NAD Levels Are Not Constant


It can be tempting to think that something so important, so fundamental to how our cells function and survive, would be protected at all costs. But we know that NAD levels are in a state of constant flux, with cells actively creating and consuming NAD. And we know that we can’t take NAD for granted -- not getting enough dietary NAD precursors leads to a nasty deficiency disease called Pellagra - the direct result of not having enough NAD in our cells [1]. 

Research has also given us insights into how NAD levels may change in sickness and in health. A rapidly growing body of evidence is revealing that decreased NAD levels are associated with a wide variety of diseases and physiologic stresses. Excess alcohol consumption, excess UV exposure, sleep deprivation, poor diet, infection and a sedentary lifestyle are among the physiologic stressors linked to NAD depletion [2, 3]. Conversely, studies in rodents and people are showing that healthy habits may be associated with higher levels of this valuable cellular resource. Across organisms -- from single-celled yeast to mice to humans -- researchers have also documented declining NAD levels with age. While there is some debate over the root cause of this decline (i.e. chronological aging vs. an accumulation of stressors over time) it is becoming clear that NAD is not something we can take for granted and could use some extra support as we age and experience physiologic stress at the cellular level.

Healthy lifestyle choices are associated with higher levels of NAD

  • Eating a whole food, balanced diet with plenty of B3s prevents NAD deficiency/maintains NAD

  • Exercise is associated with increased NAD

  • Caloric restriction (fasting) is associated with increased NAD

  • Supplementation with NAD precursors can also boost NAD beyond what is required to avoid a vitamin deficiency [4].

NAD stands for nicotinamide adenine dinucleotide. It is a molecule found in every cell in the body. NAD is used to power metabolism by enabling the mitochondria – the ‘power stations’ of the cell to convert the food we eat into the energy our body needs to sustain all its functions. It is also required to “turn on” genes implicated in cells’ response to stress [5-7].

Declining NAD associated with every day physiologic stressors: aging, disease, and overnutrition/alcohol consumption

Research in animal models suggests that there are a number of lifestyle and environmental factors that impact natural NAD levels [8-15]. A growing number of studies in rodents and humans are showing decreased NAD levels in association with aging, physiologic stress and a whole host of disease states.

FIGURE 1. Values for “% of normal” represent the approximate percentage of NAD+ observed compared to a young or healthy control. These have been estimated from graphs when not stated in the cited reference. In most cases, a decrease in NAD+ cannot b…

FIGURE 1. Values for “% of normal” represent the approximate percentage of NAD+ observed compared to a young or healthy control. These have been estimated from graphs when not stated in the cited reference. In most cases, a decrease in NAD+ cannot be distinguished from a shift in the NAD+/NADH ratio. [6, 15-42]

 

How do NAD levels change?

NAD levels, which can be influenced by changes in absolute NAD/NADH levels or shifts in NAD/NADH ratios, help cells modify their behaviors in response to changes in energy, nutrient availability, and stress. Cells are constantly synthesizing and consuming NAD. Synthesis of NAD is through the De Novo Pathway, Preiss-Handler Pathway and Salvage (or NR) Pathway. Scientists at prestigious research institutions have been investigating NAD boosting strategies as a therapy for degenerative conditions related to aging. Pre-clinical research indicates that NAD plays a unique role in cells’ innate immune response, muscle and tissue protection, as well as increasing lifespan [43, 44].

FIGURE 2. CONSUMPTION AND BIOSYNTHESIS OF NAD Figure adapted from Fang, E.F., et al., NAD(+) in Aging: Molecular Mechanisms and Translational Implications. Trends Mol Med, 2017. 23(10): p. 899-916.

FIGURE 2. CONSUMPTION AND BIOSYNTHESIS OF NAD Figure adapted from Fang, E.F., et al., NAD(+) in Aging: Molecular Mechanisms and Translational Implications. Trends Mol Med, 2017. 23(10): p. 899-916.

 

NAD consumption through enzymes such as sirtuins, Poly(ADP-ribose) polymerases (PARPS), and NADases are some of the ways NAD levels decrease with age or physiologic stress [45-47].  For example, researchers have observed that in aging rodents, NAD consumption increases while NAD synthesis activity declines, consistent with overall net decreases in NAD levels observed in aging in rodents and humans [46, 48]. In addition, NAD levels have been shown to be decreased through overnutrition, alcohol consumption, viral infection and a sedentary lifestyle in clinical and preclinical studies [6, 15, 22, 25, 36, 38, 49, 50].


References

  1. Organization, W.H., Pellagra and its prevention and control in major emergencies. 2020, World Health Organization. p. 1-42

  2. Balard, B. and P.U. Giacomoni, Nicotinamide adenosine dinucleotide level in dimethylsulfate-treated or UV-irradiated mouse epidermis. Mutat Res, 1989. 219(1): p. 71-79.

  3. Levine, D.C., et al., NAD(+) Controls Circadian Reprogramming through PER2 Nuclear Translocation to Counter Aging. Mol Cell, 2020.

  4. Conze, D., C. Brenner, and C.L. Kruger, Safety and Metabolism of Long-term Administration of NIAGEN (Nicotinamide Riboside Chloride) in a Randomized, Double-Blind, Placebo-controlled Clinical Trial of Healthy Overweight Adults. Sci Rep, 2019. 9(1): p. 9772.

  5. Canto, C., et al., The NAD(+) precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity. Cell Metab, 2012. 15(6): p. 838-847.

  6. Gariani, K., et al., Eliciting the mitochondrial unfolded protein response by nicotinamide adenine dinucleotide repletion reverses fatty liver disease in mice. Hepatology, 2016. 63(4): p. 1190-1204.

  7. Zhang, H., et al., NAD(+) repletion improves mitochondrial and stem cell function and enhances life span in mice. Science, 2016. 352(6292): p. 1436-1443.

  8. Circu, M.L. and T.Y. Aw, Reactive oxygen species, cellular redox systems, and apoptosis. Free Radic Biol Med, 2010. 48(6): p. 749-762.

  9. de Murcia, J.M., et al., Requirement of poly(ADP-ribose) polymerase in recovery from DNA damage in mice and in cells. Proc Natl Acad Sci U S A, 1997. 94(14): p. 7303-7307.

  10. Massudi, H., et al., Age-associated changes in oxidative stress and NAD+ metabolism in human tissue. PLoS One, 2012. 7(7): p. e42357.

  11. Nakahata, Y., et al., Circadian control of the NAD+ salvage pathway by CLOCK-SIRT1. Science, 2009. 324(5927): p. 654-657.

  12. Ramsey, K.M., et al., Circadian clock feedback cycle through NAMPT-mediated NAD+ biosynthesis. Science, 2009. 324(5927): p. 651-654.

  13. Sasaki, Y., T. Araki, and J. Milbrandt, Stimulation of nicotinamide adenine dinucleotide biosynthetic pathways delays axonal degeneration after axotomy. J Neurosci, 2006. 26(33): p. 8484-8491.

  14. Theorell, H., Bonnichsen, R., The Mechanism of Alcohol Dehydrogenase Action. Acta Chemica Scandinavica, 1951. 5: p. 329.

  15. Trammell, S.A., et al., Nicotinamide Riboside Opposes Type 2 Diabetes and Neuropathy in Mice. Sci Rep, 2016. 6: p. 26933.

  16. Braidy, N., et al., Age related changes in NAD+ metabolism oxidative stress and Sirt1 activity in wistar rats. PLoS One, 2011. 6(4): p. e19194.

  17. Braidy, N., et al., Mapping NAD(+) metabolism in the brain of ageing Wistar rats: potential targets for influencing brain senescence. Biogerontology, 2014. 15(2): p. 177-198.

  18. Brown, K.D., et al., Activation of SIRT3 by the NAD(+) precursor nicotinamide riboside protects from noise-induced hearing loss. Cell Metab, 2014. 20(6): p. 1059-1068.

  19. Diguet, N., et al., Nicotinamide Riboside Preserves Cardiac Function in a Mouse Model of Dilated Cardiomyopathy. Circulation, 2017.137(21) p. :2256-2273.

  20. Escande, C., et al., Deleted in breast cancer-1 regulates SIRT1 activity and contributes to high-fat diet-induced liver steatosis in mice. J Clin Invest, 2010. 120(2): p. 545-558.

  21. Fang, E.F., et al., NAD(+) Replenishment Improves Lifespan and Healthspan in Ataxia Telangiectasia Models via Mitophagy and DNA Repair. Cell Metab, 2016. 24(4): p. 566-581.

  22. Frederick, D.W., et al., Increasing NAD synthesis in muscle via nicotinamide phosphoribosyltransferase is not sufficient to promote oxidative metabolism. J Biol Chem, 2015. 290(3): p. 1546-1558.

  23. Gerdts, J., et al., SARM1 activation triggers axon degeneration locally via NAD(+) destruction. Science, 2015. 348(6233): p. 453-457.

  24. Guan, Y., et al., Nicotinamide Mononucleotide, an NAD(+) Precursor, Rescues Age-Associated Susceptibility to AKI in a Sirtuin 1-Dependent Manner. J Am Soc Nephrol, 2017. 28(8): p. 2337-2352.

  25. Guest, J., et al., Changes in oxidative damage, inflammation and [NAD(H)] with age in cerebrospinal fluid. PLoS One, 2014. 9(1): p. e85335.

  26. Hou, Y., et al., NAD(+) supplementation normalizes key Alzheimer's features and DNA damage responses in a new AD mouse model with introduced DNA repair deficiency. Proc Natl Acad Sci U S A, 2018. p. e1876-1885.

  27. Koenekoop, R.K., et al., Mutations in NMNAT1 cause Leber congenital amaurosis and identify a new disease pathway for retinal degeneration. Nat Genet, 2012. 44(9): p. 1035-1039.

  28. Li, Y., et al., Corneal Denervation Causes Epithelial Apoptosis Through Inhibiting NAD+ Biosynthesis. Invest Ophthalmol Vis Sci, 2019. 60(10): p. 3538-3546.

  29. Mukherjee, S., et al., Nicotinamide adenine dinucleotide biosynthesis promotes liver regeneration. Hepatology, 2017. 65(2): p. 616-630.

  30. North, B.J., et al., SIRT2 induces the checkpoint kinase BubR1 to increase lifespan. EMBO J, 2014. 33(13): p. 1438-1453.

  31. Park, J.H., et al., Nicotinamide mononucleotide inhibits post-ischemic NAD(+) degradation and dramatically ameliorates brain damage following global cerebral ischemia. Neurobiol Dis, 2016. 95: p. 102-110.

  32. Ryu, D., et al., NAD+ repletion improves muscle function in muscular dystrophy and counters global PARylation. Sci Transl Med, 2016. 8(361): p. 361ra139.

  33. Sambeat, A., et al., Endogenous nicotinamide riboside metabolism protects against diet-induced liver damage. Nat Commun, 2019. 10(1): p. 4291.

  34. Scheibye-Knudsen, M., et al., A high-fat diet and NAD(+) activate Sirt1 to rescue premature aging in cockayne syndrome. Cell Metab, 2014. 20(5): p. 840-855.

  35. Seyedsadjadi, N., et al., High protein intake is associated with low plasma NAD+ levels in a healthy human cohort. PLoS One, 2018. 13(8): p. e0201968.

  36. Seyssel, K., et al., Regulation of energy metabolism and mitochondrial function in skeletal muscle during lipid overfeeding in healthy men. J Clin Endocrinol Metab, 2014. 99(7): p. E1254-1262.

  37. Shi, H., et al., NAD Deficiency, Congenital Malformations, and Niacin Supplementation. N Engl J Med, 2017. 377(6): p. 544-552.

  38. Wang, S., et al., Nicotinamide riboside attenuates alcohol induced liver injuries via activation of SirT1/PGC-1alpha/mitochondrial biosynthesis pathway. Redox Biol, 2018. 17: p. 89-98.

  39. Wang, X., et al., The Inhibitory Effects of Purple Sweet Potato Color on Hepatic Inflammation Is Associated with Restoration of NAD(+) Levels and Attenuation of NLRP3 Inflammasome Activation in High-Fat-Diet-Treated Mice. Molecules, 2017. 22(8): p. 1315.

  40. Yaku, K., K. Okabe, and T. Nakagawa, Simultaneous Measurement of NAD Metabolome in Aged Mice Tissue Using Liquid Chromatography Tandem-Mass Spectrometry (LC/MS/MS). Biomed Chromatogr, 2018. p. e4205.

  41. Yoshino, J., et al., Nicotinamide mononucleotide, a key NAD(+) intermediate, treats the pathophysiology of diet- and age-induced diabetes in mice. Cell Metab, 2011. 14(4): p. 528-536.

  42. Zhou, C.C., et al., Hepatic NAD(+) deficiency as a therapeutic target for non-alcoholic fatty liver disease in ageing. Br J Pharmacol, 2016. 173(15): p. 2352-2368.

  43. Belenky, P., et al., Nicotinamide riboside promotes Sir2 silencing and extends lifespan via Nrk and Urh1/Pnp1/Meu1 pathways to NAD+. Cell, 2007. 129(3): p. 473-484.

  44. Mouchiroud, L., et al., The NAD(+)/Sirtuin Pathway Modulates Longevity through Activation of Mitochondrial UPR and FOXO Signaling. Cell, 2013. 154(2): p. 430-441.

  45. Bock, F.J. and P. Chang, New directions in poly(ADP-ribose) polymerase biology. FEBS J, 2016. 283(22): p. 4017-4031.

  46. Camacho-Pereira, J., et al., CD38 Dictates Age-Related NAD Decline and Mitochondrial Dysfunction through an SIRT3-Dependent Mechanism. Cell Metab, 2016. 23(6): p. 1127-1139.

  47. Zhang, N. and A.A. Sauve, Regulatory Effects of NAD(+) Metabolic Pathways on Sirtuin Activity. Prog Mol Biol Transl Sci, 2018. 154: p. 71-104.

  48. Frederick, D.W., et al., Loss of NAD Homeostasis Leads to Progressive and Reversible Degeneration of Skeletal Muscle. Cell Metab, 2016. 24(2): p. 269-282.

  49. Canto, C., et al., Interdependence of AMPK and SIRT1 for metabolic adaptation to fasting and exercise in skeletal muscle. Cell Metab, 2010. 11(3): p. 213-219.

  50. Costford, S.R., et al., Skeletal muscle NAMPT is induced by exercise in humans. Am J Physiol Endocrinol Metab, 2010. 298(1): p. E117-126.