011)
  • P. Bai

    PARP-1 inhibition increases mitochondrial metabolism through SIRT1 activation

    Cell Metab.

    (2011)
  • A. Berdichevsky

    C. elegans SIR-2.1 interacts with 14-3-3 proteins to activate DAF-16 and extend life span

    Cell

    (2006)
  • G. Donmez

    SIRT1 suppresses beta-amyloid production by activating the alpha-secretase gene ADAM10

    Cell

    (2010)
  • U. Rass

    Defective DNA repair and neurodegenerative disease

    Cell

    (2007)
  • B. Gong

    Nicotinamide riboside restores cognition through an upregulation of proliferator-activated receptor-gamma coactivator 1alpha regulated beta-secretase 1 degradation and mitochondrial gene expression in Alzheimer's mouse models

    Neurobiol. Aging

    (2013)
  • Y. Wang et al.

    Overlapping and distinct functions for a Caenorhabditis elegans SIR2 and DAF-16/FOXO

    Mech. Ageing Dev.

    (2006)
  • M. Gellert

    Joining of DNA strands by DNA ligase of E. coli

    Cold Spring Harb. Symp. Quant. Biol.

    (1968)
  • A. De Flora

    Autocrine and paracrine calcium signaling by the CD38/NAD+/cyclic ADP-ribose system

    Ann. N. Y. Acad. Sci.

    (2004)
  • S. Imai

    Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase

    Nature

    (2000)
  • S-J. Lin

    Calorie restriction extends yeast life span by lowering the level of NADH

    Genes Dev.

    (2004)
  • S. Imai et al.

    The importance of NAMPT/NAD/SIRT1 in the systemic regulation of metabolism and ageing

    Diabetes Obes. Metab.

    (2013)
  • N. Curtin

    PARP inhibitors for anticancer therapy

    Biochem. Soc. Trans.

    (2014)
  • M.C. Haigis et al.

    Mammalian sirtuins: biological insights and disease relevance

    Annu. Rev. Pathol.

    (2010)
  • Cited by (953)

    • The therapeutic perspective of NAD<sup>+</sup> precursors in age-related diseases

      2024, Biochemical and Biophysical Research Communications
    • The enzymes of serine synthesis pathway in cancer metastasis

      2024, Biochimica et Biophysica Acta - Molecular Cell Research
    View all citing articles on Scopus
    View full text