Skip to main content
Oxidative Medicine and Cellular Longevity logoLink to Oxidative Medicine and Cellular Longevity
. 2023 Feb 8;2023:4985726. doi: 10.1155/2023/4985726

NAD+-Consuming Enzymes in Stem Cell Homeostasis

Xiuna Ji 1, Mingyue Zheng 2, Tao Yu 3, Jie Kang 1, Tingjun Fan 1, Bin Xu 1,
PMCID: PMC9931471  PMID: 36819783

Abstract

Nicotinamide adenine dinucleotide (NAD+) is a coenzyme used in redox reactions, energy metabolism, and mitochondrial biogenesis. NAD+ is also required as a cofactor by nonredox NAD+-dependent enzymes. Hundreds of enzymes that consume NAD+ have been identified. The NAD+-consuming enzymes are involved in a variety of cellular processes such as signal transduction, DNA repair, cellular senescence, and stem cell (SC) homeostasis. In this review, we discussed how different types of NAD+-consuming enzymes regulate SC functions and summarized current research on the roles of the NAD+ consumers in SC homeostasis. We hope to provide a more global and integrative insight to the mechanism and intervention of SC homeostasis via the regulation of the NAD+-consuming enzymes.

1. Introduction

Nicotinamide adenine dinucleotide (NAD+) has been described as central coenzyme for redox reactions [1]. During the oxidation of glucose and fatty acids, NAD+ is reduced to NADH, which acts as a ubiquitous cellular electron donor. Two NADH are generated by glycolysis and converted back to NAD+ [2]. In addition to energy metabolism, NAD+ is used as a cofactor or cosubstrate by hundreds of enzymes [3]. This net consumption of NAD+ is compensated by de novo starting with tryptophan, or from salvage pathways starting with NAD+ precursors, thus maintaining a balanced pool under normal physiological conditions [2].

The diverse enzymes consuming NAD+ are found in almost all eukaryotic cells (Figure 1). They have multiple roles in the regulation of cellular processes and functions such as DNA repair, epigenetic modification, and inflammation [4]. These enzymes can be divided into two categories involved in a variety of cellular processes, including stress response, mitochondrial homeostasis, and calcium signaling [5]. The first category breaks down NAD+ and transfers the adenosine diphosphate- (ADP-) ribose units to fundamental biomolecules. The second hydrolyzes NAD+ to ADP-ribose (ADPR), cyclic ADPR (cADPR), and nicotinamide (NAM), which are dominated by multifunctional ectoenzymes with both glycohydrolase and ADP-ribosyl cyclase activities (Figure 2) [6, 7]. Recent reports show that the above NAD+ consumers attracted more attentions in the field of stem cell (SC) homeostasis mechanism and intervention [8]. Therefore, in this review, we focus on the current advances in the roles of NAD+-consuming enzymes in SC homeostasis.

Figure 1.

Figure 1

Locations of NAD+-consuming enzymes. SIRT1 is reported to locate in both in nucleus and cytosol. SIRT2, predominantly resided in cytosol, may also exist in mitochondria and nucleus. SIRT3 is a major mitochondrial deacetylase and also plays a role in nucleus. SIRT4 is only found in mitochondria. SIRT5 locates in mitochondrial matrix and intermembrane space, cytosol, peroxisome, and nucleus. SIRT6 is only found in nucleus. SIRT7 is the only sirtuin protein that mainly locates in nucleolus. PARPs reside in cytosol, mitochondria, and nucleus. CD38 is observed both in plasma membrane (catalytic domain facing outside or inside) and intracellular membranes (including endoplasmic reticulum, nucleus, mitochondria, and endolysosome). CD157 is a glycophosphatidylinositol-anchored protein with the catalytic domain facing outside. Sterile alpha and Toll/interleukin receptor (TIR) motif-containing 1 (SARM1) locates in cytosol, mitochondria, and nucleus. Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) is found in mitochondria and nucleus. NAD+ kinase (NADK) only resides in cytosol.

Figure 2.

Figure 2

Catalytic reactions of NAD+-consuming enzymes using NAD+ as a substrate. Sirtuins remove acyl groups from lysine residues on target substrates including proteins and lipids. ADP-ribose (ADPR) cleaved from NAD+ serves as an acyl group acceptor to generate acetyl-ADPR. PARPs transfer the ADP-ribose from NAD+ to proteins, DNA, and RNA (known as PARylation). CD38, CD157, and sterile alpha and Toll/interleukin receptor (TIR) motif-containing 1 (SARM1) are multifunctional ectoenzymes with both glycohydrolase and ADP-ribosyl cyclase activities. Their main catalytic activity is the hydrolysis of NAD+ to NAM and ADP-ribose (ADPR). They also catalyze NAD+ to NAM and cADPR. Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) uses NAD+ to generate NADH. NAD+ can also be consumed by NAD+ kinase (NADK) to increase NADP+ production. Then NADP+ is catalyzed to NAADP by CD38.

The literature search produced a total of 49,519 records (39,570 from Web of Science and 9,949 from PubMed). After title/abstract/language/literature evaluation screening, 99 articles were assessed for full-text eligibility, an additional 22 studies identified through review articles, and a final total of 121 articles were included in the final review (Figure S1).

2. Category I: Deacylases (Sirtuins (SIRTs) and Poly(ADP-Ribose) Polymerases (PARPs))

2.1. Sirtuins

There are seven sirtuins (SIRT1–7) with a conserved NAD+-binding domain in mammals [9, 10]. Sirtuins have various activities, containing different lysine deacetylation reactions, ADP ribosylation, and removal of lipid modifications [11, 12]. They have been found to play roles in multiple cellular functions, such as genomic stability, transcription, signal transduction, and metabolism [13]. Recent studies report that they also directly or indirectly participate in the regulation of multiple signaling pathways to maintain SC homeostasis (Table 1).

Table 1.

Sirtuin functions for stem cell homeostasis.

Sirtuins Stem cells Targets Functions References
SIRT1 MSC PGC-1α, TFAM, PARP1, FOXO1, FOXO3 Promoting mitochondrial fitness, DNA repair, and other aging-associated pathways [17]
MuSC MYLK2, MYOG Maintaining stemness and homeostasis [8]
UPRmt Maintaining mitochondrial homeostasis and self-renewal [14]
NSC FOXO3 Control over the circadian clock, limit exhaustion of their population [19]
Mouse ESC MAT2a, SMPDL3B Promoting pluripotency and embryogenesis [15, 16]
Rat MSC NAMPT Postponing senescence [18]
Liver CSC β-Catenin/NANOG, SOX2 Maintaining self-renewal [20, 21]

SIRT2 MuSC PAX7 Promoting function and differentiation [24]
HSC NLRP3 Preventing mitochondrial stress-induced cell death [25]
ALDOA, GAPDH, PGK1, ENO1 Regulating the metabolic transition [26]

SIRT3 HSC Antioxidant enzymes Maintaining homeostasis [29]
HSC Antiaging genes Delaying senescence [30]
NSC Apoptosis-related proteins Ameliorating microglia activation-induced oxidative stress injury [37]
MSC Antioxidant enzymes Counteracting senescence, restoring their differentiation capacity, and reduces oxidative stress [31, 32]
MSC laminB1, KAP1 and HP1α, etc. Counteracting senescence [34]
SSC Aerobic respiration-related factors Promoting differentiation [38, 39]
Mouse MSC Antioxidant enzymes Counteracting senescence [33]
Rat MSC PGC-1α/SIRT3/HIF-1α Regulating mitochondrial quality and function [35]
Mouse ESC TAC-related enzymes Promoting β cell maturation [36]

SIRT4 TSC LSD1 Positive effect on senescence [41]
Breast CSC SIRT1, H4K16ac, BRCA1 Negative effect on self-renewal [42]

SIRT5 ADMSC TCA-related enzymes Accelerating senescence [48]
MEF IDH2, G6PD Enhancing cellular antioxidant defense [50]

SIRT6 iPSC Pluripotent genes Differentiating into EBs and cardiomyocytes [54]
MSC NRF2 Maintaining homeostasis [58]
HSC Wnt target genes Maintaining self-renewal [53]

SIRT7 HSC, MSC NRF1 Regulating cellular energy metabolism, proliferation, and regenerative capacity [63, 64]
HFSC NFATc1 Initiating cell cycle [65]
MSC β-Catenin, AXIN Inhibiting osteogenic differentiation [66]

ADMSC: adipose-derived mesenchymal stem cell; ALDOA: aldolase A; BRCA1: breast cancer susceptibility gene 1; CSC: cancer stem cells; EBs: embryoid bodies; ENO1: enolase 1; ESC: embryonic stem cell; FOXO1: forkhead box O1; FOXO3: forkhead box O3; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; G6PD: glucose-6-phosphate dehydrogenase; HFSC: hair follicle stem cell; HIF-1α: hypoxia-inducible factor-1alpha; HP1α: heterochromatin protein 1alpha; HSC: hematopoietic stem cell; H4K16ac: acetylation of lysine 16 on histone H4; IDH2: isocitrate dehydrogenase-2; iPSC: induced pluripotent stem cell; KAP1: KRAB domain-associated protein 1; LSD1: lysine-specific demethylase 1; MAT2a: methionine adenosyltransferase 2a; MEFs: murine embryonic fibroblast; MSC: mesenchymal stem cell; MuSC: muscle stem cell; MYLK2: myosin light chain kinase 2; MYOG: myogenin; NAMPT: nicotinamide phosphoribosyltransferase; NFATc1: nuclear factor of activated T cells c1; NLRP3: nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3; NRF1: nuclear factor erythroid 2-related factor 1; NRF2: nuclear factor erythroid 2-related factor 2; NSC: neural stem cell; PARP1: poly(ADP-ribose) polymerase 1; PAX7: paired box 7; PGC-1α: peroxisome proliferator-activated receptor-gamma coactivator-1alpha; PGK1: phosphoglycerate kinase 1; SIRTs: sirtuins; SMPDL3B: sphingomyelin phosphodiesterase acid-like 3B; SSC: spermatogonial stem cell; TCA: tricarboxylic acid cycle; TFAM: mitochondrial transcription factor A; TSC: trophoblast stem cell; UPRmt: mitochondrial unfolded protein response.

2.1.1. SIRT1 (Metabolome-Epigenome Crosstalker)

SIRT1, located both in the nucleus and cytosol, is the largest one in molecular mass and the most extensively studied sirtuin protein [14]. It has been reported to play a crucial role in the SC self-renewal. SIRT1 does not only mediate mouse embryonic SC (mESC) maintenance and embryonic development through deacetylation of methionine adenosyltransferase 2a (MAT2a) [15] but, also modulate mESC differentiation via c-Myc-SMPDL3B signaling cascades [16].

There is growing evidence that decreasing intracellular NAD+ level and SIRT1 activity are associated with SC senescence in vivo. The reduction of cellular NAD+ pools blunts the adaptive mitochondrial unfolded protein response (UPRmt) pathway, ultimately leading to a loss of mitochondrial homeostasis in a SIRT1-dependent manner. Mitochondrial dysfunction is a biomarker of muscle SC (MuSC) senescence that reduces SC cell number and self-renewal capacity [8]. Moreover, cell culture expansion in vitro induces replicative senescence and loss of NAD+ homeostasis in hMSCs which correlates with the decreasing of the SIRT1 signaling activity [17]. Similarly, nicotinamide phosphoribosyltransferase (NAMPT), known as a rate-limiting enzyme in the NAD+ salvage pathway, suppresses rat MSC senescence via NAD+-SIRT1 signaling [18]. Above results demonstrate that NAD+-SIRT1 axis dysfunction might be a potential checkpoint for stemness loss and homeostasis disruption of SCs.

Furthermore, the NAD+-dependent SIRT1 switches metabolic signaling into epigenetics regulation by decreasing H4K16 acetylation and inactivation of muscle gene transcription in MuSCs [14]. SIRT1 also regulates neural SC (NSC) fates by modulating the circadian clock possibly through FOXO3a deacetylation and acts as a gatekeeper of NSCs by responding to metabolic stress [19]. Similarly, SIRT1 also plays an important role in the maintenance of cancer stem cell (CSC) self-renewal. SIRT1 deacetylates β-catenin to trigger its concentration in the nucleus. The nuclear β-catenin further promotes the transcription of NANOG, which aids in the maintenance of liver CSC self-renewal [20]. SOX2 is also a key downstream regulator of SIRT1-mediated liver CSC self-renewal and tumorigenicity. SIRT1 controls the SOX2 gene transcription through chromatin-based epigenetic modification that is reliant on DNA methylation [21]. These results imply its important regulatory roles in the metabolome-epigenome signaling cascade in SC homeostasis.

2.1.2. SIRT2 (Major Cytosolic Sirtuin)

SIRT2 is the only sirtuin protein predominantly resided in the cytosol, although it has also been found in the mitochondria and nucleus [22, 23]. It is identified as a direct regulator of PAX7 acetylation and asymmetric division in MuSCs [24]. SIRT2 also prevents mitochondrial stress-induced hematopoietic SC (HSC) death by repressing NLRP3 inflammasome and caspase 1 activation [25]. In addition, SIRT2 inhibits the activities of four glycolytic enzymes (ALDOA, GAPDH, PGK1, and ENO1) by regulating their acetylation status, thereby interfering the metabolic state during somatic reprogramming of induced pluripotent SCs (iPSCs) [26]. These findings suggest that SIRT2 might be another key enzyme in SC homeostasis, involved in transcriptional activity, inflammatory response, and metabolic switch.

2.1.3. SIRT3 (Dual Function for Mitochondrial Homeostasis and Genomic Stability)

SIRT3 is a major mitochondrial deacetylase, regulating mitochondrial metabolism [27], also reported to have a role in deacetylating histones in the nucleus [28]. It is the most abundant sirtuin family member in HSCs for their homeostasis [29]. SIRT3 cooperates with autophagy by promoting its expression to decelerate hematopoietic aging, and positive intervention to the autophagy-SIRT3 axis leads to blood rejuvenation [30]. Besides, SIRT3 decreases in human MSCs with in vitro passaging; its knockdown accelerates aging and inhibits efficient differentiation of MSCs into adipocytes and osteoblasts; instead, SIRT3 overexpression restores their differentiation capacity and reduces oxidative stress in later-passage MSCs [31]. The mechanisms involved may contribute to that SIRT3 helps to activate antioxidant enzymes (i.e., CAT and MnSOD) [32] and mitophagy inducing by enhanced mitochondrial ROS [33] and stabilize heterochromatin to counteract MSC senescence by interacting with nuclear envelope proteins and heterochromatin-associated proteins [34]. SIRT3 is also related to regulating mitochondrial quality and function for cellular adaption to hypoxia in rat MSCs [35], promoting β cell maturation of mouse ES cells through tricarboxylic acid cycle [36], and ameliorating microglia activation-induced oxidative stress injury through mitochondrial apoptosis pathway in mouse NSCs [37]. A recent research has found that aerobic respiration is upregulated in spermatogonial stem cell (SSC) differentiation in a SIRT3-dependent manner [38, 39]. Above reports show that SIRT3 might regulate SC behavior through multiple pathways, including mitochondrial homeostasis and genomic stability.

2.1.4. SIRT4 (Negative Regulatory Switch)

SIRT4 plays a critical role in cellular metabolism and DNA damage responses in mitochondria [40]. Compared with the other mitochondrial sirtuins, the enzymatic activity of SIRT4 is poorly understood in SCs [12]. Its overexpression triggers senescence in trophoblast SCs (TSCs) due to redox imbalance [41]. Loss of SIRT4 promotes the self-renewal of breast CSCs [42]. SIRT4 appears to exhibit a negative regulatory effect on SC homeostasis, while it remains fundamentally unexplored.

2.1.5. SIRT5 (the Most Widely Functional Sirtuin?)

SIRT5 is a mitochondrial sirtuin localized to the mitochondrial matrix [43]. Interestingly, its target proteins have been found in the mitochondrial matrix and intermembrane space [44], cytosol [45], peroxisome [46], and nucleus [47]. Successive passages in vitro cause SIRT5 accumulation in adipose-derived mesenchymal SCs (ADMSCs) with loss of stemness, eventually accelerating cell senescence. SIRT5-deficient ADMSCs exhibit a higher proliferation rate, delayed senescence, decreasing ROS accumulation, and elevating aerobic glycolysis and attenuating mitochondrial respiration in a more endogenous metabolic pattern [48]. In contrast, SIRT5 has also been reported to have positive effects on oxidative phosphorylation [49, 50]. Considering oxidative phosphorylation as the major source of ROS, it is quite surprising that SIRT5-deficient murine embryonic fibroblasts (MEFs) show higher ROS levels [50]. Although SIRT5 function is not well defined, it may be a potential target to promote the self-renewal capacities and maintain the physiological functions of SCs due to its multiple cellular locations [51].

2.1.6. SIRT6 (Specific Switch of Histone H3)

SIRT6, only found in the nucleus, specifically modifies histone H3 to regulate several fundamental processes about lifespan [52]. SIRT6-deficient HSCs exhibited impaired self-renewal ability [53], and its roles in maintaining SC homeostasis and human development might be related to modification of histone H3. Besides, its chromatin remodeling activity is a critical modulator of human development, regulating the transition from pluripotency to differentiated state [5457]. Human iPSCs derived from SIRT6 Asp63His (D63H) mutant fail to differentiate into embryoid bodies (EBs), functional cardiomyocytes, and neural progenitor cells (NPCs) because of sustained pluripotent gene expression [54]. However, it has been reported to maintain MSC homeostasis by serving as a NRF2 coactivator to transactivate the NRF2-driven antioxidant genes [58]. SIRT6 can also regulate HSC homeostasis through the transcriptional repression of Wnt target genes. Therefore, the function of SIRT6 in maintaining SC homeostasis may not be limited to the nucleus.

2.1.7. SIRT7 (Specific Nucleolar Sirtuin)

SIRT7 is the only sirtuin protein that mainly localizes to the nucleolus [59], involving in ribosomal biogenesis [60], stress responses [61], and senescence [62]. In HSCs, SIRT7 interacts NRF1 to regulate cellular energy metabolism and proliferation through the UPRmt. In addition, SIRT7 upregulation improved the regenerative capacity of aged HSCs [63] and hMSCs [64]. Mechanistically, SIRT7 forms a complex with nuclear lamins and heterochromatin proteins to maintain the repressive state of heterochromatin [64]. It also can activate quiescent hair follicle SCs (HFSCs) to initiate the cell cycle by destabilizing NFATc1 [65] and regulate MSC osteogenic differentiation partly by activating Wnt/β-catenin signaling [66]. Because of its special location, SIRT7 may play a special role in the regulation of SC homeostasis, which is worthy of further study.

Some sirtuins are detected in multiple cellular compartments, and to shuttle among the various cellular localizations [67]. Different sirtuins have complementary functions and extensive crosstalk to maintain SC homeostasis, besides their own distinct functions. For instance, mitochondrial sirtuins are able to manage these delicate processes accurately by crosstalk between the mitochondria and nucleus [12]. Yuan et al. reported that the reducing expressions of SIRT1 and SIRT3 fail to regulate mitochondrial fitness, DNA repair, and other aging-associated pathways during hMSC culture expansion [17]. SIRT4 and SIRT1 have an inverse relationship in breast CSCs, and SIRT4 inhibits SIRT1 expression by suppressing glutamine metabolism [42]. However, their cooperative relationship and mechanisms are still unclear and poorly understood, and more in-depth studies are needed [12].

2.2. PARPs

PARPs family consisted of 17 proteins, widely distributed in all human tissues, and involved in a variety of cellular functions, such as the cellular response to DNA damage and the regulation of gene transcription [68, 69]. PARPs transfer the ADP-ribose from NAD+ to fundamental biomolecules (known as PARylation) containing proteins, DNA, and RNA [11, 7073]. Recently, PARP research has been enriched by the discovery of novel PARP1 interaction partners that regulate its enzymatic activity [74, 75]. It shows that PARP1 contributes to pluripotency, lineage specific differentiation, and reprogramming in various SCs [76].

PARP1 is a component of the groucho/TLE-corepressor complex, which mediates dismissal of the corepressor complex from HES1-regulated promoters during neural stem/progenitor cell (NSC) differentiation [77]. During this process, PARP1 mediates histone H1 eviction from the chromatin fiber [78]. Similarly, PARP1 interacts with PARylates SOX2 directly, in which it may be required for dissociation and degradation of inhibitory SOX2 proteins from the FGF4 enhancer during ESC differentiation [79]. PARP1 can also dominate NSC proliferation by modulating platelet-derived growth factor receptor α (PDGFRα) expression [80]. PARylation is reported to promote the proliferation and self-renewal of mouse brain NSCs by inhibiting p53 activation [81]. PARP1 loss leads to defects in brain development, increased neuronal density at birth in mice, and enhanced embryonic NSC adhesion to N-cadherin in vitro [82].

These results demonstrate the chromatin-related function of PARP1, which PARylates different transcription factors to modulate their DNA binding and transcriptional activity, thereby regulating SC homeostasis [83]. Therefore, site-specific PARylation to drive cell fate may be a very promising approach in SC therapy. Remarkably, CSCs have an increased DNA damage response, and PARP1 is upregulated due to its crucial involvement in DNA repair [84, 85]. It implies that PARP inhibitors can be employed as therapeutic strategies to target CSCs, such as FDA-approved olaparib and rucaparib.

3. Category II: NAD+ Glycohydrolases (Also Referred to as NADases, including CD38, CD157, and Sterile Alpha and Toll/Interleukin Receptor (TIR) Motif-Containing 1 (SARM1))

3.1. CD38

CD38 is considered a type II protein with the catalytic domain facing outside. It can also exist in an opposite type III orientation with its catalytic domain facing the cytosol [63, 86]. CD38 is observed in intracellular membranes, including endoplasmic reticulum, nucleus, mitochondria, and endolysosome [63, 8790]. It catalyzes the synthesis of ADPR and cADPR using NAD+ as a substrate and is an important regulator of extracellular and intracellular NAD+ pools [91]. Both ADPR and cADPR act as second messengers controlling multiple cell functions through inducing intracellular Ca2+ fluxes independent of IP3 [92]. CD38 has been reported to play a role in SC differentiation [93]. For example, the NAD+/CD38/cADPR/Ca2+ signaling pathway antagonizes the cardiomyocyte differentiation of mouse ESCs [94].

Remarkably, increasing CD38 expression leads to a decline of NAD metabolites and distorts other NAD+-consuming enzyme activities in aged mice [91]. CD38 but not SIRT1 or PARPs is considered the predominant NAD+ consumers [95]. Therefore, it should also be considered whether CD38 has a specific regulatory role in SC senescence.

3.2. CD157

CD157 is a glycophosphatidylinositol-anchored protein. The ADP-ribosyl cyclase activity of CD157 is weaker than that of CD38. CD157 may participate in the embryonic and adult nervous systems partially through cADPR production [96]. CD157 upregulation increases the biosynthesis and transition of mitochondria from BMSCs to injured neurons, thus improves the neuroregeneration and inhibits cell apoptosis via calcium-dependent CD157/cyclic ADP-ribose pathway [97]. In addition, CD157 is a marker of tissue-resident vascular endothelial SCs (VESCs) [98, 99]. The CD157-positive endothelial cells have SC properties, including homeostatic capillary maintenance and regenerative capacity after vascular injury in vivo [100]. However, whether the functions of CD157 as a cell receptor are relevant to NAD+ metabolism remains unclear [3].

3.3. SARM1

The Toll/interleukin receptor (TIR) domain is necessary for SARM1 activity. Dimerization of TIR domain cleaves NAD+ into ADP-ribose, cADPR, and NAM [7, 101]. In addition to its involvement in innate immunity [102, 103], SARM1 is thought to an important NAD+-consuming enzyme during axonal injury in neurons [1, 104, 105]. Inhibition of SARM1 activation may be a compelling therapeutic target to treat neurodegenerative diseases [106108]. However, the role of SARM1 in regulating SC homeostasis is currently unclear. Our RNA-Seq results (SRA: SRP152900) revealed significantly higher levels of SARM1 expression in differentiated limbal SCs (LSCs) compared to undifferentiated ones. This suggests that maintaining SC stemness may also require inhibition of SARM1 activation.

Moreover, one-carbon metabolism enzyme methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) has been reported to use NAD+ as a cofactor [109]. It is a dual-function factor for determining the pluripotency of pluripotent SCs through both preventing mitochondrial dysfunction and promoting homologous recombination repair in nucleus [110]. NAD+ can also be consumed by NAD+ kinase (NADK) to increase NADP+ production [111]. Through CD38, NADP+ is further converted into NAADP, which is another Ca2+-mobilizing second messenger [112]. Unfortunately, there is no report on crosstalk between NADK and CD38 in the regulation of SC homeostasis.

4. Conclusion

The NAD+ consumers systematically reduce the pool of NAD+ available for NAD+-specific enzymes and processes [11, 13]. These different enzymes can also interplay with each other to perform the same cellular function (Table 2). For instance, aging triggers NAD+ loss because of PARP activation in old mice, and NAD+ reduction lowers the activity of the antiaging proteins, sirtuins, leading to a feedforward cycle of aging [8, 113]. Another study found that CD38 has a central role in age-related NAD+ decline. CD38 inhibitor 78c upregulates NAD+ levels, thereby activating prolongevity and health span-related factors, including sirtuins and PARPs [114]. Moreover, PARP inhibitor PJ34 preserved intracellular NAD+ levels, increased SIRT1 activity, and improved the function in aging-induced endothelial progenitor cells (EPCs) [115]. In CSCs, NAMPT regulates epithelial-mesenchymal transition (EMT) and tumor dedifferentiation/reprograming via controlling cellular functions that promote proliferation and pathways mediated by SIRT1 and PARP1 [116]. Above results suggest that NAD+ availability can be achieved by inhibiting one or more of the NAD+ consumers to crosstalk the regulatory networks. Of course, SC functions might be restored or improved by supplying NAD+ precursors for biosynthesis [17, 50], including nicotinamide riboside (NR) [8, 117119], NAM [120], and nicotinamide mononucleotide (NMN) (Table 3) [121]. This may be a potential approach for maintaining SC homeostasis and is a challenge for clinical treatment of related diseases.

Table 2.

Functions of NAD+-consuming enzymes in stem cells.

Functions NAD+-consuming enzymes
Stemness and homeostasis SIRT1, SIRT3, SIRT6, PARP1, CD157, MTHFD2
Self-renewal SIRT1, SIRT4, SIRT6, PARP1
Mitochondrial fitness SIRT1, SIRT2, SIRT3, SIRT5, CD157
Senescence SIRT1, SIRT3, SIRT4, SIRT5, SIRT7, CD38
Differentiation SIRT2, SIRT3, SIRT6, SIRT7, PARP1, CD38, SARM1?, NADK?
Metabolic regulation SIRT2, SIRT7, CD38

↑: positive effect on the function; ↓: negative effect on the function; MTHFD2: methylenetetrahydrofolate dehydrogenase 2; NADK: NAD+ kinase; PARP: poly(ADP-ribose) polymerase; SARM1: sterile alpha and Toll/interleukin receptor (TIR) motif-containing 1; SIRT: sirtuin.

Table 3.

Improvement of stem cell homeostasis by adding NAD+ precursors.

NAD+ precursors Concentrations Stem cells Functions References
NR 400 mg/kg/day MuSC Antiaging [8]
500 mg/kg/day ISC Antiaging [117]
500 μM iPSC Ameliorating mitochondrial function [118]
400 mg/kg/day HSC Restoring youthful metabolic capacity [119]

NAM 10 mM hESC Promoting pancreatic differentiation [120]

NMN 0.03–2.25 μM MSC Maintaining self-renewal [121]

hESC: human embryonic stem cell; HSC: hematopoietic stem cell; iPSC: induced pluripotent stem cell; ISC: intestinal stem cell; MSC: mesenchymal stromal cell; MuSC: muscle stem cell; NAM: nicotinamide; NMN: nicotinamide mononucleotide; NR: nicotinamide riboside.

Acknowledgments

This work was supported by the National Natural Science Foundation of China (No. 31601104) and the Natural Science Foundation of Shandong Province (No. ZR2021MC158).

Abbreviations

ADMSC:

Adipose-derived mesenchymal stem cell

ADP:

Adenosine diphosphate

ADPR:

ADP-ribose

ALDOA:

Aldolase A

cADPR:

Cyclic ADPR

CAT:

Catalase

CSC:

Cancer stem cells

EB:

Embryoid body

EPC:

Endothelial progenitor cell

EMT:

Epithelial-mesenchymal transition

ENO1:

Enolase 1

FGF4:

Fibroblast growth factor 4

FOXO3a:

Forkhead box O3a

GAPDH:

Glyceraldehyde-3-phosphate dehydrogenase

HFSC:

Hair follicle stem cell

HSC:

Hematopoietic stem cell

iPSC:

Induced pluripotent stem cell

LSC:

Limbal stem cell

MAT2a:

Methionine adenosyltransferase 2a

MEF:

Murine embryonic fibroblast

mESC:

Mouse embryonic stem cell

MTHFD2:

Methylenetetrahydrofolate dehydrogenase 2

MuSC:

Muscle stem cell

MnSOD:

Manganese superoxide dismutase

NAD+:

Nicotinamide adenine dinucleotide

NADK:

NAD+ kinase

NAM:

Nicotinamide

NAMPT:

Nicotinamide phosphoribosyltransferase

NFATc1:

Nuclear factor of activated T cells c1

NLRP3:

Nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3

NPC:

Neural progenitor cell

NRF1:

Nuclear factor erythroid 2-related factor 1

NRF2:

Nuclear factor erythroid 2-related factor 2

NMN:

Nicotinamide mononucleotide

NR:

Nicotinamide riboside

NSC:

Neural stem cell

PARP:

Poly(ADP-ribose) polymerase

PAX7:

Paired box 7

PDGFRα:

Platelet-derived growth factor receptor α

PGK1:

Phosphoglycerate kinase 1

ROS:

Reactive oxygen species

SARM1:

Sterile alpha and Toll/interleukin receptor (TIR) motif-containing 1

SC:

Stem cell

SIRT:

Sirtuin

SMPDL3B:

Sphingomyelin phosphodiesterase acid-like 3B

SOX2:

SRY-box transcription factor 2

SSC:

Spermatogonial stem cell

TET2:

Ten-eleven translocation-2

TIR:

Toll/interleukin receptor

TSC:

Trophoblast stem cell

UPRmt:

Mitochondrial unfolded protein response

VESC:

Vascular endothelial stem cell.

Data Availability

Our RNA-Seq data supporting this review have been deposited in the Sequence Read Archive (SRA) database at NCBI under the accession number SRP152900. The raw data are available (https://www.ncbi.nlm.nih.gov/sra/SRP152900).

Conflicts of Interest

The authors declare that there is no conflict of interest regarding the publication of this paper.

Supplementary Materials

Supplementary Materials

Supplementary Figure S1: literature search method and selection criteria. Searches in Web of Science and PubMed were restricted to papers published between 2016 and 2022. The basic framework of the intervention program was determined by searching for a combination of subject terms and free words, such as “stem cell,” “NAD enzyme,” and “homeostasis.” Exclusion criteria for literature are as follows: no complete text accessible; publications written in a language other than English; and studies with inadequate literature evaluation.

References

  • 1.Katsyuba E., Romani M., Hofer D., Auwerx J. NAD+ homeostasis in health and disease. Nature Metabolism . 2020;2(1):9–31. doi: 10.1038/s42255-019-0161-5. [DOI] [PubMed] [Google Scholar]
  • 2.Tannous C., Booz G. W., Altara R., et al. Nicotinamide adenine dinucleotide: biosynthesis, consumption and therapeutic role in cardiac diseases. Acta Physiologica . 2021;231(3, article e13551) doi: 10.1111/apha.13551. [DOI] [PubMed] [Google Scholar]
  • 3.Covarrubias A. J., Perrone R., Grozio A., Verdin E. NAD+ metabolism and its roles in cellular processes during ageing. Nature Reviews. Molecular Cell Biology . 2021;22(2):119–141. doi: 10.1038/s41580-020-00313-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Navarro M. N., de Las Heras M. M. G., Mittelbrunn M. Nicotinamide adenine dinucleotide metabolism in the immune response, autoimmunity and inflammageing. British Journal of Pharmacology . 2022;179(9):1839–1856. doi: 10.1111/bph.15477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ralto K. M., Rhee E. P., Parikh S. M. NAD+ homeostasis in renal health and disease. Nature Reviews Nephrology . 2020;16(2):99–111. doi: 10.1038/s41581-019-0216-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Shang J., Smith M. R., Anmangandla A., Lin H. NAD+-consuming enzymes in immune defense against viral infection. The Biochemical Journal . 2022;478(23):4071–4092. doi: 10.1042/BCJ20210181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Xie N., Zhang L., Gao W., Huang C., Zou B. NAD+ metabolism: pathophysiologic mechanisms and therapeutic potential. Signal Transduction and Targeted Therapy . 2020;5(1):p. 227. doi: 10.1038/s41392-020-00311-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zhang H., Ryu D., Wu Y., et al. NAD+ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science . 2016;352(6292):1436–1443. doi: 10.1126/science.aaf2693. [DOI] [PubMed] [Google Scholar]
  • 9.Du J., Zhou Y., Su X., et al. Sirt5 is a NAD-dependent protein lysine demalonylase and desuccinylase. Science . 2011;334(6057):806–809. doi: 10.1126/science.1207861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Imai S. I., Armstrong C. M., Kaeberlein M., Guarente L. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature . 2000;403(6771):795–800. doi: 10.1038/35001622. [DOI] [PubMed] [Google Scholar]
  • 11.Cambronne X. A., Kraus W. L. Location, Location, Location: Compartmentalization of NAD+ Synthesis and Functions in Mammalian Cells. Trends in Biochemical Sciences . 2020;45(10):858–873. doi: 10.1016/j.tibs.2020.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Elkhwanky M. S., Hakkola J. Extranuclear sirtuins and metabolic stress. Antioxidants & Redox Signaling . 2018;28(8):662–676. doi: 10.1089/ars.2017.7270. [DOI] [PubMed] [Google Scholar]
  • 13.Wang M., Lin H. Understanding the function of mammalian sirtuins and protein lysine acylation. Annual Review of Biochemistry . 2021;90(1):245–285. doi: 10.1146/annurev-biochem-082520-125411. [DOI] [PubMed] [Google Scholar]
  • 14.Ryall J. G., Dell’Orso S., Derfoul A., et al. The NAD+-dependent SIRT1 deacetylase translates a metabolic switch into regulatory epigenetics in skeletal muscle stem cells. Cell Stem Cell . 2015;16(2):171–183. doi: 10.1016/j.stem.2014.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Tang S., Fang Y., Huang G., et al. Methionine metabolism is essential for SIRT1-regulated mouse embryonic stem cell maintenance and embryonic development. The EMBO Journal . 2017;36(21):3175–3193. doi: 10.15252/embj.201796708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Fan W., Tang S., Fan X., et al. SIRT1 regulates sphingolipid metabolism and neural differentiation of mouse embryonic stem cells through c-Myc-SMPDL3B. eLife . 2021;10, article e67452 doi: 10.7554/eLife.67452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Yuan X., Liu Y., Bijonowski B. M., et al. NAD+/NADH redox alterations reconfigure metabolism and rejuvenate senescent human mesenchymal stem cells in vitro. Communications Biology . 2020;3(1):p. 774. doi: 10.1038/s42003-020-01514-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Pi C., Yang Y., Sun Y., et al. Nicotinamide phosphoribosyltransferase postpones rat bone marrow mesenchymal stem cell senescence by mediating NAD+-Sirt1 signaling. Aging . 2019;11(11):3505–3522. doi: 10.18632/aging.101993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Draijer S., Chaves I., Hoekman M. F. M. The circadian clock in adult neural stem cell maintenance. Progress in Neurobiology . 2019;173:41–53. doi: 10.1016/j.pneurobio.2018.05.007. [DOI] [PubMed] [Google Scholar]
  • 20.Chen X., Huan H., Liu C., et al. Deacetylation of β-catenin by SIRT1 regulates self-renewal and oncogenesis of liver cancer stem cells. Cancer Letters . 2019;463:1–10. doi: 10.1016/j.canlet.2019.07.021. [DOI] [PubMed] [Google Scholar]
  • 21.Liu L., Liu C., Zhang Q., et al. SIRT1-mediated transcriptional regulation of SOX2 is important for self- renewal of liver cancer stem cells. Hepatology . 2016;64(3):814–827. doi: 10.1002/hep.28690. [DOI] [PubMed] [Google Scholar]
  • 22.Vaquero A., Scher M. B., Lee D. H., et al. SirT2 is a histone deacetylase with preference for histone H4 Lys 16 during mitosis. Genes & Development . 2006;20(10):1256–1261. doi: 10.1101/gad.1412706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Liu G., Park S. H., Imbesi M., et al. Loss of NAD-dependent protein deacetylase sirtuin-2 alters mitochondrial protein acetylation and dysregulates mitophagy. Antioxidants & Redox Signaling . 2017;26(15):849–863. doi: 10.1089/ars.2016.6662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sincennes M. C., Brun C. E., Lin A. Y. T., et al. Acetylation of PAX7 controls muscle stem cell self-renewal and differentiation potential in mice. Nature Communications . 2021;12(1):p. 3253. doi: 10.1038/s41467-021-23577-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Luo H., Mu W. C., Karki R., et al. Mitochondrial stress-initiated aberrant activation of the NLRP3 inflammasome regulates the functional deterioration of hematopoietic stem cell aging. Cell Reports . 2019;26(4):945–954.e4. doi: 10.1016/j.celrep.2018.12.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Liu T. M., Shyh-Chang N. SIRT2 and glycolytic enzyme acetylation in pluripotent stem cells. Nature Cell Biology . 2017;19(5):412–414. doi: 10.1038/ncb3522. [DOI] [PubMed] [Google Scholar]
  • 27.Shimazu T., Hirschey M. D., Hua L., et al. SIRT3 deacetylates mitochondrial 3-hydroxy-3-methylglutaryl CoA synthase 2 and regulates ketone body production. Cell Metabolism . 2010;12(6):654–661. doi: 10.1016/j.cmet.2010.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Scher M. B., Vaquero A., Reinberg D. SirT3 is a nuclear NAD(+)-dependent histone deacetylase that translocates to the mitochondria upon cellular stress. Genes & Development . 2007;21(8):920–928. doi: 10.1101/gad.1527307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Brown K., Xie S., Qiu X., et al. SIRT3 reverses aging-associated degeneration. Cell Reports . 2013;3(2):319–327. doi: 10.1016/j.celrep.2013.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Fang Y., An N., Zhu L., et al. Autophagy-Sirt3 axis decelerates hematopoietic aging. Aging Cell . 2020;19(10, article e13232) doi: 10.1111/acel.13232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Denu R. A. SIRT3 enhances mesenchymal stem cell longevity and differentiation. Oxidative Medicine and Cellular Longevity . 2017;2017:11. doi: 10.1155/2017/5841716.5841716 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zhang D. Y., Gao T., Xu R. J., et al. SIRT3 transfection of aged human bone marrow-derived mesenchymal stem cells improves cell therapy-mediated myocardial repair. Rejuvenation Research . 2020;23(6):453–464. doi: 10.1089/rej.2019.2260. [DOI] [PubMed] [Google Scholar]
  • 33.Guo Y., Jia X., Cui Y., Song Y., Fu D. Sirt3-mediated mitophagy regulates AGEs-induced BMSCs senescence and senile osteoporosis. Redox Biology . 2021;41(1, article 101915) doi: 10.1016/j.redox.2021.101915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Diao Z., Ji Q., Wu Z., et al. SIRT3 consolidates heterochromatin and counteracts senescence. Nucleic Acids Research . 2021;49(8):4203–4219. doi: 10.1093/nar/gkab161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Wang X., Shen K., Wang J., et al. Hypoxic preconditioning combined with curcumin promotes cell survival and mitochondrial quality of bone marrow mesenchymal stem cells, and accelerates cutaneous wound healing via PGC-1α/SIRT3/HIF-1α signaling. Free Radical Biology & Medicine . 2020;159:164–176. doi: 10.1016/j.freeradbiomed.2020.07.023. [DOI] [PubMed] [Google Scholar]
  • 36.Lu L., Zhitao L., Nannan C., et al. Mitofusin2 promotes beta cell maturation from mouse embryonic stem cells via Sirt3/Idh2 activation. Stem Cells International . 2022;2022:18. doi: 10.1155/2022/1172795.1172795 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Jiang D. Q., Wang Y., Li M. X., Ma Y. J., Wang Y. SIRT3 in neural stem cells attenuates microglia activation-induced oxidative stress injury through mitochondrial pathway. Frontiers in Cellular Neuroscience . 2017;11:p. 7. doi: 10.3389/fncel.2017.00007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hermann B. P., Cheng K., Singh A., et al. The mammalian spermatogenesis single-cell transcriptome, from spermatogonial stem cells to spermatids. Cell Reports . 2018;25(6):1650–1667.e8. doi: 10.1016/j.celrep.2018.10.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lord T., Nixon B. Metabolic changes accompanying spermatogonial stem cell differentiation. Developmental Cell . 2020;52(4):399–411. doi: 10.1016/j.devcel.2020.01.014. [DOI] [PubMed] [Google Scholar]
  • 40.Van D., Santos D., Haigis M. C. Mitochondrial sirtuins and molecular mechanisms of aging. Trends in Molecular Medicine . 2017;23(4):320–331. doi: 10.1016/j.molmed.2017.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Castex J., Willmann D., Kanouni T., et al. Inactivation of Lsd1 triggers senescence in trophoblast stem cells by induction of Sirt4. Cell Death & Disease . 2017;8(2, article e2631) doi: 10.1038/cddis.2017.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Du L., Liu X., Ren Y., et al. Loss of SIRT4 promotes the self-renewal of breast cancer stem cells. Theranostics . 2020;10(21):9458–9476. doi: 10.7150/thno.44688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kumar S., Lombard D. B. Functions of the sirtuin deacylase SIRT5 in normal physiology and pathobiology. Critical Reviews in Biochemistry and Molecular Biology . 2018;53(3):311–334. doi: 10.1080/10409238.2018.1458071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Schlicker C., Gertz M., Papatheodorou P., Kachholz B., Becker C. F. W., Steegborn C. Substrates and regulation mechanisms for the human mitochondrial sirtuins Sirt3 and Sirt5. Journal of Molecular Biology . 2008;382(3):790–801. doi: 10.1016/j.jmb.2008.07.048. [DOI] [PubMed] [Google Scholar]
  • 45.Chinopoulos C. The mystery of extramitochondrial proteins lysine succinylation. International Journal of Molecular Sciences . 2021;22(11):p. 6085. doi: 10.3390/ijms22116085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Chen X. F., Tian M. X., Sun R. Q., et al. SIRT5 inhibits peroxisomalACOX1 to prevent oxidative damage and is downregulated in liver cancer. EMBO Reports . 2018;19(5, article e45124) doi: 10.15252/embr.201745124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zorro Shahidian L., Haas M., Le Gras S., et al. Succinylation of H3K122 destabilizes nucleosomes and enhances transcription. EMBO Reports . 2021;22(3, article e51009) doi: 10.15252/embr.202051009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ou T., Yang W., Li W., et al. SIRT5 deficiency enhances the proliferative and therapeutic capacities of adipose-derived mesenchymal stem cells via metabolic switching. Clinical and Translational Medicine . 2020;10(5, article e172) doi: 10.1002/ctm2.172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zhou L., Wang F., Sun R., et al. SIRT5 promotes IDH2 desuccinylation and G6PD deglutarylation to enhance cellular antioxidant defense. EMBO Reports . 2016;17(6):811–822. doi: 10.15252/embr.201541643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Zhang Y., Bharathi S. S., Rardin M. J., et al. SIRT5 binds to cardiolipin. The Journal of Biological Chemistry . 2017;292(24):10239–10249. doi: 10.1074/jbc.M117.785022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Salazar-Noratto G. E., Luo G., Denoeud C., et al. Understanding and leveraging cell metabolism to enhance mesenchymal stem cell transplantation survival in tissue engineering and regenerative medicine applications. Stem Cells . 2020;38(1):22–33. doi: 10.1002/stem.3079. [DOI] [PubMed] [Google Scholar]
  • 52.Liu G., Chen H., Liu H., Zhang W., Zhou J. Emerging roles of SIRT6 in human diseases and its modulators. Medicinal Research Reviews . 2021;41(2):1089–1137. doi: 10.1002/med.21753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Wang H., Diao D., Shi Z., et al. SIRT6 controls hematopoietic stem cell homeostasis through epigenetic regulation of Wnt signaling. Cell Stem Cell . 2016;18(4):495–507. doi: 10.1016/j.stem.2016.03.005. [DOI] [PubMed] [Google Scholar]
  • 54.Ferrer C. M., Alders M., Postma A. V., et al. An inactivating mutation in the histone deacetylase SIRT6 causes human perinatal lethality. Genes & Development . 2018;32(5–6):373–388. doi: 10.1101/gad.307330.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Zhang W., Wan H., Feng G., et al. SIRT6 deficiency results in developmental retardation in cynomolgus monkeys. Nature . 2018;560(7720):661–665. doi: 10.1038/s41586-018-0437-z. [DOI] [PubMed] [Google Scholar]
  • 56.Xu P., Wang T. T., Liu X. Z., et al. Sirt6 regulates efficiency of mouse somatic reprogramming and maintenance of pluripotency. Stem Cell Research & Therapy . 2019;10(1):p. 9. doi: 10.1186/s13287-018-1109-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wei H., Khawar M. B., Tang W., Wang L., Li W. Sirt6 is required for spermatogenesis in mice. Aging . 2020;12(17):17099–17113. doi: 10.18632/aging.103641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Pan H., Guan D., Liu X., et al. SIRT6 safeguards human mesenchymal stem cells from oxidative stress by coactivating NRF2. Cell Research . 2016;26(2):190–205. doi: 10.1038/cr.2016.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Michishita E., Park J. Y., Burneskis J. M., Barrett J. C., Horikawa I. Evolutionarily conserved and nonconserved cellular localizations and functions of human SIRT proteins. Molecular Biology of the Cell . 2005;16(10):4623–4635. doi: 10.1091/mbc.e05-01-0033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Chen S., Blank M. F., Iyer A., et al. SIRT7-dependent deacetylation of the U3-55k protein controls pre-rRNA processing. Nature Communications . 2016;7(1, article 10734) doi: 10.1038/ncomms10734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Shin J., He M., Liu Y., et al. SIRT7 represses Myc activity to suppress ER stress and prevent fatty liver disease. Cell Reports . 2013;5(3):654–665. doi: 10.1016/j.celrep.2013.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Paredes S., Angulo-Ibanez M., Tasselli L., et al. SIRT7 regulates rDNA instability-induced senescence. The Journal of Biological Chemistry . 2018;293(28):11242–11250. doi: 10.1074/jbc.AC118.003325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Liu J., Zhao Y. J., Li W. H., Hou Y. N., Lee H. C. Cytosolic interaction of type III human CD38 with CIB1 modulates cellular cyclic ADP-ribose levels. Proceedings of the National Academy of Sciences of the United States of America . 2017;114(31):8283–8288. doi: 10.1073/pnas.1703718114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Bi S., Liu Z., Wu Z., et al. SIRT7 antagonizes human stem cell aging as a heterochromatin stabilizer. Protein & Cell . 2020;11(7):483–504. doi: 10.1007/s13238-020-00728-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Li G., Tang X., Zhang S., et al. SIRT7 activates quiescent hair follicle stem cells to ensure hair growth in mice. The EMBO Journal . 2020;39(18, article e104365) doi: 10.15252/embj.2019104365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Chen E. E. M., Zhang W., Ye C. C. Y., et al. Knockdown of SIRT7 enhances the osteogenic differentiation of human bone marrow mesenchymal stem cells partly via activation of the Wnt/ β-catenin signaling pathway. Cell Death & Disease . 2017;8(9, article e3042) doi: 10.1038/cddis.2017.429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Nakamura Y., Ogura M., Tanaka D., Inagaki N. Localization of mouse mitochondrial SIRT proteins: shift of SIRT3 to nucleus by co-expression with SIRT5. Biochemical and Biophysical Research Communications . 2008;366(1):174–179. doi: 10.1016/j.bbrc.2007.11.122. [DOI] [PubMed] [Google Scholar]
  • 68.Koh D. W., Dawson T. M., Dawson V. L. Mediation of cell death by poly(ADP-ribose) polymerase-1. Pharmacological Research . 2005;52(1):5–14. doi: 10.1016/j.phrs.2005.02.011. [DOI] [PubMed] [Google Scholar]
  • 69.Zhang L., Cao J., Dong L., Lin H. TiPARP forms nuclear condensates to degrade HIF-1α and suppress tumorigenesis. Proceedings of the National Academy of Sciences of the United States of America . 2020;117(24):13447–13456. doi: 10.1073/pnas.1921815117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Diefenbach J., Burkle A. Poly-ADP-ribosylation in health and disease. Cellular and Molecular Life Sciences: CMLS . 2005;62(7–8):721–730. doi: 10.1007/s00018-004-4503-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hassa P. O., Haenni S. S., Elser M., Hottiger M. O. Nuclear ADP-ribosylation reactions in mammalian cells: where are we today and where are we going? Microbiology and Molecular Biology Reviews . 2006;70(3):789–829. doi: 10.1128/MMBR.00040-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Groslambert J., Prokorova E., Ahel I. ADP-ribosylation of DNA and RNA. DNA Repair . 2021;105, article 103144 doi: 10.1016/j.dnarep.2021.103144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Cohen M. S. Interplay between compartmentalized NAD+ synthesis and consumption: a focus on the PARP family. Genes & Development . 2020;34(5–6):254–262. doi: 10.1101/gad.335109.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Azarm K., Smith S. Nuclear PARPs and genome integrity. Genes & Development . 2020;34(5–6):285–301. doi: 10.1101/gad.334730.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Alemasova E. E., Lavrik O. I. Poly(ADP-ribosyl) ation by PARP1: reaction mechanism and regulatory proteins. Nucleic Acids Research . 2019;47(8):3811–3827. doi: 10.1093/nar/gkz120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Zeniou M., Nguekeu-Zebaze L., Dantzer F. Therapeutic considerations of PARP in stem cell biology: relevance in cancer and beyond. Biochemical Pharmacology . 2019;167:107–115. doi: 10.1016/j.bcp.2019.06.012. [DOI] [PubMed] [Google Scholar]
  • 77.Ju B. G., Solum D., Song E. J., et al. Activating the PARP-1 Sensor Component of the Groucho/ TLE1 Corepressor Complex Mediates a CaMKinase IIδ-Dependent Neurogenic Gene Activation Pathway. Cell . 2004;119(6):815–829. doi: 10.1016/j.cell.2004.11.017. [DOI] [PubMed] [Google Scholar]
  • 78.Hau A. C., Grebbin B. M., Agoston Z., et al. MEIS homeodomain proteins facilitate PARP1/ARTD1-mediated eviction of histone H1. The Journal of Cell Biology . 2017;216(9):2715–2729. doi: 10.1083/jcb.201701154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Gao F., Kwon S. W., Zhao Y., Jin Y. PARP1 Poly(ADP-ribosyl)ates Sox2 to Control Sox2 Protein Levels and FGF4 Expression during Embryonic Stem Cell Differentiation. Journal of Biological Chemistry . 2009;284(33):22263–22273. doi: 10.1074/jbc.M109.033118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Dis A., Sh B., Kssc D., Sjka B. PARP-1 regulates mouse embryonic neural stem cell proliferation by regulating PDGFRα expression. Biochemical and Biophysical Research Communications . 2020;526(4):986–992. doi: 10.1016/j.bbrc.2020.03.166. [DOI] [PubMed] [Google Scholar]
  • 81.Okuda A., Kurokawa S., Takehashi M., et al. Poly(ADP-ribose) polymerase inhibitors activate the p53 signaling pathway in neural stem/progenitor cells. BMC Neuroscience . 2017;18(1):p. 14. doi: 10.1186/s12868-016-0333-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Nelson M. M., Hoff J. D., Zeese M. L., Corfas G. Poly (ADP-ribose) polymerase 1 regulates cajal-retzius cell development and neural precursor cell adhesion. Frontiers in Cell and Developmental Biology . 2021;9, article 693595 doi: 10.3389/fcell.2021.693595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Luo X., Ryu K. W., Kim D. S., et al. PARP-1 Controls the Adipogenic Transcriptional Program by PARylating C/EBPβ and Modulating Its Transcriptional Activity. Molecular Cell . 2017;65(2):260–271. doi: 10.1016/j.molcel.2016.11.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Chaudhuri A. R., Nussenzweig A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nature Reviews. Molecular Cell Biology . 2017;18(10):610–621. doi: 10.1038/nrm.2017.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Manic G., Sistigu A. A., Corradi F., Musella M., de Maria R., Vitale I. Replication stress response in cancer stem cells as a target for chemotherapy. Seminars in Cancer Biology . 2018;53:31–41. doi: 10.1016/j.semcancer.2018.08.003. [DOI] [PubMed] [Google Scholar]
  • 86.Chini C. C. S., Peclat T. R., Warner G. M., Kashyap S., Chini E. N. CD38 ecto-enzyme in immune cells is induced during aging and regulates NAD+ and NMN levels. Nature Metabolism . 2020;2(11):1284–1304. doi: 10.1038/s42255-020-00298-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Muñoz P., Mittelbrunn M., de la Fuente H., et al. Antigen-induced clustering of surface CD38 and recruitment of intracellular CD38 to the immunologic synapse. Blood . 2008;111(7):3653–3664. doi: 10.1182/blood-2007-07-101600. [DOI] [PubMed] [Google Scholar]
  • 88.van de Donk N., Richardson P. G., Malavasi F. CD38 antibodies in multiple myeloma: back to the future. Blood . 2018;131(1):13–29. doi: 10.1182/blood-2017-06-740944. [DOI] [PubMed] [Google Scholar]
  • 89.Chini E. N., Chini C. C. S., Espindola Netto J. M., de Oliveira G. C., van Schooten W. The pharmacology of CD38/NADase: an emerging target in cancer and diseases of aging. Trends in Pharmacological Sciences . 2018;39(4):424–436. doi: 10.1016/j.tips.2018.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Bock K. W. Functions of aryl hydrocarbon receptor (AHR) and CD38 in NAD metabolism and nonalcoholic steatohepatitis (NASH) Biochemical Pharmacology . 2019;169, article 113620 doi: 10.1016/j.bcp.2019.08.022. [DOI] [PubMed] [Google Scholar]
  • 91.Camacho-Pereira J., Tarragó M. G., Chini C. C., et al. CD38 dictates age-related NAD decline and mitochondrial dysfunction through an SIRT3-dependent mechanism. Cell Metabolism . 2016;23(6):1127–1139. doi: 10.1016/j.cmet.2016.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Piedra-Quintero Z. L., Wilson Z., Nava P., Guerau-de-Arellano M. CD38: An immunomodulatory molecule in inflammation and autoimmunity. Frontiers in Immunology . 2020;11, article 597959 doi: 10.3389/fimmu.2020.597959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Hao B., Webb S. E., Miller A. L., Yue J. The role of Ca2+ signaling on the self-renewal and neural differentiation of embryonic stem cells (ESCs) Cell Calcium . 2016;59(2–3):67–74. doi: 10.1016/j.ceca.2016.01.004. [DOI] [PubMed] [Google Scholar]
  • 94.Wei W., Sun H., Ting K., et al. Inhibition of Cardiomyocytes differentiation of mouse embryonic stem cells by CD38/cADPR/Ca2+ signaling pathway. Journal of Biological Chemistry . 2012;287(42):35599–35611. doi: 10.1074/jbc.M112.392530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Sun C., Wang K., Stock A. J., et al. Re-equilibration of imbalanced NAD metabolism ameliorates the impact of telomere dysfunction. The EMBO Journal . 2020;39(21, article e103420) doi: 10.15252/embj.2019103420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Higashida H., Liang M., Yoshihara T., et al. An immunohistochemical, enzymatic, and behavioral study of CD157/BST-1 as a neuroregulator. BMC Neuroscience . 2017;18(1):p. 35. doi: 10.1186/s12868-017-0350-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Li J., Li H., Cai S., Bai S., Cai H., Zhang X. CD157 in bone marrow mesenchymal stem cells mediates mitochondrial production and transfer to improve neuronal apoptosis and functional recovery after spinal cord injury. Stem Cell Research & Therapy . 2021;12(1):p. 289. doi: 10.1186/s13287-021-02305-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Wakabayashi T., Naito H., Iba T., Nishida K., Takakura N. Identification of CD157-positive vascular endothelial stem cells in mouse retinal and choroidal vessels: fluorescence-activated cell sorting analysis. Investigative Ophthalmology & Visual Science . 2022;63(4):p. 5. doi: 10.1167/iovs.63.4.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Naito H., Wakabayashi T., Ishida M., et al. Isolation of tissue-resident vascular endothelial stem cells from mouse liver. Nature Protocols . 2020;15(3):1066–1081. doi: 10.1038/s41596-019-0276-x. [DOI] [PubMed] [Google Scholar]
  • 100.Wakabayashi T., Naito H., Suehiro J. I., et al. CD157 marks tissue-resident endothelial stem cells with homeostatic and regenerative properties. Cell Stem Cell . 2018;22(3):384–397.e6. doi: 10.1016/j.stem.2018.01.010. [DOI] [PubMed] [Google Scholar]
  • 101.Summers D. W., Gibson D. A., Diantonio A., Milbrandt J. SARM1-specific motifs in the TIR domain enable NAD(+) loss and regulate injury-induced SARM1 activation. Proceedings of the National Academy of Sciences of the United States of America . 2016;113(41):E6271–E6280. doi: 10.1073/pnas.1601506113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Doran C., Sugisawa R., Carty M., et al. CRISPR/Cas9-mediated SARM1 knockout and epitope-tagged mice reveal that SARM1 does not regulate nuclear transcription, but is expressed in macrophages. The Journal of Biological Chemistry . 2021;297(6, article 101417) doi: 10.1016/j.jbc.2021.101417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Carty M., Kearney J., Shanahan K. A., et al. Cell survival and cytokine release after inflammasome activation is regulated by the Toll-IL-1R protein SARM. Immunity . 2019;50(6):1412–1424.e6. doi: 10.1016/j.immuni.2019.04.005. [DOI] [PubMed] [Google Scholar]
  • 104.Essuman K., Summers D. W., Sasaki Y., Mao X., DiAntonio A., Milbrandt J. The SARM1 Toll/interleukin-1 receptor domain possesses intrinsic NAD+ cleavage activity that promotes pathological axonal degeneration. Neuron . 2017;93(6):1334–1343.e5. doi: 10.1016/j.neuron.2017.02.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Chen Y. H., Sasaki Y., DiAntonio A., Milbrandt J. SARM1 is required in human derived sensory neurons for injury-induced and neurotoxic axon degeneration. Experimental Neurology . 2021;339, article 113636 doi: 10.1016/j.expneurol.2021.113636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Lu Q., Botchway B. O. A., Zhang Y., Jin T., Liu X. SARM1 can be a potential therapeutic target for spinal cord injury. Cellular and molecular life sciences : CMLS . 2022;79(3):p. 161. doi: 10.1007/s00018-022-04195-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Sasaki Y., Zhu J., Shi Y., et al. Nicotinic acid mononucleotide is an allosteric SARM1 inhibitor promoting axonal protection. Experimental Neurology . 2021;345, article 113842 doi: 10.1016/j.expneurol.2021.113842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Bosanac T., Hughes R. O., Engber T., et al. Pharmacological SARM1 inhibition protects axon structure and function in paclitaxel-induced peripheral neuropathy. Brain . 2021;144(10):3226–3238. doi: 10.1093/brain/awab184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Tibbetts A. S., Appling D. R. Compartmentalization of mammalian folate-mediated one-carbon metabolism. Annual Review of Nutrition . 2010;30(1):57–81. doi: 10.1146/annurev.nutr.012809.104810. [DOI] [PubMed] [Google Scholar]
  • 110.Yue L., Pei Y., Zhong L., et al. Mthfd2 modulates mitochondrial function and DNA repair to maintain the pluripotency of mouse stem cells. Stem Cell Reports . 2020;15(2):529–545. doi: 10.1016/j.stemcr.2020.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Hoxhaj G., Ben-Sahra I., Lockwood S. E., et al. Direct stimulation of NADP+ synthesis through Akt-mediated phosphorylation of NAD kinase. Science . 2019;363(6431):1088–1092. doi: 10.1126/science.aau3903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Xu W., Li L., Zhang L. NAD+ metabolism as an emerging therapeutic target for cardiovascular diseases associated with sudden cardiac death. Frontiers in Physiology . 2020;11:p. 901. doi: 10.3389/fphys.2020.00901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Guarente L. Cell metabolism. The resurgence of NAD+ Science . 2016;352(6292):1396–1397. doi: 10.1126/science.aag1718. [DOI] [PubMed] [Google Scholar]
  • 114.Tarragó M. G., Chini C. C. S., Kanamori K. S., et al. A potent and specific CD38 inhibitor ameliorates age-related metabolic dysfunction by reversing tissue NAD+ decline. Cell Metabolism . 2018;27(5):1081–1095.e10. doi: 10.1016/j.cmet.2018.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Zha S., Li Z., Cao Q., Wang F., Liu F. PARP1 inhibitor (PJ34) improves the function of aging-induced endothelial progenitor cells by preserving intracellular NAD+ levels and increasing SIRT1 activity. Stem Cell Research & Therapy . 2018;9(1):p. 224. doi: 10.1186/s13287-018-0961-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Lucena-Cacace A., Otero-Albiol D., Jiménez-García M. P., Muñoz-Galvan S., Carnero A. NAMPT is a potent oncogene in colon cancer progression that modulates cancer stem cell properties and resistance to therapy through Sirt1 and PARP. Clinical Cancer Research : an official Journal of the American Association for Cancer Research . 2018;24(5):1202–1215. doi: 10.1158/1078-0432.CCR-17-2575. [DOI] [PubMed] [Google Scholar]
  • 117.Igarashi M., Miura M., Williams E., et al. NAD+ supplementation rejuvenates aged gut adult stem cells. Aging Cell . 2019;18(3, article e12935) doi: 10.1111/acel.12935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Schöndorf D. C., Ivanyuk D., Baden P., et al. The NAD+ Precursor Nicotinamide Riboside Rescues Mitochondrial Defects and Neuronal Loss in iPSC and Fly Models of Parkinson's Disease. Cell Reports . 2018;23(10):2976–2988. doi: 10.1016/j.celrep.2018.05.009. [DOI] [PubMed] [Google Scholar]
  • 119.Sun X., Cao B., Naval-Sanchez M., et al. Nicotinamide riboside attenuates age-associated metabolic and functional changes in hematopoietic stem cells. Nature Communications . 2021;12(1):p. 2665. doi: 10.1038/s41467-021-22863-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Zhang Y., Xu J., Ren Z., et al. Nicotinamide promotes pancreatic differentiation through the dual inhibition of CK1 and ROCK kinases in human embryonic stem cells. Stem Cell Research & Therapy . 2021;12(1):p. 362. doi: 10.1186/s13287-021-02426-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Song J., Li J., Yang F., et al. Nicotinamide mononucleotide promotes osteogenesis and reduces adipogenesis by regulating mesenchymal stromal cells via the SIRT1 pathway in aged bone marrow. Cell Death & Disease . 2019;10(5):p. 336. doi: 10.1038/s41419-019-1569-2. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Materials

Supplementary Figure S1: literature search method and selection criteria. Searches in Web of Science and PubMed were restricted to papers published between 2016 and 2022. The basic framework of the intervention program was determined by searching for a combination of subject terms and free words, such as “stem cell,” “NAD enzyme,” and “homeostasis.” Exclusion criteria for literature are as follows: no complete text accessible; publications written in a language other than English; and studies with inadequate literature evaluation.

Data Availability Statement

Our RNA-Seq data supporting this review have been deposited in the Sequence Read Archive (SRA) database at NCBI under the accession number SRP152900. The raw data are available (https://www.ncbi.nlm.nih.gov/sra/SRP152900).


Articles from Oxidative Medicine and Cellular Longevity are provided here courtesy of Wiley

RESOURCES