ArticlePDF Available

The Role of NAD+ in Anti-Aging Therapies

Authors:
446
The Role of NAD+ in Anti-Aging Therapies
Xuqian Liu1 and Taosheng Huang1,2*
1Human Aging Research Institute, Nanchang University, China
2Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, China
*Corresponding author: Taosheng Huang, Human Aging Research Institute, Nanchang University, Nanchang 330031 and Division of
Human Genetics, Cincinnati Children’s Hospital Medical Center, China.
To Cite This Article: Taosheng Huang, The Role of NAD+ in Anti-Aging Therapies. Am J Biomed Sci & Res. 2019 - 6(5). AJBSR.MS.ID.001072.
DOI: 10.34297/AJBSR.2019.06.001080.
Received: December 11, 2019 ; Published: December 20, 2019
Copy Right@ Taosheng Huang
This work is licensed under Creative Commons Attribution 4.0 License
AJBSR.MS.ID.001080.
American Journal of
Biomedical Science & Research
www.biomedgrid.com
---------------------------------------------------------------------------------------------------------------------------------
ISSN: 2642-1747
Research Article
Abstract
The aging process involves accumulation of DNA damage, mitochondrial defects, progressive tissue degeneration and atrophy, and the
development of metabolic dysfunction and weakness. Aging is also accompanied by decreased levels of nicotinamide adenine dinucleotide (NAD+),
which can result in cell damage and even shorter life spans. NAD+ acts as an enzyme cofactor in many essential biological pathways and is a substrate
for several regulatory proteins. Many studies have suggested that the upregulation of NAD+ precursors can increase levels of NAD+ in tissues or cells
+. Here we provide a review of NAD+ metabolism and its role in
aging-related therapy.
Keywords: NAD+; Aging; Clinical trials
Introduction
-
sidered to be an irreversible process. Aging of an organism is ac-
companied by metabolic disorders and the impairment of physio-
logical function, as well as the development of age-related diseases
[1-4]. There is abundant evidence that NAD+ plays an important role
in aging, as it is involved in various biological functions and is a key
regulator of stress resistance [5,6]. Levels of NAD+ steadily decline
with age, resulting in altered metabolism and increased disease
susceptibility [7-9]. NAD+ plays a key role in various energy metab-
olism pathways [6,10]. Additionally, NAD+ is a cofactor for many en-
zymes, such as poly (ADP-ribose) polymerases (PARPs), CD38, and
sirtuins [11]. Sirtuins are NAD+-dependent histone deacetylase for
a wide range of transcriptional regulators [10,12]. Overexpression
of SIRT1 in the brains of mice has been shown to delay aging [13].
PARP is a major NAD+-degrading enzyme, which plays diverse roles
in many molecular and cellular processes [14]. Inhibition of PARP-1
increases mitochondrial metabolism via modulation of SIRT1 activ-
ity [15]. Another NAD+-degrading enzyme, CD38, had been associ-
ated with the decline in NAD+ levels during aging [16].
Mammalian cells cannot import NAD+ in vivo, so they must
synthesize it either from tryptophan or the various forms of niacin
taken up in the diet including nicotinamide mononucleotide (NMN)
and nicotinamide riboside (NR) [17-20]. Recently, it was found in
mice that supplementing with NAD+ precursors (including NMN,
NR, and nicotinamide) or inhibiting the activity of NAD+-consum-
ing enzymes can increase the level of NAD+ in tissues and improve
energy metabolism, thereby delaying aging and extending healthy
life [15,21-24].
Currently, the anti-aging activity of NAD+ precursors is primar-
ily evaluated through measurement of aging markers in mouse
behavior, accumulation of DNA damage, and mitochondrial activi-
ty. RNA sequencing has also been used to identify genes and path-
ways involved in the anti-aging mechanisms of NAD+ [20,22,25,29].
Furthermore, recent research has shown that biological age can be
measured by analyzing the 353 DNA methylation sites of the Hor-
vath clock [30,31].
NAD+ Biosynthesis-Salvage Pathway
In vivo, NAD+ is an essential cofactor of dehydrogenase [32,33].
Nicotinamide coenzyme is an electron carrier which plays an im-
portant role in various oxidation-reduction reactions. Therefore
NAD+ is a cofactor of many key enzymes in glycolysis, the tricarbox-
ylic acid cycle, and oxidative phosphorylation [34]. Age-associated
American Journal of Biomedical Science & Research
Am J Biomed Sci & Res Copy@ Taosheng Huang
447
decline in NAD+ availability has an important effect on the aging
process of many species [8,35,36]. There are three pathways for the
synthesis of NAD+ in cells, involving many different precursors [37-
39]. Here we focus on the salvage pathway, which is important from
a translational research perspective because it is the main source
of NAD+ [40,41].
There are three pathways for the synthesis of NAD+ in cells:
a. de novo from tryptophan;
b. from nicotinic acid via the Preiss-Handler pathway; and
c. from nicotinamide (NAM) via the salvage pathway [37].
NAM itself is a by-product of NAD+-degrading enzymes such as
       -
vage pathway is catalyzed by nicotinamide phosphoribosyl-trans-
ferase (NAMPT), which converts nicotinamide and 5-phosphoribo-
syl-1-pyrophosphate into NMN [42]. Subsequently, nicotinamide
mononucleotide adenylyltransferase (NMNAT) produces NAD+
from NMN and ATP [43,44]. NR can be converted by nicotinamider-
iboside kinase (NRK) into NMN which participating in the Salvage
pathway [45]. NAMPT is the rate-limiting enzyme of the salvage
pathway [42]. It has been hypothesized that reduced NAD+ synthe-
sis is one of the causes of lower NAD+ levels with aging, and this
may be due to decreased activity of NAMPT [42,46]. Indeed, NAMPT
levels are known to decline with age in many types of tissues [47-
49], whereas exercise increases skeletal muscle NAMPT expression
[50].
Figure 1: The Salvage pathway NAM and NR are the main precursor for the salvage pathway.
In mammals, NAMPT has two different forms: intra- and ex-
tracellular [51]. The intracellular form is the one that participates
in the salvage pathway of NAD+ synthesis [42], while the extra-
cellular form likely functions as a circulating cytokine [52]. Stud-
ies have shown that secretion of NAMPT is regulated by SIRT1 in
vivo [53,54], and SIRT1 activity in turn depends on NAMPT which
regulates level of NAD+ [55]. Increasing level of NAMPT may delay
aging of individuals via SIRT1-dependent pathways [56]. NAMPT
has been shown to regulate osteoblast differentiation in primary
culture of mouse bone marrow-derived mesenchymal stem cells via
NAD+     
bone aging or fractures [48,57-59].
        

in aged mice resulted in increased levels of circulating eNAMPT, in-
creased levels of NAD+ in multiple tissues, and extended lifespan
[60].
In mammals NMNAT is the central enzyme of the NAD bio-
synthetic pathway [43,62]. There are three isoforms, NMNAT1,
NMNAT2, and NMNAT3, encoded by different genes and localized
to nucleus, Golgi apparatus, and mitochondria, respectively [43].
NMNAT1 directly control SIRT1 deacetylase activity at a set of
target gene promoters [63]. Homozygous knockout of Nmnat1 in
mice results in embryonic death [64]. Low levels of NMNAT2, high-
ly expressed in the brain and nervous system, could impair axon
regeneration as well as axon survival in aging and disease [65,66].
-
chondrial NAD+ biosynthesis [67]. In addition, down-regulation of

for mitochondrial respiration, suggesting that NMNAT3 plays a key
role in mitochondrial NAD+ homeostasis [68].
Anti-aging effects of NAD+
Through its role as a substrate for sirtuins, CD38, and PARP,
NAD+ regulates a variety of cellular process including energy me-
tabolism, DNA damage repair, gene expression, and oxidative stress
response [11,34,69,70].
a. The Sirtuins Pathway
Sirtuins are evolutionarily conserved NAD+-dependent deacety-
lases. Increasing sirtuin expression has been shown to affect lifes-
Am J Biomed Sci & Res Copy@ Taosheng Huang
American Journal of Biomedical Science & Research
448
pan across various species [13,36,71,72]. Sirtuins have received
 
silent information regulator 2 (Sir2) can extend yeast lifespan [72].
The closest mammalian homologue of this regulator is SIRT1[69],
mainly localized in the nucleus but also present in cytosol [73]. Its

circumstance [12]. It has been shown in vivo that NAD+-SIRT1 sig-
naling promotes mitochondrial activity [25]. Previous research has
suggested that increasing SIRT1 in the brain, especially in the dor-
somedial and lateral hypothalamic nuclei, can delay aging and ex-
-
sion of Sirt1 was found to delay aging and protect against oxidative
stress in the heart [74]. NR-SIRT1 signaling can inhibit cardiac stem
cell senescence by improving mitochondrial function and muscle
stem cell function, thereby enhancing life span in mice [25]. More
research is needed to determine whether increased level of NAD+ in
vivo can improve SIRT1 activity, thereby delaying aging [75].
b. The PARPs Pathway
PARPs are expressed by most eukaryotic cells and are involved
in DNA damage detection and repair, cell death pathways and so on
[14]. Aging is associated with an accumulation of DNA damage [76].
Depletion of NAD+ is involved in cell death through PARP-1 [70]. Al-
though this enzyme plays an important role in cells, over-activation
of PARP-1 can lead to depletion of NAD+, reduction of ATP, reducing
the activity of SIRT1, loss of mitochondrial function, and even cell
death [70,77,78]. Increased level of NAD+, when SIRT1 is intact, can
reduce the cell death caused by activation of PARP-1 in cardiac myo-
cyte [79].
c. CD38 and NAD+
CD38 is a multi-functional protein. Studies have shown that
CD38 is the NADase in mammalian tissues [80,81]. It is thought
to contribute to the age-related decline in NAD+ levels [23,80,82].
CD38 also acts as an antigen for B-lymphocyte activation and as an
  -
cent cells are known to express small molecules including secret-

       
process, known as the senescence-associated secretory phenotype
[84-86], involves secretion of factors by senescent cells which in-
duce the expression of CD38 in non-senescent cells [82,87]. This
+
and its reduced form, NADH, within a cell [82]. Recently, the small
molecule CD38 inhibitor 78c was shown to reverse the age-related
loss of NAD+ [28,83]. By increasing tissue levels of NAD+, 78c may
be able to ameliorate metabolic disorder and other disruptions in-
volved in the aging process. In addition, animals treated with 78c
show activation of longevity genes, which inhibit DNA damage [28].
NAD+ and NADH are in dynamic equilibrium within the cell
[75]. Intracellular NAD+ can be increased in vivo through oral ad-
ministration of NAD+ precursor or by inhibiting the degradation of
NAD+ [15,23,88]. Regulation of the NAD+/ NADH ratio in this way
can improve mitochondrial function and has been shown to treat
senile deafness in elderly mice [89].
NAD+ Repletion and Aging
One of the major causes of aging is progressive tissue degen-
eration and atrophy due to reduced somatic or stem cell function
[22,90,91]. Adult stem cells are not only essential in continuously
proliferating tissues (such as hematopoietic, intestinal, and skin
systems) but also in normally quiescent tissues (such as skeletal
muscle and the brain) that require regeneration after damage or
exposure to disease [92]. NR supplementation improved meta-
bolic function in muscle and neural stem cells, in both young and
old mice, thereby increasing lifespan [25]. NR treatment has also
been shown to rejuvenate stem cells from aged mice and restore
the impaired ability to repair gut damage [22]. Previous studies
have shown that DNA damage of nerve cells, nerve stem cells, and
muscle stem cells in mice can be reduced by NR supplementation
[25,93]. NR has also been shown to ameliorate mitochondrial dys-
function and enhance oxidative metabolism in obese mice [94,95]
and prolong the lifespan of mice through neuronal DNA repair and
mitochondrial quality improvement [96,97].
Supplementation with NMN can restore age-related capillary

novel therapy to restore SIRT1 activity and reverse age-related arte-
rial dysfunction by reducing oxidative stress [98,99]. Mitochondrial
disorders due to impaired oxidative phosphorylation (OXPHOS) are
a cause of aging [100]. Long-term treatment with NMN in elderly
C57BL/6J mice can improve metabolic dysfunction and ameliorate
age-associated physiological decline [20]. NMN can also restore
mitochondrial function, prevent neural death, and delay cognitive
decline in a mouse model of Alzheimer’s disease [101,102]. Sup-
plementation with NAM was shown to improve blood sugar levels
and metabolic capacity in HFD-fed mice. However, it had no effect
on lifespan [103].
There are data showing that supplementation with NAD+ pre-
cursors enhanced the mitochondrial function of cells or stem cells
in a SIRT1-dependent manner [25,94]. Furthermore, supplement-
ing NAD+ precursors in elderly mice improved mitochondrial func-
tion in hematopoietic stem cells and muscle stem cells, as well as

the biological activity of mesenchymal stromal cells through the up-
regulation of SIRT1, thereby stimulating osteogenesis of the cells
and protecting bone from aging to delay the aging of mice [105]. In
elderly mice, NMN treatment improved capillary density through
the NAD+-H2
and physiological status [27,29,106].
Inhibition of some NAD+-degrading enzymes could also lead
to increased levels of NAD+ [15,23]. CD38, PARPs, and SARM1 all
American Journal of Biomedical Science & Research
Am J Biomed Sci & Res Copy@ Taosheng Huang
449
degrade NAD+ inside the cell [23,77,80,107]. The activity of CD38
increases with aging, contributing to the age-related decline in
NAD+ [16]. Several small-molecule inhibitors of CD38 have been
described [83,108-110]. Thiazoloquin(az)olin(on)e is one such in-
hibitor which could potentially be used as a therapeutic agent to in-
crease intracellular NAD+ level [28]. Inhibition of PARP1 has recent-
ly been reported to correct mitochondrial impairment [111,112]
and has strong metabolic implications through its modulation of
SIRT1 activity [15].
Measure Biological Age
Thus far, the anti-aging activity of NAD+ is mainly examined us-
ing RNA sequencing and gene set enrichment analysis to identify
pathways of candidate biomarkers [20,22,25]. However, there is
not yet a gold standard for aging biomarkers. The DNA clock may
offer a better objective biomarker for the study of aging [30,31].
DNA methylation plays a critical role in the regulation of gene tran-
scription [113-118]. Senescence can be predicted and evaluated by
detecting cytosine-5 methylation within CpG dinucleotides [30,31].
These age-related CpG characteristics are independent of gender or
tissue type. Recent research has shown that biological age can be
approximated by measuring levels of DNA methylation, a process
known as the Horvath (or DNA) clock [30,31,119,120]. Age-related
   -
parison of thousands of CpG sites in Illumina Bead Chip microarray
data [121,122]. Many of these age-associated CpG sites were then
used as epigenetic age-predictors [30,31,120,123]. Three hun-
    
of biological age, independent of chromatin status or tissue source
[30,31].
Petkovich et al. developed a robust predictor of mouse biologi-
cal age based on 90 CpG sites derived from partial blood DNA meth-
      -
sue predictor to estimate age based on DNA methylation at 329
unique CpG sites from various different mouse tissues [125]. One
group claims to have found three methylation sites, Prima1, Hsf4,
and Kcns1, which are enough to predict biological age in mice [126].
However, this study has yet to be replicated. The most accurate
clock results from applying elastic net regression to all CpGs for
multi-tissue in mice [127].
Together these studies suggest that the DNA clock provides an
objective biomarker for the study of aging [30,31]. Recently, met-
formin has shown that reversed subject’s biological age, based on
assessment of Horvath clock [128,129]. Its use will allow us to ex-
amine the anti-aging effectiveness of NAD+ and its precursors more
objectively and accurately.
Clinical Research
NAD+ precursors can be delivered orally to humans or ani-
mals to alter the dynamic balance of NAD+/NADH in vivo [24,130].
Preliminary clinical studies in humans showed that NR supple-
mentation could improve muscle NAD+ metabolism in the elderly
[131,132]. Healthy volunteers, who underwent an 8-day course
of NR, with doses increasing from 250 mg to 1000 mg, showed
increased levels of circulating NAD+ and experienced no adverse
side effects [133]. Similarly, NR supplementation increased NADH
and NADPH levels and improved exercise performance in elderly
subjects [134]. Therefore, NMN is considered safe in clinical trials
[135]. However, high dose supplementation with NAD+ precursors
may increase rates of glycolysis and mitochondrial respiratory me-
 -
tokines in cells [136]. Thus, use of supplements should be carefully
observed to ensure that they strike a proper balance between an-
ti-aging effects and potential detrimental effects.
Conclusions
NAD+ is a cofactor for many important enzymes. Reduced
levels of NAD+ have been associated with aging. Evidence suggests
that supplementation with NAD+ precursors, or inhibition of
NAD+ degradation, could improve metabolic function. While
supplementation with NAD+ precursors has been found to delay
aging in mice, anti-aging effects of NAD+ have yet to be demonstrated
in human subjects. Use of a more accurate biomarker for aging,
     

References
1. Fraga MF, Agrelo R, Esteller M (2007) Crosstalk between aging and
cancer: the epigenetic language. Ann N Y Acad Sci 1100: 60-74.
2. Fraga MF, Esteller M (2007) Epigenetics and aging: the targets and the
marks. Trends Genet 23(8): 413-418.
3. Braidy N, Grant R, Sachdev PS (2018) Nicotinamide adenine
dinucleotide and its related precursors for the treatment of Alzheimer’s
disease. Curr Opin Psychiatry 31(2): 160-166.
4. Fuellen G, Jansen L, Cohen AA, Walter Luyten, Manfred Gogol, et al.
(2019) Health and Aging: Unifying Concepts, Scores, Biomarkers and
Pathways. Aging Dis 10(4): 883-900.
5. Hosseini L, Vafaee MS, Mahmoudi J, Badalzadeh R (2019) Nicotinamide
adenine dinucleotide emerges as a therapeutic target in aging and
ischemic conditions. Biogerontology 20(4): 381-395.
6. Ying W (2008) NAD+/NADH and NADP+/NADPH in cellular functions
and cell death: regulation and biological consequences. Antioxid Redox
Signal 10(2): 179-206.
7.            
Pathological Factor of a Number of Diseases and Aging: Mechanisms
and Therapeutic Implications. Antioxid Redox Signal 30(6): 890-905.
8. Mouchiroud L, Houtkooper RH, Moullan N, Katsyuba E, Ryu D, et al.
(2013) The NAD (+)/Sirtuin Pathway Modulates Longevity through
Activation of Mitochondrial UPR and FOXO Signaling. Cell 154(2): 430-
441.
9.            
stem cells recapitulates their functional defects during aging. EMBO
J 33(12): 1321-1340.
10. Houtkooper RH, Pirinen E, Auwerx J (2012) Sirtuins as regulators of
metabolism and healthspan. Nat Rev Mol Cell Biol 3(4): 225-238.
11. Katsyuba E, Auwerx J (2017) Modulating NAD (+) metabolism, from
bench to bedside. EMBO J 36(18): 2670-2683.
Am J Biomed Sci & Res Copy@ Taosheng Huang
American Journal of Biomedical Science & Research
450
12. Tanno M, Sakamoto J, Miura T, Shimamoto K, Horio Y (2007)
Nucleocytoplasmic shuttling of the NAD+-dependent histone
deacetylase SIRT1. J Biol Chem 282(9): 6823-6832.
13. Satoh A, Brace CS, Rensing N, Cliften P, Wozniak DF, et al. (2013) Sirt1
extends life span and delays aging in mice through the regulation of
Nk2 homeobox 1 in the DMH and LH. Cell Metab 18(3): 416-430.
14. Kim MY, Zhang T, Kraus WL (2005) Poly (ADP-ribosyl)ation by PARP-
1: ‘PAR-laying’ NAD+ into a nuclear signal. Genes Dev 2005; 19(17):
1951-1967.
15. Bai P, Canto C, Oudart H, Attila Brunyánszki, Yana Cen, et al. (2011)
PARP-1 inhibition increases mitochondrial metabolism through SIRT1
activation. Cell Metab 13(4): 461-468.
16. Camacho Pereira J, Tarrago MG, Chini CCS, Nin V, Escande C, et al.
(2016) CD38 Dictates Age-Related NAD Decline and Mitochondrial
Dysfunction through an SIRT3-Dependent Mechanism. Cell Metab
23(6): 1127-1139.
17. Trammell SA, Yu L, Redpath P, Migaud ME, Brenner C (2016)
Nicotinamide Riboside Is a Major NAD+ Precursor Vitamin in Cow
Milk. J Nutr 146(5): 957-963.
18. Bogan KL, Brenner C (2008) Nicotinic acid, nicotinamide, and
nicotinamide riboside: a molecular evaluation of NAD+ precursor
vitamins in human nutrition. Annu Rev Nutr 28: 115-130.
19. Ummarino S, Mozzon M, Zamporlini F, Amici A, Mazzola F, et al. (2017)
Simultaneous quantitation of nicotinamide riboside, nicotinamide
mononucleotide and nicotinamide adenine dinucleotide in milk by a
novel enzyme-coupled assay. Food Chem 221: 161-168.
20. Mills KF, Yoshida S, Stein LR, Grozio A, Kubota S, et al. (2016) Long-
Term Administration of Nicotinamide Mononucleotide Mitigates Age-
Associated Physiological Decline in Mice. Cell Metab 24(6): 795-806.
21. Chaturvedi P, Tyagi SC (2018) NAD (+): A big player in cardiac and
skeletal muscle remodeling and aging. J Cell Physiol 233(3): 1895-
1896.
22. Igarashi M, Miura M, Williams E, Jaksch F, Kadowaki T, et al. (2019)
NAD (+) supplementation rejuvenates aged gut adult stem cells. Aging
Cell 18(3): e12935.
23. Chini EN, Chini CCS, Espindola Netto JM, de Oliveira GC, van Schooten
W (2018) The Pharmacology of CD38/NADase: An Emerging Target in
Cancer and Diseases of Aging. Trends Pharmacol Sci 39(4): 424-436.
24. Yoshino J, Baur JA, Imai SI (2018) NAD (+) Intermediates: The Biology
and Therapeutic Potential of NMN and NR. Cell Metab 2018; 27(3):
513-528.
25. Zhang H, Ryu D, Wu Y, Gariani K, Wang X, et al. (2016) NAD+ repletion
improves mitochondrial and stem cell function and enhances life span
in mice. Science 352(6292): 1463-1443.
26. Tarantini S, Valcarcel-Ares MN, Toth P, Yabluchanskiy A, Tucsek Z, et
al. (2019) Nicotinamide mononucleotide (NMN) supplementation
rescues cerebromicrovascular endothelial function and neurovascular
coupling responses and improves cognitive function in aged mice.
Redox Biol 24: 101192.
27. Das A, Huang GX, Bonkowski MS, Longchamp A, Li C, et al. (2018)
Impairment of an Endothelial NAD (+)-H2S Signaling Network Is a
Reversible Cause of Vascular Aging. Cell 173(1): 74-89.
28. Tarrago MG, Chini CCS, Kanamori KS, Warner GM, Caride A, et al.

Metabolic Dysfunction by Reversing Tissue NAD (+) Decline. Cell
Metab 27(5): 1081-1095.
29. Das A, Huang GX, Bonkowski MS et al. (2019) Impairment of an
Endothelial NAD (+)-H2S Signaling Network Is a Reversible Cause of
Vascular Aging. Cell 176: 944-945.
30. Horvath S (2013) DNA methylation age of human tissues and cell
types. Genome Biol 14(10): R115.
31. Horvath S (2015) Erratum to: DNA methylation age of human tissues
and cell types. Genome Biol 16: 96.
32. Xiao W, Wang RS, Handy DE, Loscalzo J (2018) NAD(H) and NADP(H)
Redox Couples and Cellular Energy Metabolism. Antioxid Redox Signal
28(3): 251-272.
33. Yaku K, Okabe K, Nakagawa T (2018) NAD metabolism: Implications in
aging and longevity. Ageing Res Rev 47: 1-17.
34. Berger F, Ramirez-Hernandez MH, Ziegler M. The new life of a
centenarian: signalling functions of NAD(P). Trends Biochem Sci
29(3): 111-118.
35. Imai SI (2016) The NAD World 2.0: the importance of the inter-tissue
communication mediated by NAMPT/NAD(+)/SIRT1 in mammalian
aging and longevity control. NPJ Syst Biol Appl 2: 16018.
36. Imai S, Guarente L (2014) NAD+ and sirtuins in aging and disease.
Trends Cell Biol 24(8): 464-471.
37. Verdin E (2015) NAD (+) in aging, metabolism, and neurodegeneration.
Science 350(6265): 1208-1213.
38. Chi Y, Sauve AA (2013) Nicotinamide riboside, a trace nutrient in foods,
is a vitamin B3 with effects on energy metabolism and neuroprotection.
Curr Opin Clin Nutr Metab Care 16(6): 657-661.
39. Bieganowski P, Brenner C (2004) Discoveries of nicotinamide riboside
as a nutrient and conserved NRK genes establish a Preiss-Handler
independent route to NAD+ in fungi and humans. Cell 117(4): 495-
502.
40. Kennedy BE, Sharif T, Martell E, Dai C, Kim Y, et al. (2016) NAD (+)
salvage pathway in cancer metabolism and therapy. Pharmacol Res
114: 274-283.
41. Houtkooper RH, Canto C, Wanders RJ, Auwerx J (2010) The secret
life of NAD+: an old metabolite controlling new metabolic signaling
pathways. Endocr Rev 31(2): 194-223.
42. Revollo JR, Grimm AA, Imai S (2004) The NAD biosynthesis pathway
mediated by nicotinamide phosphoribosyltransferase regulates Sir2
activity in mammalian cells. J Biol Chem 279(49): 50754-50763.
43. Berger F, Lau C, Dahlmann M, Ziegler M (2005) Subcellular
compartmentation and differential catalytic properties of the three
human nicotinamide mononucleotide adenylyltransferase isoforms. J
Biol Chem 280(43): 36334-36341.
44. Schweiger M, Hennig K, Lerner F, Niere M, Hirsch-Kauffmann M, et
al. (2001) Characterization of recombinant human nicotinamide
mononucleotide adenylyl transferase (NMNAT), a nuclear enzyme
essential for NAD synthesis. FEBS Lett 492(1-2): 95-100.
45. Canto C, Menzies KJ, Auwerx J (2015) NAD (+) Metabolism and the
Control of Energy Homeostasis: A Balancing Act between Mitochondria
and the Nucleus. Cell Metab 22(1): 31-53.
46. van der Veer E, Ho C, O’Neil C, Barbosa N, Scott R, et al. (2007) Extension
of human cell lifespan by nicotinamide phosphoribosyltransferase. J
Biol Chem 282(15): 10841-10845.
47. Liu LY, Wang F, Zhang XY, Huang P, Lu YB, et al. (2012) Nicotinamide
phosphoribosyltransferase may be involved in age-related brain
diseases. PLoS One 7(10): e44933.
48. Frederick DW, Loro E, Liu L, Davila A Jr, Chellappa K, et al. (2016) Loss
of NAD Homeostasis Leads to Progressive and Reversible Degeneration
of Skeletal Muscle. Cell Metab 24(2): 269-282.
49.            
(2017) Nicotinamide adenine dinucleotide biosynthesis promotes
liver regeneration. Hepatology 65(2): 616-630.
50. Costford SR, Bajpeyi S, Pasarica M, Albarado DC, Thomas SC, et al.
(2010) Skeletal muscle NAMPT is induced by exercise in humans. Am J
Physiol Endocrinol Metab 298(1): E117-E126.
American Journal of Biomedical Science & Research
Am J Biomed Sci & Res Copy@ Taosheng Huang
451
51. Revollo JR, Korner A, Mills KF, Satoh A, Wang T, et al. (2007) Nampt/
PBEF/Visfatin regulates insulin secretion in beta cells as a systemic
NAD biosynthetic enzyme. Cell Metab 6(5): 363-375.
52. Imai S (2009) Nicotinamide phosphoribosyltransferase (Nampt): a
link between NAD biology, metabolism, and diseases. Curr Pharm Des
15(1): 20-28.
53. Yoon MJ, Yoshida M, Johnson S, Takikawa A, Usui I, et al. (2015)
SIRT1-Mediated eNAMPT Secretion from Adipose Tissue Regulates
Hypothalamic NAD+ and Function in Mice. Cell Metab 21(5): 706-717.
54. Nakahata Y, Sahar S, Astarita G, Kaluzova M, Sassone-Corsi P (2009)
Circadian control of the NAD+ salvage pathway by CLOCK-SIRT1.
Science 324(5927): 654-657.
55. Wang LF, Wang XN, Huang CC, Hu L, Xiao YF, et al. (2017) Inhibition
of NAMPT aggravates high fat diet-induced hepatic steatosis in mice
through regulating Sirt1/AMPKalpha/SREBP1 signaling pathway.
Lipids Health Dis 16(1): 82.
56. Koltai E, Szabo Z, Atalay M, Boldogh I, Naito H, et al. (2010) Exercise
alters SIRT1, SIRT6, NAD and NAMPT levels in skeletal muscle of aged
rats. Mech Ageing Dev 131(1): 21-28.
57. He X, He J, Shi Y, Pi C, Yang Y, et al. (2017) Nicotinamide
phosphoribosyltransferase (Nampt) may serve as the marker for
osteoblast differentiation of bone marrow-derived mesenchymal stem
cells. Exp Cell Res 352(1): 45-52.
58. Ma C, Pi C, Yang Y, Lin L1, Shi Y, et al. (2017) Nampt Expression
Decreases Age-Related Senescence in Rat Bone Marrow Mesenchymal
Stem Cells by Targeting Sirt1. PLoS One 12(1): e0170930.
59. Li Y, He X, Li Y, He J, Anderstam B, et al. (2011) Nicotinamide
phosphoribosyltransferase (Nampt) affects the lineage fate
determination of mesenchymal stem cells: a possible cause for reduced
osteogenesis and increased adipogenesis in older individuals. J Bone
Miner Res 26(11): 2656-2664.
60. Yoshida M, Satoh A, Lin JB, Mills KF, Sasaki Y, et al. (2019) Extracellular
Vesicle-Contained eNAMPT Delays Aging and Extends Lifespan in
Mice. Cell Metab 30(2): 329-342.
61. Yoshino J, Mills KF, Yoon MJ, Imai S (2011) Nicotinamide mononucleotide,
a key NAD(+) intermediate, treats the pathophysiology of diet- and
age-induced diabetes in mice. Cell Metab 14(4): 528-536.
62. Magni G, Amici A, Emanuelli M, Orsomando G, Raffaelli N, et al. (2004)
Enzymology of NAD+ homeostasis in man. Cell Mol Life Sci 61(1): 19-
34.
63. Zhang T, Berrocal JG, Frizzell KM, Gamble MJ, DuMond ME, et al. (2009)
Enzymes in the NAD+ salvage pathway regulate SIRT1 activity at target
gene promoters. J Biol Chem 284(30): 20408-20417.
64. Conforti L, Janeckova L, Wagner D, Mazzola F, Cialabrini, et al. (2011)
Reducing expression of NAD+ synthesizing enzyme NMNAT1 does not
affect the rate of Wallerian degeneration. FEBS J 278(15): 2666-2679.
65. Gilley J, Adalbert R, Yu G, Coleman MP (2013) Rescue of peripheral and
CNS axon defects in mice lacking NMNAT2. J Neurosci 33(33): 13410-
13424.
66. Hicks AN, Lorenzetti D, Gilley J, Lu B, Andersson KE, et al. (2012)
Nicotinamide mononucleotide adenylyltransferase 2 (Nmnat2)
regulates axon integrity in the mouse embryo. PLoS One 7(10):
e47869.
67. Yue Z, Ma Y, You J, Li Z, Ding Y, et al. (2016) NMNAT3 is involved in the
protective effect of SIRT3 in Ang II-induced cardiac hypertrophy. Exp
Cell Res 347(2): 261-273.
68. VanLinden MR, Dolle C, Pettersen IK, Kulikova VA, Niere M, et al. (2015)
Subcellular Distribution of NAD+ between Cytosol and Mitochondria

27644-27659.
69. Herranz D, Munoz-Martin M, Canamero M, Francisca Mulero, Barbara
Martinez Pastor, et al. (2010) Sirt1 improves healthy ageing and
protects from metabolic syndrome-associated cancer. Nat Commun 1:
3.
70. Julio C Morales, Li L, Farjana J Fattah, Ying Dong, Erik A Bey, et al.
(2014) Review of Poly (ADP-ribose) Polymerase (PARP) Mechanisms
of Action and Rationale for Targeting in Cancer and Other Diseases.
Crit Rev Eukaryot Gene Expr 24(1) :15-28.
71. 
a pathway related to calorie restriction. Proc Natl Acad Sci U S A
101(45): 15998-16003.
72. Matt Kaeberlein MM, Leonard Guarente (1999) The SIR2/3/4 complex
and SIR2 alone promote longevity in Saccharomyces cerevisiae by two
different mechanisms. Genes Dev 13(19): 2570-2580.
73. Tang BL (2016) Sirt1 and the Mitochondria. Mol Cells 39(2): 87-95.
74. Alcendor RR, Gao S, Zhai P, Zablocki D, Holle E, et al. (2007) Sirt1
regulates aging and resistance to oxidative stress in the heart. Circ Res
100(10): 1512-1521.
75. Sultani G, Samsudeen AF, Osborne B, Turner N (2017) NAD (+):
A key metabolic regulator with great therapeutic potential. J
Neuroendocrinol 29(10).
76. Nakamura AJ, Chiang YJ, Hathcock KS, Horikawa I, Sedelnikova
OA, et al. (2008) Both telomeric and non-telomeric DNA damage
are determinants of mammalian cellular senescence. Epigenetics
Chromatin 1(1): 6.
77. Hou WH, Chen SH, Yu X (2019) Poly-ADP ribosylation in DNA damage
response and cancer therapy. Mutat Res 780: 82-91.
78. Sas K, Szabo E, Vecsei L (2018) Mitochondria, Oxidative Stress and
the Kynurenine System, with a Focus on Ageing and Neuroprotection.
Molecules 23(1).
79. Pillai JB, Isbatan A, Imai S, Gupta MP (2005) Poly (ADP-ribose)
polymerase-1-dependent cardiac myocyte cell death during heart
failure is mediated by NAD+ depletion and reduced Sir2alpha
deacetylase activity. J Biol Chem 280(52): 43121-43130.
80. Aksoy P, Escande C, White TA, Thompson M, Soares S, et al. (2006)
Regulation of SIRT 1 mediated NAD dependent deacetylation: a novel
role for the multifunctional enzyme CD38. Biochem Biophys Res
Commun 349(1): 353-359.
81. Aksoy P, White TA, Thompson M, Chini EN, Benech JC, et al. (2006)
Regulation of intracellular levels of NAD: a novel role for CD38.
Biochem Biophys Res Commun 345(1): 1386-1392.
82. Chini C, Hogan KA, Warner GM, Tarragó MG, Peclat TR, et al. (2019)
The NADase CD38 is induced by factors secreted from senescent cells
providing a potential link between senescence and age-related cellular
NAD(+) decline. Biochem Biophys Res Commun 513(2): 486-493.
83. Dong M, Si YQ, Sun SY, Pu XP, Yang ZJ, et al. (2011) Design, synthesis
and biological characterization of novel inhibitors of CD38. Org Biomol
Chem 9(9): 3246-3257.
84. Biran A, Zada L, Abou Karam P, Vadai E, Roitman L, et al. (2017)
        
Aging Cell 16(4): 661-671.
85. Wiley CD, Velarde MC, Lecot P, Liu S, Sarnoski EA, et al. (2016)
Mitochondrial Dysfunction Induces Senescence with a Distinct
Secretory Phenotype. Cell Metab 23(2): 303-314.
86. Rodier F, Coppé JP, Patil CK, Hoeijmakers WA, Muñoz DP, et al. (2009)
Persistent DNA damage signalling triggers senescence-associated

87. Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, et al. (2018)
Senolytics improve physical function and increase lifespan in old age.
Nat Med 24: 1246-1256.
Am J Biomed Sci & Res Copy@ Taosheng Huang
American Journal of Biomedical Science & Research
452
88. Belenky P, Bogan KL, Brenner C (2007) NAD+ metabolism in health
and disease. Trends Biochem Sci 32: 12-19.
89. Hyung-Jin Kim, Wa Cao, Gi-Su Oh, Seung Hoon Lee, AiHua Shen, et al.
(2018) Augmentation of cellular NAD(+) by NQO1 enzymatic action
improves age-related hearing impairment. Aging Cell 18(5): e13016.
90. Sharpless NE, DePinho RA (2007) How stem cells age and why this
makes us grow old. Nat Rev Mol Cell Biol 8: 703-713.
91. Lopez-Otin C, Blasco MA, Partridge L, Manuel Serrano, Guido Kroemer,
et al. (2013) The hallmarks of aging. Cell 153: 1194-1217.
92. Wagers AJ, Weissman IL (2004) Plasticity of adult stem cells. Cell 116:
639-648.
93. Hou Y, Lautrup S, Cordonnier S, Wang Y, Croteau DL, et al. (2018)
NAD supplementation normalizes key Alzheimer’s features and DNA
damage responses in a new AD mouse model with introduced DNA

94. Cantó C, Houtkooper RH, Pirinen E, Youn DY, Oosterveer MH, et
al. (2012) The NAD(+) precursor nicotinamide riboside enhances
oxidative metabolism and protects against high-fat diet-induced
obesity. Cell Metab 15: 838-847.
95. Gariani K, Menzies KJ, Ryu D, Wegner CJ, Wang X, et al. (2016) Eliciting
the mitochondrial unfolded protein response by nicotinamide adenine
dinucleotide repletion reverses fatty liver disease in mice. Hepatology
63: 1190-1204.
96. Fang EF, Kassahun H, Croteau DL, Scheibye-Knudsen M, Marosi K, et
al. (2016) NAD(+) Replenishment Improves Lifespan and Healthspan
in Ataxia Telangiectasia Models via Mitophagy and DNA Repair. Cell
Metab 24: 566-581.
97.         
NAD(+)/SIRT1 Axis. Cell Metab 24: 526-528.
98. Csiszar A, Tarantini S, Yabluchanskiy A, Balasubramanian P, Kiss T, et

dysfunction. 316: H1253-H1266.
99. de Picciotto NE, Gano LB, Johnson LC, Martens CR, Sindler AL, et al.
(2016) Nicotinamide mononucleotide supplementation reverses
vascular dysfunction and oxidative stress with aging in mice. Aging
Cell 15: 522-530.
100. Koopman WJ, Willems PH, Smeitink JA (2012) Monogenic
mitochondrial disorders. N Engl J Med 366: 1132-1141.
101. Long AN, Owens K, Schlappal AE, Kristian T, Fishman PS, et al. (2015)
Effect of nicotinamide mononucleotide on brain mitochondrial
  
BMC Neurol 15:19.
102. Wang X, Hu X, Yang Y, Takata T, Sakurai T, et al. (2016) Nicotinamide
mononucleotide protects against beta-amyloid oligomer-induced
cognitive impairment and neuronal death. Brain Res 1643: 1-9.
103. Mitchell SJ, Bernier M, Aon MA, Cortassa S, Kim EY, et al. (2018)
Nicotinamide Improves Aspects of Healthspan, but Not Lifespan, in
Mice. 27: 667-676.e664.
104. Moon J, Kim HR, Shin MG (2018) Rejuvenating Aged Hematopoietic
Stem Cells Through Improvement of Mitochondrial Function. Ann Lab
Med 38: 395-401.
105. Jie Song, Jing Li, Fangji Yang, Gang Ning, Limin Zhen, et al. (2019)
Nicotinamide mononucleotide promotes osteogenesis and reduces
adipogenesis by regulating mesenchymal stromal cells via the SIRT1
pathway in aged bone marrow. Cell Death Dis 10: 336.
106. TV A, Cell KBJ (2018) HS to Mitigate Vascular Aging: A SIRT1
Connection. 173: 8-10.
107. Gerdts J, Brace EJ, Sasaki Y, DiAntonio A, Milbrandt J, et al. (2015)
       
destruction. 348: 453-457.
108. Zhang S, Xue X, Zhang L, Zhang L, Liu Z, et al. (2015) Comparative
Analysis of Pharmacophore Features and Quantitative Structure-
Activity Relationships for CD38 Covalent and Non-covalent Inhibitors.
Chem Biol Drug Des 86: 1411-1424.
109. Kellenberger E, Kuhn I, Schuber F, Muller-Steffner H (2011) Flavonoids
as inhibitors of human CD38. Bioorg Med Chem Lett 21: 3939-3942.
110. KA Wall, M Klis, J Kornet, D Coyle, JC Amé, et al. (1998) Inhibition of
the intrinsic NAD+ glycohydrolase activity of CD38 by carbocyclic NAD
analogues. Biochem J 335(Pt 3): 631-636.
111. Mukhopadhyay P, Rajesh M, Cao Z, Horváth B, Park O, et al. (2014) Poly
        

112. Fang EF, Scheibye-Knudsen M, Brace LE, Kassahun H, SenGupta T, et
al. (2014) Defective mitophagy in XPA via PARP-1 hyperactivation and
NAD(+)/SIRT1 reduction. Cell 157: 882-896.
113. Day K, Waite LL, Thalacker-Mercer A, West A, Bamman MM, et al.
(2013) Differential DNA methylation with age displays both common

landscape. Genome Biol 14: R102.
114. Jones PA (2012) Functions of DNA methylation: islands, start sites,
gene bodies and beyond. Nat Rev Genet 13: 484-492.
115. Maegawa S, Hinkal G, Kim HS, Shen L, Zhang L, et al. (2010) Widespread
        
Genome Res 20: 332-340.
116. Smith ZD, Meissner A (2013) DNA methylation: roles in mammalian
development. Nat Rev Genet 14: 204-220.
117. Schultz MD, He Y, Whitaker JW, Hariharan M, Mukamel EA, et al. (2015)
Human body epigenome maps reveal noncanonical DNA methylation
variation. Nature 523: 212-216.
118. Schultz MD, He Y, Whitaker JW, Hariharan M, Mukamel EA, et al. (2016)
Corrigendum: Human body epigenome maps reveal noncanonical DNA
methylation variation. Nature 530: 242.
119. Levine ME, Lu AT, Bennett DA, Horvath S (2015) Epigenetic age of the
pre-frontal cortex is associated with neuritic plaques, amyloid load,
and Alzheimer’s disease related cognitive functioning. Aging (Albany
NY) 7: 1198-1211.
120. Hannum G, Guinney J, Zhao L, Zhang L, Hughes G, et al. (2013) Genome-
       
rates. Mol Cell 49: 359-367.
121. Bock landt S, Lin W, Sehl ME, Sehl ME, Sánchez FJ, Sinsheimer JS et al.
(2011) Epigenetic predictor of age. PLoS One 6(6): e14821.
122. Koch CM, Wagner W (2011) Epigenetic-aging-signature to determine
age in different tissues. Aging (Albany NY) 3(10): 1018-1027.
123. Weidner CI, Lin Q, Koch CM, Eisele L, Beier F, et al. (2014) Aging of
blood can be tracked by DNA methylation changes at just three CpG
sites. Genome Biol 15(2): R24.
124. Petkovich DA, Podolskiy DI, Lobanov AV, Lee SG, Miller RA, et al.

Longevity Interventions. Cell Metab 25(4): 954-960.
125. Stubbs TM, Bonder MJ, Stark AK, Krueger F, et al. (2017) Multi-tissue
DNA methylation age predictor in mouse. Genome Biol 18(1): 68.
126. Han Y, Eipel M, Franzen J, Sakk V, Dethmers-Ausema B, et al. (2018)
Epigenetic age-predictor for mice based on three CpG sites. Elife 24: 7.
127. Thompson MJ, Chwialkowska K, Rubbi L, Lusis AJ, Davis RC, et al.
(2018) A multi-tissue full lifespan epigenetic clock for mice. Aging
(Albany NY) 10(10): 2832-2854.
128. Fahy GM, Brooke RT, Watson JP, Michael S Kobor, Steve Horvath, et al.
(2019) Reversal of epigenetic aging and immunosenescent trends in
humans. Aging Cell 18(6): e13028.
American Journal of Biomedical Science & Research
Am J Biomed Sci & Res Copy@ Taosheng Huang
453
129. Abbott A (2019) First hint that body’s ‘biological age’ can be reversed.
Nature 573(7773): 173.
130. Trammell SA, Schmidt MS, Weidemann BJ, Redpath P, Jaksch F, et al.
(2016) Nicotinamide riboside is uniquely and orally bioavailable in
mice and humans. Nat Commun 10(7): 12948.
131. Martens CR, Denman BA, Mazzo MR, Armstrong ML, Reisdorph N, et
al. (2018) Chronic nicotinamide riboside supplementation is well-
tolerated and elevates NAD+ in healthy middle-aged and older adults.
Nat Commun 9(1): 1286.
132. Elhassan YS, Kluckova K, Fletcher RS, Schmidt MS, Garten A, et al.
(2019) Nicotinamide Riboside Augments the Aged Human Skeletal
Muscle NAD Metabolome and Induces Transcriptomic and Anti-

133. Airhart SE, Shireman LM, Risler LJ, Anderson GD, Nagana Gowda
GA, et al. (2017) An open-label, non-randomized study of the
pharmacokinetics of the nutritional supplement nicotinamide riboside
(NR) and its effects on blood NAD+ levels in healthy volunteers. PLoS
One 12(12): e0186459.
134. Dolopikou CF, Kourtzidis IA, Margaritelis NV, Vrabas IS, Koidou I et al.
(2019) Acute nicotinamide riboside supplementation improves redox
homeostasis and exercise performance in old individuals: a double-
blind cross-over study. Eur J Nutr 6.
135. 
Japan. NPJ Aging Mech Dis 2: 16021.
136. Nacarelli T, Lau L, Fukumoto T, Zundell J, Fatkhutdinov N, et al.
(2019) NAD+    
associated secretome. Nat Cell Biol 21(3): 397-407.
... There are various pathways of NAD + synthesis, primarily taken from various dietary sources, such as de novo synthesis of tryptophan via kynurenine pathway; from nicotinic acid by Preiss-Handler pathway; and from nicotinamide, nicotinamide ribose through the Salvage pathway. Some researchers stated that NAD + precursor supplementation in lab mice showed a delayed aging process [18,19]. At the same time, studies have found that the Salvage pathway is defective during the senility period, mostly involving the skeletal muscles, white adipose tissues, and other organs [19]. ...
... Studies have been carried out in humans, which have revealed that supplementation with NAD + precursors by restoring its balance, might contribute to an aging delay. However, its overdose might trigger harmful effects and excessive secretion of pro-inflammatory cytokines [18], as well as predispose the body to development of autoimmune diseases and tumors. NAD + is correspondingly involved in neuroprotection, whereas a defective metabolism can lead to the onset of neurodegenerative diseases [19]. ...
Article
Full-text available
Background. Researchers tried to find out how age-related pathologies can be prevented by keeping the biomarkers levels under control since their changes might induce abnormalities. The purpose was to analyze aspects that might accelerate, and fight off the action of endogenous and exogenous factors on the metabolism of aging. Material and methods. This study is based on the critical analysis of 35 literature sources out of 74 selected from PubMed, NCBI, Google Scholar data that highlight the most essential concepts. Results. Biological age represents the modified aspects of metabolism and physiological processes. The antioxidant system and reactive oxygen species play a key role in slowing down, and accelerating the aging metabolism; genetic markers detect the aging factors; genome mutations are responsible for premature aging. Conclusions. Hence, it's important to make a balance between a healthy lifestyle, a proper skincare routine, and maintaining cognitive functions, in order to hope for a "successful aging" by preventing diseases and increasing life expectancy.
... Ageing-associated increase in NFκ-B could be illuminated by NAD+ decline accompanying the senescence process through two mechanisms: First, marked elevation of ROS during ageing [26] . Second, the reduction in sirtuin deacetylases like SIRT1, 2 & 6 [NAD+-dependant enzymes responsible for protein deacetylation, proper mitochondrial functions, effective stem cells, DNA repair and efficient metabolic pathways [27,28] secondary to NAD+ decline [3] . ...
Chapter
Extracellular vesicles (EVs) are membrane-encapsulated vesicles carrying various signal molecules, including metabolites, proteins, enzymes, genetic material, nucleic acids, mRNAs, noncoding RNAs, intact RNAs, lipids, organelles, ions, pathogenic microorganisms, and disease-specific molecules. The International Society for Extracellular Vesicles (ISEV) describes EVs as lipid-enveloped vesicles released from cells that do not contain a nucleus and cannot replicate themselves. EVs are categorized based on size and biogenesis as apoptotic bodies, exosomes, macrovesicles, and others. EVs are released from prokaryotic and eukaryotic cells, such as epithelial cells, mesenchymal stem cells, neurons, and endothelial cells, to regulate intracellular regulation and communication through blood circulation. Therefore, EVs are potential and promising biomarkers, drug delivery tools, and therapeutic approaches. These vesicles transfer knowledge of the parent cell’s biochemical and physiological status to the neighbor cells. The information of the neighbor cell from the current healthy or pathological conditions can be used not only for intercellular conditions but also as a biomarker for many biological or pathological processes. Crosstalk between the cells may have many crucial functions in health, aging-related disease, and cancer cell signaling. Delayed diagnosis and treatment lead to poor prognosis and increased risk for mortality in cancer patients; thus, novel cancer biomarkers are urgently required for early prognosis, disease progression monitoring, and personalized therapies. Since cell-cell interactions between tumor cells and cancer cell communication with stromal and immune cells occur via EVs, they are considered promising biomarkers for the early detection of cancer since they are secreted from cells circulating in the blood and are easy to collect. EVs have superior properties to overcome chemotherapy-related limitations because they have extended circulation time, excellent tumor-targeting capabilities, lower immunogenicity, higher biocompatibility, and reduced systemic toxicity in the peripheral tissues. In this chapter, we discussed the biogenesis, role, functions, and challenges of various types of EVs in aging-related diseases and cancer treatment.
Article
Full-text available
Nicotinamide adenine dinucleotide (NAD+) is modulated by conditions of metabolic stress and has been reported to decline with aging in preclinical models, but human data are sparse. Nicotinamide riboside (NR) supplementation ameliorates metabolic dysfunction in rodents. We aimed to establish whether oral NR supplementation in aged participants can increase the skeletal muscle NAD+ metabolome and if it can alter muscle mitochondrial bioenergetics. We supplemented 12 aged men with 1 g NR per day for 21 days in a placebo-controlled, randomized, double-blind, crossover trial. Targeted metabolomics showed that NR elevated the muscle NAD+ metabolome, evident by increased nicotinic acid adenine dinucleotide and nicotinamide clearance products. Muscle RNA sequencing revealed NR-mediated downregulation of energy metabolism and mitochondria pathways, without altering mitochondrial bioenergetics. NR also depressed levels of circulating inflammatory cytokines. Our data establish that oral NR is available to aged human muscle and identify anti-inflammatory effects of NR.
Article
Full-text available
Age-related hearing loss (ARHL) is a major neurodegenerative disorder and the leading cause of communication deficit in the elderly population, which remains largely untreated. The development of ARHL is a multifactorial event that includes both intrinsic and extrinsic factors. Recent studies suggest that NAD+ /NADH ratio may play a critical role in cellular senescence by regulating sirtuins, PARP-1, and PGC-1α. Nonetheless, the beneficial effect of direct modulation of cellular NAD+ levels on aging and age-related diseases has not been studied, and the underlying mechanisms remain obscure. Herein, we investigated the effect of β-lapachone (β-lap), a known plant-derived metabolite that modulates cellular NAD+ by conversion of NADH to NAD+ via the enzymatic action of NADH: quinone oxidoreductase 1 (NQO1) on ARHL in C57BL/6 mice. We elucidated that the reduction of cellular NAD+ during the aging process was an important contributor for ARHL; it facilitated oxidative stress and pro-inflammatory responses in the cochlear tissue through regulating sirtuins that alter various signaling pathways, such as NF-κB, p53, and IDH2. However, augmentation of NAD+ by β-lap effectively prevented ARHL and accompanying deleterious effects through reducing inflammation and oxidative stress, sustaining mitochondrial function, and promoting mitochondrial biogenesis in rodents. These results suggest that direct regulation of cellular NAD+ levels by pharmacological agents may be a tangible therapeutic option for treating various age-related diseases, including ARHL.
Article
Full-text available
Mesenchymal stromal cells (MSCs) can differentiate to various cell types including osteoblasts, chondrocytes, and adipocytes. This cellular flexibility contributes to widespread clinical use of MSCs in tissue repair. However, challenges remain in efficient cellular expansion of MSCs for stem cell therapy. Current MSC culture methods have resulted in reduced self-renewal of MSCs and compromised therapeutic outcomes. This study identifies that nicotinamide mononucleotide (NMN), a key natural NAD+ intermediate, effectively encourages MSC expansion in vitro and in vivo. The in vitro expanded MSCs had heightened osteogenesis, but reduced adipogenesis. Furthermore, NMN supplementation stimulated osteogenesis of endogenous MSCs, and protected bone from aging and irradiation induced damage in mice. Mechanistically, we found that NMN treatment upregulated SIRT1. Genetically overexpressing SIRT1 in MSCs by using Prx1 cre; ColA1flox-stop-flox-SIRT1 mice promoted osteogenesis and reduced adipogenesis in aged mice. Overall, our data demonstrate that NMN promoted MSC self-renewal with strengthened osteogenesis and reduced adipogenesis via upregulating SIRT1 in aged mice.
Article
Full-text available
Tissue nicotinamide adenine dinucleotide (NAD ⁺ ) decline has been implicated in aging. We have recently identified CD38 as a central regulator involved in tissue NAD ⁺ decline during the aging process. CD38 is an ecto-enzyme highly expressed in endothelial and inflammatory cells. To date, the mechanisms that regulate CD38 expression in aging tissues characterized by the presence of senescent cells is not completely understood. Cellular senescence has been described as a hallmark of the aging process and these cells are known to secrete several factors including cytokines and chemokines through their senescent associated secretory phenotype (SASP). Here we investigated if the cellular senescence phenotype is involved in the regulation of CD38 expression and its NADase activity. We observed that senescent cells do not have high expression of CD38. However, the SASP factors secreted by senescent cells induced CD38 mRNA and protein expression and increased CD38-NADase activity in non-senescent cells such as endothelial cells or bone marrow derived macrophages. Our data suggest a link between cellular senescence and NAD ⁺ decline in which SASP-mediated upregulation of CD38 can disrupt cellular NAD ⁺ homeostasis.
Article
Full-text available
Cellular senescence is a stable growth arrest that is implicated in tissue ageing and cancer. Senescent cells are characterized by an upregulation of proinflammatory cytokines, which is termed the senescence-associated secretory phenotype (SASP). NAD+ metabolism influences both tissue ageing and cancer. However, the role of NAD+ metabolism in regulating the SASP is poorly understood. Here, we show that nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme of the NAD+ salvage pathway, governs the proinflammatory SASP independent of senescence-associated growth arrest. NAMPT expression is regulated by high mobility group A (HMGA) proteins during senescence. The HMGA–NAMPT–NAD+ signalling axis promotes the proinflammatory SASP by enhancing glycolysis and mitochondrial respiration. HMGA proteins and NAMPT promote the proinflammatory SASP through NAD+-mediated suppression of AMPK kinase, which suppresses the p53-mediated inhibition of p38 MAPK to enhance NF-κB activity. We conclude that NAD+ metabolism governs the proinflammatory SASP. Given the tumour-promoting effects of the proinflammatory SASP, our results suggest that anti-ageing dietary NAD+ augmentation should be administered with precision. Nacarelli et al. show that the nicotinamide-phosphoribosyltransferase-regulated NAD+ biogenesis pathway promotes the proinflammatory senescence-associated secretory phenotype by enhancing glycolysis and mitochondrial respiration during senescence.
Article
Full-text available
Purpose Older individuals suffer from low NADH levels. We have previously shown that nicotinamide riboside [NR; a NAD(P)(H) precursor] administration impaired exercise performance in young rats. It has been suggested that supplementation of redox agents exerts ergogenic effect only in deficient individuals. We hypothesized that old individuals would more likely benefit from NR supplementation. We investigated the effect of acute NR supplementation on redox homeostasis and physical performance in young and old individuals. Methods Twelve young and twelve old men received NR or placebo in a double-blind cross-over design. Before and 2 h after NR or placebo supplementation, blood and urine samples were collected, while physical performance (VO2max, muscle strength, and fatigue) was assessed after the second blood sample collection. Results At rest, old individuals exhibited lower erythrocyte NAD(P)H levels, higher urine F2-isoprostanes and lower erythrocyte glutathione levels compared to young (P < 0.05). NR supplementation increased NADH (51% young; 59% old) and NADPH (32% young; 38% old) levels in both groups (P < 0.05), decreased F2-isoprostanes by 18% (P < 0.05), and tended to increase glutathione (P = 0.078) only in the old. NR supplementation did not affect VO2max and concentric peak torque, but improved isometric peak torque by 8% (P = 0.048) and the fatigue index by 15% (P = 0.012) in the old. In contrast, NR supplementation did not exert any redox or physiological effect in the young. Conclusions NR supplementation increased NAD(P)H levels, decreased oxidative stress, and improved physical performance only in old subjects, substantiating that redox supplementation may be beneficial only in individuals with antioxidant deficiencies.
Article
Full-text available
Human DNA-methylation data have been used to develop highly accurate biomarkers of aging ("epigenetic clocks"). Recent studies demonstrate that similar epigenetic clocks for mice (Mus Musculus) can be slowed by gold standard anti-aging interventions such as calorie restriction and growth hormone receptor knock-outs. Using DNA methylation data from previous publications with data collected in house for a total 1189 samples spanning 193,651 CpG sites, we developed 4 novel epigenetic clocks by choosing different regression models (elastic net- versus ridge regression) and by considering different sets of CpGs (all CpGs vs highly conserved CpGs). We demonstrate that accurate age estimators can be built on the basis of highly conserved CpGs. However, the most accurate clock results from applying elastic net regression to all CpGs. While the anti-aging effect of calorie restriction could be detected with all types of epigenetic clocks, only ridge regression based clocks replicated the finding of slow epigenetic aging effects in dwarf mice. Overall, this study demonstrates that there are trade-offs when it comes to epigenetic clocks in mice. Highly accurate clocks might not be optimal for detecting the beneficial effects of anti-aging interventions.
Article
Full-text available
ELife digest Epigenetic marks are chemical modifications found throughout the genome – the DNA within cells. By influencing the activity of nearby genes, the marks govern developmental processes and help cells to adapt to changes in their surroundings. Some epigenetic marks can be gained or lost with age. A lot of aging research focuses on one type of mark, called “DNA methylation”. By measuring the presence or absence of specific methyl groups, scientists can estimate biological age – which may differ from calendar age. Recent studies have developed computer models called epigenetic aging clocks to predict the biological age of mouse cells. These clocks use epigenetic data collected from the entire genomes of mice, and are useful for understanding how the aging process is affected by genetic parameters, diet, or other environmental factors. Yet, the genome sequencing methods used to construct most existing epigenetic clocks are expensive, labor-intensive, and cannot be easily applied to large groups of mice. Han et al. have developed a new way to predict biological aging in mice that needs methylation information from just three particular sections of the genome. Even though this approach is much faster and less expensive than other epigenetic approaches to measuring aging, it has a similar level of accuracy to existing models. Han et al. use the new method to show that cells from different strains of laboratory mice age at different rates. Furthermore, in a strain that has a shorter life expectancy, aging seems to be accelerated. The new approach developed by Han et al. will make it easier to study how aging in mice is affected by different interventions. Further studies will also be needed to better understand how epigenetic marks relate to biological aging.
Article
In a small trial, drugs seemed to rejuvenate the body’s ‘epigenetic clock’, which tracks a person’s biological age. In a small trial, a cocktail of drugs seemed to rejuvenate the body’s ‘epigenetic clock’. Man participating in the 5k race during the Rock n Roll Virginia Beach Half Marathon, Virginia
Article
Aging is a significant risk factor for impaired tissue functions and chronic diseases. Age-associated decline in systemic NAD+ availability plays a critical role in regulating the aging process across many species. Here, we show that the circulating levels of extracellular nicotinamide phosphoribosyltransferase (eNAMPT) significantly decline with age in mice and humans. Increasing circulating eNAMPT levels in aged mice by adipose-tissue-specific overexpression of NAMPT increases NAD+ levels in multiple tissues, thereby enhancing their functions and extending healthspan in female mice. Interestingly, eNAMPT is carried in extracellular vesicles (EVs) through systemic circulation in mice and humans. EV-contained eNAMPT is internalized into cells and enhances NAD+ biosynthesis. Supplementing eNAMPT-containing EVs isolated from young mice significantly improves wheel-running activity and extends lifespan in aged mice. Our findings have revealed a novel EV-mediated delivery mechanism for eNAMPT, which promotes systemic NAD+ biosynthesis and counteracts aging, suggesting a potential avenue for anti-aging intervention in humans.