Abstract
NAD+ is a versatile biomolecule acting as a master regulator and substrate in various cellular processes, including redox regulation, metabolism, and various signaling pathways. In this article, we concisely and critically review the role of NAD+ in mechanisms promoting genome maintenance. Numerous NAD+-dependent reactions are involved in the preservation of genome stability, the cellular DNA damage response, and other pathways regulating nucleic acid metabolism, such as gene expression and cell proliferation pathways. Of note, NAD+ serves as a substrate to ADP-ribosyltransferases, sirtuins, and potentially also eukaryotic DNA ligases, all of which regulate various aspects of DNA integrity, damage repair, and gene expression. Finally, we critically analyze recent developments in the field as well as discuss challenges associated with therapeutic actions intended to raise NAD+ levels.
Keywords: NAD, DNA repair, PARPs, ARTs, sirtuins
Abbreviations: ADPR, ADP-ribose; ART, ADP-ribosyl transferase; ARTD, diphtheria toxin–like ADP-ribosyltransferase; ATM, ataxia–telangiectasia mutated; BER, base excision repair; CD, cluster of differentiation; CS, Cockayne syndrome; DSB, double-strand break; LIG, ligase; NA, nicotinic acid; NAAD, nicotinic acid adenine dinucleotide; NADK, NAD+ kinase; NAM, nicotinamide; NAMPT, nicotinamide phosphoribosyl transferase; NAR, nicotinic acid riboside; NHEJ, nonhomologous end-joining; NMN, nicotinamide mononucleotide; NMNAT, nicotinamide mononucleotide adenylyl transferase; NR, nicotinamide riboside; PAR, poly-ADP-ribose; PARP, poly-ADP-ribose polymerase; PARylation, poly(ADP-ribosyl)ation; ROS, reactive oxygen species; SIRT, sirtuin; Trp, tryptophan; XPA, xeroderma pigmentosum group A
DNA is constantly exposed to a plethora of damaging factors of both exogenous and endogenous origin, such as replication stress, alkylating and oxidative molecules, or UV radiation. To protect DNA from lesions caused by such stressors and to ensure genomic stability, cells have developed several pathways that recognize and repair specific DNA lesions (Fig. 1). Thus, in particular, small lesions are subject to direct reversal mediated by single proteins, for example, O6-alkylguanine-DNA alkyltransferase repairs O-alkylated DNA damage, and the alkB homolog dioxygenases reverse N-alkylated base adducts (1). Yet, the majority of repair mechanisms involves multiple events mediated by different proteins and orchestrated in a complex network of DNA repair pathways: base excision repair (BER) targets small lesions like oxidized bases and apuric/apyrimidic sites, in a mechanism overlapping with single-strand break repair. Larger nucleotide adducts are removed by nucleotide excision repair, and mismatch repair targets errors produced during DNA replication and recombination. Double-strand breaks (DSBs) are repaired either through homologous recombination or nonhomologous end-joining (NHEJ) comprising a canonical pathway (C-NHEJ) or alternative end-joining (A-EJ). As a comprehensive review of DNA repair pathways falls outside the scope of this article, we refer the reader to other in-depth review articles on this topic (2, 3). The diversity of active repair mechanisms ensures DNA stability, but when it is impaired, the persistence of genomic errors may lead to severe cellular consequences, including senescence and cell death (2). In the long term, inefficient DNA repair and compromised genome maintenance contribute to carcinogenesis and determines the adaptational capacity of cancer cells as well as aging and age-related diseases (4, 5, 6).
NAD+ fulfills multiple cellular roles, ranging from its function as a cofactor in energy production and redox regulation up to various signaling pathways essential for cell survival (7, 8). Interestingly, numerous NAD+-dependent reactions are involved in the preservation of genomic stability, cellular response to DNA damage, and other pathways concerning nucleic acids, such as gene expression or cell proliferation. Thus, the cellular NAD+ status influences genomic stability and sensitivity to DNA-damaging agents (9, 10, 11, 12, 13). Of note, NAD+ depletion in aging and age-related diseases implies supplementation of NAD+ boosting molecules as a plausible strategy in addressing age-dependent DNA damage (7, 14, 15).
Here, we first introduce the reader to the NAD+ biosynthesis and major cellular roles of this ubiquitous and versatile molecule. Furthermore, we turn our focus on the NAD+-dependent molecular mechanisms involved in mammalian genome maintenance. We concisely review both well-established concepts as well as new findings in the field. Finally, we discuss current controversies around NAD+-dependent therapeutic approaches that target various pathophysiological conditions in the light of its role in genome maintenance processes.
NAD+ biosynthesis
In mammals, NAD+ is synthesized from various precursors ingested through the diet, such as forms of vitamin B3: nicotinic acid (NA), nicotinamide (NAM), and nicotinamide riboside (NR); as well as the essential amino acid tryptophan (Trp), which is metabolized via three major pathways: (i) salvage pathway, (ii) de novo pathway, and (iii) Preiss–Handler pathway (Fig. 2). In addition, alternative precursors such as nicotinic acid riboside (NAR) (16), biosynthesis intermediates, such as nicotinamide mononucleotide (NMN) (17), or products of NAD+ consumption (NAM, NMN, and NR) are salvaged to replenish cellular NAD+ pools. In order to supply cells with “fresh” exogenous precursors, those can enter the cell during a passive process (e.g., NAM) or via membrane transporters—members of the solute carrier transporter family (for Trp and NA) or equilibrative nucleoside transporter (for NR and NAR). Furthermore, exogenous NMN can be taken up by cells, yet the cellular transport of this precursor molecule remains unclear (18, 19), with some studies suggesting that NMN uptake requires the prior conversion to NR by glycohydrolase, cluster of differentiation 73 (CD73) (20, 21).
The salvage pathway is a primary source of NAD+ in mammals (22) (Fig. 2). The first and rate-limiting step in this pathway—conversion of NAM to NMN, catalyzed by nicotinamide phosphoribosyl transferase (NAMPT)—is crucial in cellular NAD+ synthesis because numerous NAD+-consuming reactions release NAM. The enzyme NAMPT is present both extracellularly (eNAMPT) and intracellularly (iNAMPT) in the cytoplasm and nucleus, whereas its mitochondrial presence is disputed (23). Besides, NMN can be produced during ATP-dependent phosphorylation of NR by nicotinamide riboside kinase (17), and cleavage of the NR glycosidic bond by purine nucleoside phosphorylase produces NAM. Moreover, nicotinamide riboside kinase and purine nucleoside phosphorylase convert NAR to NAMN and NA, respectively, which feed the Preiss–Handler pathway (see later) (24). In the second (and final) step of the salvage pathway, NMN is converted to NAD+ by ATP-dependent nicotinamide mononucleotide adenylyl transferase (NMNAT). This enzyme in human tissues exists in three isoforms: the most ubiquitous nuclear NMNAT1, cytoplasmic NMNAT2 (25), and mitochondrial NMNAT3, whose function remains controversial (26, 27, 28). In addition, NMNAT catalyzes the formation of nicotinic acid adenine dinucleotide (NAAD) from NAMN, which comes from either Preiss–Handler or de novo pathways. In the Preiss–Handler pathway, NAMN is produced from NA by nicotinic acid phosphoribosyl transferase (29). In the de novo pathway, Trp is first transformed to quinolinic acid in the multistep process during the kynurenine pathway, and then quinolinic acid is converted to NAMN by quinolinic acid phosphoribosyl transferase. While originally thought to play a minor role in mammalian NAD+ synthesis, it has been recently suggested that the de novo pathway may play an important role in NAD+ homeostasis in humans (30, 31). In the final step of both Preiss–Handler or de novo pathways, NAAD is amidated to NAD+ by the cytosolic enzyme NAD+ synthase, which requires ATP, and ammonia or l-glutamine (32).
Cellular NAD+ levels are maintained in the range of 100 μM in the cytoplasm and nucleus and 200 μM in mitochondria (8). Such differences in subcellular NAD+ pools reflect its biological functions (see later) and are variable in tissues with different metabolic roles. Similarities in nuclear and cytoplasmic levels are likely because of the passive diffusion of NAD+ and its precursors through nuclear pores (33), although the detailed relationships between nuclear and cytoplasmic pools appear to be complex, since depletion of NAD+ in one compartment cannot be fully compensated by the other pool (28). As NAD+ has an overall negative charge, it is unable to passively cross lipid bilayers in mitochondria and other cellular membranes. Therefore, for a long time, the cytoplasmic/nuclear NAD+ pool has been thought to be strictly separated from the mitochondrial one, and it remained rather obscure how mitochondria build up their significant intraorganelle NAD+ levels. However, recently, this issue had been at least partially resolved. Thus, in 2020, a series of studies demonstrated the existence of a mitochondrial NAD+ carrier, that is, solute carrier transporter family SLC25A51 (or MCART1), providing mechanistic insight how mitochondria are supplied with NAD+ (28, 34, 35, 36).
The extracellular NAD+ presence, around 0.1 μM in mammals (37), is a result of either passive release from damaged cells (38) or active transport, for example, via connexin hemichannels like connexin 43 (39, 40). In turn, extracellular NAD+ can be degraded to precursors that can be taken up into the cell (41). Depending on the individual tissue, NAD+ molecules have a quite variable half-life that varies from 15 min to 15 h (42, 43, 44). Therefore, in tissues with high fluxes such as the small intestine and spleen, quick and efficient biosynthesis is necessary to compensate for a rapid cellular consumption via numerous NAD+-dependent reactions.
Cellular roles of NAD+
Numerous biochemical reactions require NAD+ as a cosubstrate (Fig. 2). NAD+ and its reduced form (NADH) are critical regulators of cellular bioenergetics, serving as cofactors in reactions of glucose metabolism, fatty acid β-oxidation, or the tricarboxylic acid cycle. For example, NAD+ is reduced to NADH during glycolysis by GAPDH. In turn, NADH is oxidized by lactate dehydrogenase, when pyruvate is converted to lactate under anaerobic conditions. NADH is a cofactor for fatty acid desaturases, and NAD+ is essential for alcohol dehydrogenase. NAD+/NADH-dependent malate dehydrogenase (malate–aspartate shuttle) and glycerol-3-phosphate dehydrogenase (glycerol-3-phosphate shuttle) generate reducing equivalents transported across the membrane into the mitochondria. There, NAD+ is reduced by pyruvate dehydrogenase complex and the tricarboxylic acid cycle enzymes: malate dehydrogenase, α-ketoglutarate dehydrogenase, and isocitrate dehydrogenase. NAD+ is also reduced by hydroxyacyl-CoA dehydrogenase in β-oxidation during fatty acid metabolism. Subsequently generated NADH is the major reducing factor in complex I mitochondrial electron transport chain, for the transfer of electrons in oxidative phosphorylation that produces ATP. Thus, the NAD+/NADH abundance reflects the energy status of the cell (7, 45).
ATP-dependent NAD+ kinases (NADKs) in the cytosol (cNADK) and mitochondria (mNADK) phosphorylate NAD+ to NADP+, which together with its reduced form NADPH, is critical for the maintenance of cellular redox homeostasis. It regenerates molecules responsible for xenobiotic detoxification, for example, cytochrome P450, and eradication of reactive oxygen species (ROS), such as GSH, thioredoxin, or peroxiredoxin (46). NADP+ is also a cosubstrate for glucose-6-phosphate dehydrogenase, a key enzyme in the pentose phosphate pathway, which produces precursors for the synthesis of nucleotides and aromatic amino acids. Moreover, it contributes to the synthesis of fatty acids and nicotinic acid adenine dinucleotide phosphate, which serves as a second messenger for intracellular calcium (Ca2+) signaling (46, 47). Because of its reducing role, NADP+ is found predominantly in the reduced state, with NADP+/NADPH ratios 1:200, which contrasts the NAD+/NADH ratio of 1000:1 in the cytoplasm and of 10:1 in the mitochondria (48).
In addition to the numerous reactions where NAD(P)+/NAD(P)H couples serve as an electron carrier, NAD+ can be also irreversibly consumed (Fig. 3). ADP-ribosyl transferases (ARTs) cleave the N-glycosidic bond, which releases NAM and ADP-ribose (ADPR) and further covalently attach the latter to the target molecule. Cholera toxin–like ADP-ribosyltransferases are localized often in the extracellular space and catalyze the addition of a single ADPR unit, called mono-ADP-ribosylation. Diphtheria toxin–like ADP-ribosyltransferases (ARTDs; also called poly-ADP-ribose polymerases [PARPs]) come in three flavors: (i) without any known enzymatic activity, (ii) as mono-ARTs catalyzing mono-ADP-ribosylation, or (iii) as poly-ARTs, catalyzing the attachment of numerous ADPR moieties, forming poly-ADP-ribose (PAR) chains in the process called poly(ADP-ribosyl)ation (PARylation). ADP-ribosylation is a post-translational modification that serves multiple cellular functions, such as signal transduction, energy metabolism, intracellular trafficking, or cell death (49, 50, 51). Yet, one of its major roles is the maintenance of nuclear homeostasis, regulation of DNA repair mechanism, chromatin remodeling, and transcription (52).
NAD+ is also consumed by histone deacetylases known as sirtuins (SIRTs). They use NAD+ as an acceptor during the transfer of acetyl (Ac) and other groups from target protein, which is a common post-translational modification, implemented in numerous cellular pathways, including those involved in genomic maintenance (53, 54). Another family of NAD+ consuming enzymes are glycohydrolases (NADases), such as CD73, CD38, or sterile α and TIR motif–containing 1. Apart from generating ADPR, this enzyme exhibits ADP-ribosyl cyclase activity, which produces cyclic ADPR, a second messenger in Ca2+ signaling. For instance, CD38 is a major extracellular NAD+ consumer, and its activity has a wide range of implications in the context of infection, metabolic dysfunction, aging, or tumor biology (55, 56). For more information regarding NAD+ multiple cellular roles, we refer the reader to recent reviews (7, 43, 45). In further parts of this review, we focus exclusively on NAD+-dependent pathways involved in the maintenance of genome integrity.
NAD+-dependent processes in genome maintenance
There is now a growing body of evidence that NAD+-dependent processes are heavily involved in genome maintenance and DNA repair mechanisms (57). This is exemplified in a study by Kiss et al. (58), demonstrating that CRISPR–Cas9-generated NMNAT1 U2OS knockout cells show reduced intracellular NAD+ levels, which was accompanied by hypersensitivity toward DNA-damaging cisplatin treatment. Furthermore, this study revealed that genetic inactivation of NMNAT1 completely blocked PARP1 activation in the nucleus, leading to increased γ-H2A.X DNA damage foci formation (58). In addition to PARP1, NAD+ also fuels the enzymatic activities of other PARPs involved in genome maintenance, such as PARP2, PARP3, TNKS1 (tankyrase 1), and TNKS2 (tankyrase 2). Those PARPs share NAD+ as a substrate with the SIRTs, of which some family members, such as SIRT1, SIRT6, and SIRT7, are directly involved in genome maintenance (59). Because of a functional and physical interplay between NAD+, PARPs, and SIRTs, the existence of a PARP–NAD–SIRT axis has been proposed that actively supports DNA repair mechanisms (57). In addition, DNA ligases (LIGs) have been suggested as a third class of enzymes that may use NAD+ as a substrate to support DNA repair (60). In the following paragraphs, we summarize and discuss the current state of the art on the role of NAD+ in functions of these enzymes during genome maintenance.
ARTDs (aka PARPs)
ARTDs (aka PARPs) are the major consumers of cellular NAD+. ARTDs use NAD+ to synthesize a monomer or polymer of ADPR (MAR or PAR, respectively) covalently attached to the target molecule. Thereby, ARTDs modify protein residues (Ser, Asp, Glu, Arg, and Lys) or terminal phosphates at DNA breaks (61, 62). During the process, the N-glycosidic bond of NAD+ is cleaved and NAM is released. The elongation of PAR chains involves the formation of the glycosidic bond between two riboses: 2′–1′′ for linear chains and 1′–2′′ for branched molecules and can involve up to 200 ADPR units (Fig. 3A). Although PARylation is reversible, NAD+ is not restored—PAR chains are degraded by poly(ADP-ribose) glycohydrolase to free PAR and/or ADPR molecules because of the cleavage of O-glycosidic bond. The terminal protein-ADPR moiety is cleaved by ADP-ribosyl hydrolases, the macrodomain containing proteins MacroD1/2, or the terminal ADP-ribose protein glycohydrolase 1 (TARG1) (63). The highly dynamic process of PARylation serves multiple cellular roles, including regulation of cellular stress response, survival, energy metabolism, inflammation, host–virus interactions, and many more, as reviewed elsewhere (49, 50). Furthermore, major roles of ARTDs are detection of DNA damage and facilitation of DNA repair and other nuclear mechanism maintaining genomic integrity (52, 64).
The first member identified within the ARTD family, that is, PARP1, accounts for the majority of PARylation activity, both constitutively and upon hyperactivation during DNA damage, when it consumes up to 80% of the nuclear NAD+ pool (65, 66, 67). PARP1 acts as a DNA nick sensor, which recognizes, binds to, and becomes activated by DNA strand breaks, and catalyzes immediate PAR synthesis, including autoPARylation (52, 64, 68, 69, 70). That serves as a scaffold for the recruitment of factors involved in different DNA repair mechanism, such as single-strand break repair /BER (e.g., X-ray repair cross-complementing protein 1 [XRCC1]) (71), homologous recombination (e.g., breast cancer type 1 susceptibility protein [BRCA1]), (72) or NHEJ (e.g., DNA-dependent protein kinase catalytic subunit, DNA-PKcs) (73) (more examples can be found in Table 1). PARP1 is recruited to stalled replication forks and helps to stabilize those during DNA damage repair (74, 75). PARP1-mediated PARylation of histone tails results in chromatin relaxation and recruitment of chromatin remodeling proteins, such as SMARCA5 (SWI/SNF related, matrix associated, actin-dependent regulator of chromatin, subfamily A, member 5) facilitating DNA repair (76). Moreover, such chromatin modifications may alter transcription. Further, PARP1 can act as a multitasking transcriptional regulator by binding to promoter sequence of transcription factors (e.g., NF-κB) or to transcription enhancers (e.g., cAMP response element–binding protein) (77, 78). PARP1 activity inhibits the negative elongation factor (NELF), thereby facilitating transcript elongation (79). Other ARTDs family members, particularly PARP2, PARP3, TNKS1, and TNKS2 exhibit partly overlapping as wells as non-redundant functions with PARP1 (Table 1). For a detailed discussion, we refer the reader to previous reviews focusing on the role of ARTDs in DNA damage repair (51, 52, 80, 81, 82, 83), chromatin remodeling (84) and transcription (85, 86). Moreover, PARylation have been also shown to regulate numerous steps in mechanisms maintaining RNA stability, processing, and translation (84, 87, 88, 89), as well as mitotic events and telomere integrity (90).
Table 1.
Class of enzymes | Specific NAD+ consuming members followed by target proteins |
---|---|
ARTDs |
|
SIRTs | |
NAD+-dependent LIG | LIG IV (60) |
Of importance, PARP1 overactivation is believed to determine the fate of the cell following DNA damage (91), which has been linked to patho-mechanisms of numerous disorders and cancer development (92). If DNA damage is low, PARP1 activation contributes to cell survival by mobilizing the DNA repair machinery. But if the damage is extensive, hyperactivation of PARP may cause severe depletion of cellular NAD+ levels. ATP-dependent NAD+ re-synthesis can lead to an energy crisis and metabolic shifts (e.g., impaired glycolysis, stimulation of pentose phosphate pathway) which cause cellular damage and subsequent cell death via necrosis (93). Furthermore, PARP1 regulates p53 activity (94), which initiates caspase-dependent apoptosis, although in the later phase nuclear caspases cleave PARP1, thereby inactivating it and preventing the decline of ATP, which is required for the execution of apoptosis (49, 95). Besides, PAR initiates caspase-independent apoptosis known as parthanatos. Fragments of PAR, released through poly(ADP-ribose) glycohydrolase activity, act as signaling molecules that can be distributed to subcellular compartments and attached to various targets. Binding of PAR fragments to mitochondrial apoptosis-inducing factor (AIF) promotes its nuclear translocation, initiation of large-scale DNA fragmentation, and subsequently cell death (49, 96). Interestingly, knock-in mice expressing the a PARP1 variant (PARP1D993A) resulting in hypo-PARylation activity and the formation of hypobranched PAR, revealed compromised genotoxic stress response during replication and led to a shift of the cell fate of mouse embryonic fibroblasts after genotoxic treatment away from DNA repair towards cell death and senescence (97). By these mechanisms, activation of PARP1 after DNA damage, may serve as a molecular switch to determine the post-damage cell fate in terms of DNA repair, cell death, or senescence.
There is room for speculation that besides the quality and quantity of the genotoxic stress, the local and systemic availability of NAD+ contributes to such PARylation dependent cell fate decision making. What speaks in favor for such a hypothesis is that PARP1 activity is tightly regulated by the availability and accessibility of NAD+. The Michaelis-Menten constant Km (NAD+) for major PARPs are similar to or higher than the concentration of free NAD+, which indicates that NAD+ levels in different subcellular compartments can significantly affect the activity of PARP1 and other PARP family members (98). Under normal conditions, PARP1 has limited NAD+ binding ability that accounts for the basal (unstimulated) PARP1 activity, but upon DNA damage, PARP1 interaction with DNA strand breaks leads to allosteric changes that allow full NAD+ access to its catalytic site and promotes activation of the enzyme (99). Interestingly, PARP1 activity also stimulates NAD+ synthesis, i.e., autoPARylated PARP1 has been shown to bind to NMNAT1, which may provide NAD+ to support PARP1 catalytic activity, but also stimulate PARP1 independently of NAD+ synthesis (100). Thereby, the interplay between PARP1 and NMNAT1 depends on the phosphorylation state of NMNAT1, which is subject to phosphorylation by protein kinase C (101). Moreover, in response to DNA damage, PARylation has been shown to stimulate the expression of salvage pathway enzymes NAMPT and NMNAT1 (102). Besides, a negative feedback mechanism that prevents excessive depletion of NAD+ by PARP1 upon DNA damage has been observed – decreased NAD+ levels promoted the binding of PARP1 to DBC1 (deleted in breast cancer 1), a potential tumor suppressor, which inhibits PARP1 activity and leads to DNA damage accumulation (103). Moreover, the NAD+–PARP1 interplay fosters crosstalk between subcellular compartments. Nuclear PARP1-dependent NAD+ depletion may impact NAD+ metabolism in distant regions of the cell (104). On the other hand, mitochondrial (105) and extracellular (21) pools of NAD+ have been demonstrated to influence nuclear PARP1 activation and DNA repair. Such observations indicate a complex, multilayer NAD+-PARP1 interaction. Taken together, the NAD+-dependent process of PARylation has been well established as a critical regulator of DNA damage repair factors, triggering chromatin structural changes, modulating the recruitment of transcription factors, modulating post-transcriptionally the stability of mRNAs, or interfering with mitotic events.
SIRTs
SIRTs (SITRs) are NAD+-dependent deacetylases associated with longevity, aging, and age-related pathologies (106, 107). SIRTs catalyze the removal of an acetyl group from protein lysine (Lys) residues, using NAD+ as an acetyl acceptor. During the process cleavage of NAD+ N-glycosidic bond releases NAM and the ADPR moiety is covalently attached to the acetyl group on the Lys. Then, the cleavage between acetyl and amino group produces 2-O-acetyl-ADPR (OAADPR) and deacetylated protein (Fig. 3B) (8, 108). In mammals, seven homologs are distributed within the cell in the nucleus (SIRT1, 6, 7), cytoplasm (SIRT2), and mitochondria (SIRT3, 4, 5) (109), and the shuttle between cellular compartments has been observed in response to pathological conditions (110, 111). While deacetylation accounts for the main activity, some SIRT family members have been shown to catalyze ADP-ribosylation or removal of different acyl groups, such as malonyl, glutaryl, succinyl, propionyl, myristoyl, and palmitoyl (112, 113). Owing to their wide involvement in post-translational modification, SIRTs are recognized as regulators of diverse biological processes, including energy metabolism (114, 115), inflammation (116), circadian rhythm control (117), redox homeostasis (118), autophagy (119), apoptosis (120), as well as genomic stability (53, 54), and tumorigenesis (108).
Analogously to PARPs, nuclear SIRTs contribute to genome integrity and DNA repair at numerous levels (Table 1). SIRT1 (121, 122), SIRT6 (123), and SIRT7 (124) deacetylate histones leading to the formation of higher-order chromatin compaction, which decreases accessibility for transcription factors. SIRT1 has been shown to directly deacetylate transcription factors, including p53 (125), NF-κB (126), or cAMP response element–binding protein (127), and has been implicated in other aspects of DNA integrity, including replication (128), recombination, DNA damage repair, and tumorigenesis (54, 129). Likewise, SIRT6 (130, 131, 132, 133) and SIRT7 (134) also regulate proteins involved in DNA damage repair. For example, SIRT7 is recruited to DSBs in a PARP1-dependent manner and catalyze H3K122 desuccinylation causing chromatin condensation facilitating damage repair (135). SIRT7-mediated H3 deacetylation affects the accumulation of the DNA damage response factor 53BP1 (134), and direct deacetylation of ataxia–telangiectasia mutated (ATM) kinase promotes activation of various components of DNA damage response (136). SIRT7 has been also implicated in the regulation of ribosome biogenesis and protein synthesis (137) as well as RNA stability (138, 139). SIRT6 association with H3 telomeres has been shown to facilitate the stability of telomeric chromatin (140). Albeit non-nuclear SIRTs have been involved predominantly in the regulation of metabolic homeostasis (107, 141), evidence on their role in genome maintenance is growing. SIRT2 shuttle between cytoplasm and nuclei under pathological and stress conditions has been implicated in the regulation of transcription (142), chromatin condensation during mitosis (111), and DNA damage response (143, 144, 145). SIRT3-mediated deacetylation of Ku70 (146) or RAD52 (147) points to its role in DSBR, whereas modification of 8-oxoguanine-DNA glycosylase 1 (OGG1) facilitates the excision of 7,8-dihydro-8-oxoguanine (8-oxoG) from damaged mitochondrial DNA (148). Overall, the broad and complex involvement of SIRT family members in genomic maintenance is well established and has been a subject on recent reviews (53, 54, 59, 149, 150).
Like PARPs, the activities of SIRTs are tightly regulated by NAD+ availability (151, 152), and age-associated decline in NAD+ levels and subsequent decrease in SIRT activity have been implicated in the pathogenesis of numerous aging-related diseases (107, 114). Targeting NAD+ biosynthesis enzymes such as NAMPT or NMNAT modulate SIRT activity (153, 154). Thus, increased dosage of NAMPT enhanced the transcriptional regulatory activity of the catalytic domain of SIRT2 recruited onto a reporter gene in mouse fibroblasts (153). Furthermore, NMNAT-1 has been shown to interact with SIRT1 in MCF-7 breast cancer cells and was recruited to target gene promoters by SIRT1 to support its deacetylase activity and transcriptional regulation (154). Experiments in mice suggested that modulation of NAD+ biosynthesis via the de novo synthesis pathway and the salvage synthesis pathway activated SIRT1 and prevented ototoxicity of the DNA damaging chemotherapeutic cisplatin (155). In turn, SIRTs affect NAD+ biosynthetic pathways to promote an increase in NAD+ levels (154, 156), albeit they consume a significantly lower amount of NAD+ than PARPs (8). NAMPT was identified as a direct substrate of SIRT6 deacetylation, with a mechanism that upregulates NAMPT enzymatic activity in the cancer cell lines BxPC-3 and MCF-7 (156). Interestingly, SIRT1, but not ARTDs, has been showed to maintain NAD+ homeostasis in conditions of excess NAD+ availability—SIRT1 silencing prevented intracellular NAD+ resetting under prolonged extracellular NAD+ exposure in HeLa cells (157).
In addition, a complex interplay between SIRTs and ARTDs occurs, since the enzymes compete for NAD+ as a substrate (158, 159). SIRT1 activity seems to be more affected by fluctuating NAD+ levels than PARP1 activity, whereas PARP1 displays a higher catalytic turnover of NAD+ than SIRT1 (158) that might lead in consequence to reduced SIRT1 activity during PARP1 overactivation following DNA damage (160). SIRT1 (161) and SIRT3 (162) deacetylate PARP1, which blocks its catalytic activity. Moreover, PARP1 and SIRT6 have been shown to bind to each other, which was potentiated by DNA damage (163). Besides crossmodifications, there is evidence on transcriptional coregulation—SIRT1 has been shown to negatively regulate the PARP1 promoter (164), whereas the SIRT1 promoter has been shown to be regulated by PARP2 (165). That adds an additional dimension to SIRT–ARTD crosstalk via coregulation of common pathways and targets in maintaining genomic integrity (133, 166).
NAD+-dependent DNA LIGs
In prokaryotes, DNA LIGs can catalyze the ATP/NAD+-dependent formation of phosphodiester bonds between adjacent DNA strands, required in the process of replication, recombination, and DNA repair (167). The reaction is initiated by an active site Lys, which attacks the α-phosphate moiety of either ATP or NAD+ to form a covalent enzyme–AMP intermediate, and the release of either pyrophosphate (from ATP), or NMN (from NAD+). Upon binding to nicked DNA, the AMP moiety is transferred from the Lys to the 5′-phosphate end. Subsequently, the hydroxyl group at 3′ end (3′-OH) attacks the adenylated 5′-phosphate (5′-PO4) to establish the new phosphodiester bond between the two ends, which releases AMP and finalizes the ligation (Fig. 3C) (168, 169). Mammalian cells have three genes encoding for DNA LIGs—LIGI, LIGIII, and LIGIV, which are essential for normal development and survival (170, 171, 172). Previously, they have been known to use only the ATP for the adenylation, unlike some archaeal LIGs, which utilize either ATP or NAD+ (169, 173). Of note, a recent study showed that human LIG IV, involved in the final step of NHEJ, indeed can use NAD+ as an alternative substrate for double-stranded DNA ligation (60). Although this finding has been doubted in a subsequent study (174) and requires further confirmation, it questions the exclusiveness of NAD+-dependent LIGs in prokaryote and points out a potential caveat in the concept to target them in a novel generation of antibiotics (175).
NAD+-dependent cellular homeostasis
Besides direct reactions mediated by NAD+-dependent enzymes, NAD+ affects genomic integrity through the maintenance of cellular homeostasis, such as energy production, redox balance, or Ca2+ levels. The efficiency of mechanisms involved in genomic stability depends greatly on energy. ATP is required for chromatin expansion, mediated by chromatin remodelers, for example, chromodomain helicase DNA-binding or switch/sucrose nonfermentable (SWI/SNF) (176, 177), which facilitates the recruitment of DNA repair proteins or gene expression. Numerous molecules involved in DNA damage signaling and repair are ATP dependent: DNA LIGs (178), kinases, for example, ATM, ataxia–telangiectasia and Rad3 related (ATR), DNA-PKcs (179, 180), or ATPases such as DNA-binding protein Rad50 (181). Likewise, numerous steps in genome expression and cell division are energy dependent (182, 183).
Disrupted redox balance and accumulation of ROS are major effectors in genotoxic stress. ROS induce DNA damage by oxidation of nucleobases, especially adenine and guanine (8-oxoA and 8-oxo-G), impair the activity of molecules regulating DNA damage response, DNA repair, and other nuclear processes, and influence the activity of transcription factors, modulating gene expression. Such a wide array of ROS-induced alterations affects genomic integrity, contribute to mutagenesis, and cell death (4, 184, 185, 186). An interesting example linking NAD+-dependent energy and redox homeostasis with genome maintenance is GAPDH. When oxidized by ROS, it acquires nonglycolytic functions and translocates to the nucleus, where it contributes to the maintenance of DNA integrity, transcriptional regulation, and tRNA export (187).
NAD+-derived cyclic ADPR regulates Ca2+ signaling (55, 56), which is another important factor in DNA integrity. Dysregulated Ca2+ homeostasis leads to mitochondrial accumulation of ROS and subsequently DNA damage (188). Ca2+ stimulates proteins involved in DNA damage repair (189, 190), mRNA stability (191), and mitosis (192, 193). Thus, dysregulation of NAD+-dependent homeostasis is often deleterious and linked to diseases and cancer (4, 194, 195).
NAD+ supplementation in maintaining genomic stability
Numerous studies confirmed that NAD+ status and its supplementation affect genomic stability, DNA repair, and sensitivity to cytotoxic effects of DNA-damaging agents or associated diseases. For instance, Kirkland et al. in a number of seminal articles have demonstrated that in rat bone marrow niacin is required for the maintenance of chromosomal stability and facilitates PARP-mediated DNA repair when challenged by the chemotherapy drugs etoposide (12) and ethylnitrosourea (13, 196, 197, 198, 199). These data suggest that niacin supplementation might be beneficial for cancer patients in decreasing the severity of the side effects of chemotherapy (200). NAM supplementation ameliorated the DNA damage induced by exposure of human lymphocytes to various genotoxins, UV irradiation, N-methyl-N′-nitro-N-nitroso guanidine, or dimethyl sulfate, ex vivo (201). Our group showed that NA supplementation increased DNA repair efficiency and enhanced genomic stability after X-ray exposure in human peripheral blood mononuclear cells ex vivo (10). NR, investigated extensively by Bohr et al., has shown numerous beneficial effects in aging-related and DNA repair–deficient disease models. Xeroderma pigmentosum group A (XPA) is a disorder associated with decreased ability to repair DNA via nucleotide excision repair. NR treatment ameliorated numerous endpoints in mitochondrial dysfunction in XPA protein–deficient cells and rescued the life span defect in XPA-1–deficient Caenorhabditis elegans (202). Analogously, NR treatment stimulated neuronal DNA repair and improved mitochondrial quality in animal models of ataxia–telangiectasia, a disease linked to ATM deficiency. Moreover, NR supplementation reduced the neuropathological and neurobehavioral disease outcomes and extends the animal life span (203). Cockayne syndrome (CS) is a segmental accelerated aging disorder characterized by progressive neurodegeneration caused by a deficiency of DNA repair proteins CS group A or B. In CS models, because of deficient DNA repair, PARP overactivation leads to decreased cellular NAD+ levels and subsequently decreased SIRT1 activity and mitochondrial dysfunction. One-week-long NR treatment reduced CS-associated increased mitochondrial membrane potential or ROS production and normalized the cerebellar transcriptome involved in the mitochondrial pathways, oxidative stress, transcription, DNA repair, DNA damage response, and histone acetylation in CS mice (204). In a mouse model of Alzheimer's disease (3xTgAD mice), which in addition lack the BER enzyme DNA polymerase β, an exacerbation of major Alzheimer's disease features was ameliorated by NR supplementation, such as DNA damage, neuroinflammation, neuronal apoptosis, decreased SIRT3 activity, increased PARP1 activity, but NR also improved cognitive function in multiple behavioral tests and restored hippocampal synaptic plasticity (205). Moreover, short-term NR supplementation prevented the hearing loss in CS mice (206). NR restored mitochondrial quality via mitophagy and delayed accelerated aging in C. elegans and Drosophila melanogaster models of Werner syndrome, a disease caused by mutations in the gene encoding the Werner syndrome (WRN) DNA helicase (207). Furthermore, NR treatment attenuated age-associated and functional changes in hematopoietic stem cells in mice, which was accompanied by significant transcriptional changes of genes involved in DNA damage response and DNA repair (208). Such well and extensively documented benefits of NAD+ supplements, particularly NR, imply the universal therapeutic application of these compounds in treating diseases associated with impaired genomic stability. However, because of the complex and still not completely understood cellular function of NAD+, the translation to humans remains challenging.
Yet, what has been shown in humans by now is that supplementation with NR ameliorated telomere damage, and several other endpoints of the pathophysiology of dyskeratosis congenita evaluated in primary fibroblasts from dyskeratosis congenita patients (209). Moreover, phase 2 and 3 clinical studies in high-risk patients receiving 500 mg of NAM twice daily for up to 12 months significantly reduced the rates of nonmelanoma skin cancers and actinic keratoses by 10 to 30% compared with placebo control groups. These studies impressively indicate the chemopreventive potential of NAM treatment (210, 211). However, as discussed in the next section, several caveats must be taken into account, when considering the broader use of NAD+ boosting molecules as genoprotective agents in humans.
A double-faced role of NAD+ in genome maintenance
With a broad (and constantly growing) number of mechanisms relying on NAD+, maintaining its cellular pool emerges as critical to ensure genome integrity and expression. That becomes particularly important in the light of age-related NAD+ decline. Therefore, boosting NAD+ levels has been considered as a potential therapeutic or preventive factor in multiple pathological conditions (212). In laboratory conditions, supplementation with NAD+ precursors, such as NR and NMN, has been shown beneficial against aging and age-related diseases (213, 214). Similar effects have been reached by approaches aiming to increase NAD+ levels and metabolism via caloric restriction, intermittent fasting, or exercise (7, 106). However, these promising preclinical studies are not easily translated to humans (215). The reason for that might be the complexity of the cellular roles of NAD+. For example, the involvement of NAD+ in numerous redox reactions may be detrimental for cellular homeostasis when exposed to supraphysiologic NAD+ levels (98).
Another issue regarding supplementing NAD+ involves the risk of fueling cancer cells. Increased metabolic demands of rapidly growing cancer cells require more NAD+ (216). The proliferation-promoting shift toward lactate fermentation in the cytoplasm and reduced oxidative phosphorylation in mitochondria (i.e., the Warburg effect) (217) may impair the NAD+/NADH ratio. High NAD+ levels may lead to cancer promotion (218). Numerous genes, associated with NAD+ biosynthesis and metabolism, have been identified as promoting tumor development, for example, NAMPT (219, 220), nicotinic acid phosphoribosyl transferase (221, 222), NADK (47), which makes them promising targets in cancer therapy. [NB: Yet, NAD+ supplementation has also been shown to reduce the incidence and size of tumors (223, 224).] Besides, NAD+-consuming enzymes, such as PARPs and SIRTs, play a complex role in cancer development. Under normal conditions, their beneficial role in the maintenance of genome integrity prevents the occurrence of cancer-driving mutations. But at conditions, when tumor already exists, their activity may promote cancer development and resistance to treatment. In such cases, enhancing NAD+ levels might have a tumor-promoting effect (14, 225).
Concluding remarks
Although our understanding of the role of NAD+ in nucleic acid maintenance has significantly expanded during recent years, the complexity of NAD+ cellular functions makes this topic of relevance in future studies. Many aspects of NAD+ metabolism remain unresolved, such as cell type–specific differences or subcellular compartmentalization. Topics, such as the role of NAD+ in mitochondrial DNA repair, or NAD+ capping, are yet poorly studied and deserve further exploration. Equally important is an understanding and evaluation of the risk–benefit ratio associated with elevating NAD+ levels, which may act as a double-edged sword in the context of maintaining DNA integrity and overall NAD+-dependent cellular homeostasis. It will be exciting to see how future studies will provide a deeper understanding of these mechanisms.
Conflict of interest
The authors declare that they have no conflicts of interest with the contents of this article.
Acknowledgments
The experimental work in our group has been supported by grants from the German Ministry of Defense (grant numbers: E/U2AD/ID015/IF561 and E/U2CB/EA199/EF552) as well as the German Research Foundation (DFG, grant number: MA-4905/4-1).
Author contributions
J. A. R. and A. M. conceptualization; J. A. R. writing–original draft; J. A. R., A. B., and A. M. writing–review & editing; A. B. and A. M. funding acquisition.
Edited by Patrick Sung
Contributor Information
Joanna A. Ruszkiewicz, Email: joanna.ruszkiewicz@uni-konstanz.de.
Alexander Bürkle, Email: alexander.buerkle@uni-konstanz.de.
Aswin Mangerich, Email: aswin.mangerich@uni-konstanz.de.
References
- 1.Eker A.P., Quayle C., Chaves I., van der Horst G.T. DNA repair in mammalian cells: direct DNA damage reversal: elegant solutions for nasty problems. Cell Mol. Life Sci. 2009;66:968–980. doi: 10.1007/s00018-009-8735-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Jackson S.P., Bartek J. The DNA-damage response in human biology and disease. Nature. 2009;461:1071–1078. doi: 10.1038/nature08467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Scully R., Panday A., Elango R., Willis N.A. DNA double-strand break repair-pathway choice in somatic mammalian cells. Nat. Rev. Mol. Cell. Biol. 2019;20:698–714. doi: 10.1038/s41580-019-0152-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Srinivas U.S., Tan B.W.Q., Vellayappan B.A., Jeyasekharan A.D. ROS and the DNA damage response in cancer. Redox Biol. 2019;25:101084. doi: 10.1016/j.redox.2018.101084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Petr M.A., Tulika T., Carmona-Marin L.M., Scheibye-Knudsen M. Protecting the aging genome. Trends Cell Biol. 2020;30:117–132. doi: 10.1016/j.tcb.2019.12.001. [DOI] [PubMed] [Google Scholar]
- 6.Schumacher B., Pothof J., Vijg J., Hoeijmakers J.H.J. The central role of DNA damage in the ageing process. Nature. 2021;592:695–703. doi: 10.1038/s41586-021-03307-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Katsyuba E., Romani M., Hofer D., Auwerx J. NAD(+) homeostasis in health and disease. Nat. Metab. 2020;2:9–31. doi: 10.1038/s42255-019-0161-5. [DOI] [PubMed] [Google Scholar]
- 8.Kulkarni C.A., Brookes P.S. Cellular compartmentation and the redox/nonredox functions of NAD. Antioxid. Redox Signal. 2019;31:623–642. doi: 10.1089/ars.2018.7722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Durkacz B.W., Omidiji O., Gray D.A., Shall S. ADP-ribose)n participates in DNA excision repair. Nature. 1980;283:593–596. doi: 10.1038/283593a0. [DOI] [PubMed] [Google Scholar]
- 10.Weidele K., Beneke S., Burkle A. The NAD(+) precursor nicotinic acid improves genomic integrity in human peripheral blood mononuclear cells after X-irradiation. DNA Repair (Amst) 2017;52:12–23. doi: 10.1016/j.dnarep.2017.02.001. [DOI] [PubMed] [Google Scholar]
- 11.Weitberg A.B. Effect of nicotinic acid supplementation in vivo on oxygen radical-induced genetic damage in human lymphocytes. Mutat. Res. 1989;216:197–201. doi: 10.1016/0165-1161(89)90005-8. [DOI] [PubMed] [Google Scholar]
- 12.Spronck J.C., Kirkland J.B. Niacin deficiency increases spontaneous and etoposide-induced chromosomal instability in rat bone marrow cells in vivo. Mutat. Res. 2002;508:83–97. doi: 10.1016/s0027-5107(02)00188-4. [DOI] [PubMed] [Google Scholar]
- 13.Kostecki L.M., Thomas M., Linford G., Lizotte M., Toxopeus L., Bartleman A.P., et al. Niacin deficiency delays DNA excision repair and increases spontaneous and nitrosourea-induced chromosomal instability in rat bone marrow. Mutat. Res. 2007;625:50–61. doi: 10.1016/j.mrfmmm.2007.05.008. [DOI] [PubMed] [Google Scholar]
- 14.Sharif T., Martell E., Dai C., Ghassemi-Rad M.S., Kennedy B.E., Lee P.W.K., et al. Regulation of cancer and cancer-related genes via NAD. Antioxid. Redox Signal. 2019;30:906–923. doi: 10.1089/ars.2017.7478. [DOI] [PubMed] [Google Scholar]
- 15.Covarrubias A.J., Perrone R., Grozio A., Verdin E. NAD(+) metabolism and its roles in cellular processes during ageing. Nat. Rev. Mol. Cel. Biol. 2021;22:119–141. doi: 10.1038/s41580-020-00313-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kulikova V., Shabalin K., Nerinovski K., Dolle C., Niere M., Yakimov A., et al. Generation, release, and uptake of the NAD precursor nicotinic acid riboside by human cells. J. Biol. Chem. 2015;290:27124–27137. doi: 10.1074/jbc.M115.664458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ratajczak J., Joffraud M., Trammell S.A., Ras R., Canela N., Boutant M., et al. NRK1 controls nicotinamide mononucleotide and nicotinamide riboside metabolism in mammalian cells. Nat. Commun. 2016;7:13103. doi: 10.1038/ncomms13103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Grozio A., Mills K.F., Yoshino J., Bruzzone S., Sociali G., Tokizane K., et al. Slc12a8 is a nicotinamide mononucleotide transporter. Nat. Metab. 2019;1:47–57. doi: 10.1038/s42255-018-0009-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Schmidt M.S., Brenner C. Absence of evidence that Slc12a8 encodes a nicotinamide mononucleotide transporter. Nat. Metab. 2019;1:660–661. doi: 10.1038/s42255-019-0085-0. [DOI] [PubMed] [Google Scholar]
- 20.Kulikova V., Shabalin K., Nerinovski K., Yakimov A., Svetlova M., Solovjeva L., et al. Degradation of extracellular NAD(+) intermediates in cultures of human HEK293 cells. Metabolites. 2019;9:293. doi: 10.3390/metabo9120293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wilk A., Hayat F., Cunningham R., Li J., Garavaglia S., Zamani L., et al. Extracellular NAD(+) enhances PARP-dependent DNA repair capacity independently of CD73 activity. Sci. Rep. 2020;10:651. doi: 10.1038/s41598-020-57506-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Magni G., Amici A., Emanuelli M., Raffaelli N., Ruggieri S. Enzymology of NAD+ synthesis. Adv. Enzymol. Relat. Areas Mol. Biol. 1999;73:135–182. doi: 10.1002/9780470123195.ch5. [DOI] [PubMed] [Google Scholar]
- 23.Garten A., Schuster S., Penke M., Gorski T., de Giorgis T., Kiess W. Physiological and pathophysiological roles of NAMPT and NAD metabolism. Nat. Rev. Endocrinol. 2015;11:535–546. doi: 10.1038/nrendo.2015.117. [DOI] [PubMed] [Google Scholar]
- 24.Belenky P., Christensen K.C., Gazzaniga F., Pletnev A.A., Brenner C. Nicotinamide riboside and nicotinic acid riboside salvage in fungi and mammals. Quantitative basis for Urh1 and purine nucleoside phosphorylase function in NAD+ metabolism. J. Biol. Chem. 2009;284:158–164. doi: 10.1074/jbc.M807976200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Hicks A.N., Lorenzetti D., Gilley J., Lu B., Andersson K.E., Miligan C., et al. Nicotinamide mononucleotide adenylyltransferase 2 (Nmnat2) regulates axon integrity in the mouse embryo. PLoS One. 2012;7 doi: 10.1371/journal.pone.0047869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Berger F., Lau C., Dahlmann M., Ziegler M. Subcellular compartmentation and differential catalytic properties of the three human nicotinamide mononucleotide adenylyltransferase isoforms. J. Biol. Chem. 2005;280:36334–36341. doi: 10.1074/jbc.M508660200. [DOI] [PubMed] [Google Scholar]
- 27.Yamamoto M., Hikosaka K., Mahmood A., Tobe K., Shojaku H., Inohara H., et al. Nmnat3 is dispensable in mitochondrial NAD level maintenance in vivo. PLoS One. 2016;11 doi: 10.1371/journal.pone.0147037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ziegler M., Monné M., Nikiforov A., Agrimi G., Heiland I., Palmieri F. Welcome to the family: identification of the NAD+ transporter of animal mitochondria as member of the solute carrier family SLC25. Biomolecules. 2021;11:880. doi: 10.3390/biom11060880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Piacente F., Caffa I., Ravera S., Sociali G., Passalacqua M., Vellone V.G., et al. Nicotinic acid phosphoribosyltransferase regulates cancer cell metabolism, susceptibility to NAMPT inhibitors, and DNA repair. Cancer Res. 2017;77:3857–3869. doi: 10.1158/0008-5472.CAN-16-3079. [DOI] [PubMed] [Google Scholar]
- 30.Shi H., Enriquez A., Rapadas M., Martin E., Wang R., Moreau J., et al. NAD deficiency, congenital malformations, and niacin supplementation. New Engl. J. Med. 2017;377:544–552. doi: 10.1056/NEJMoa1616361. [DOI] [PubMed] [Google Scholar]
- 31.Poyan Mehr A., Tran M.T., Ralto K.M., Leaf D.E., Washco V., Messmer J., et al. De novo NAD(+) biosynthetic impairment in acute kidney injury in humans. Nat. Med. 2018;24:1351–1359. doi: 10.1038/s41591-018-0138-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hara N., Yamada K., Terashima M., Osago H., Shimoyama M., Tsuchiya M. Molecular identification of human glutamine- and ammonia-dependent NAD synthetases. Carbon-nitrogen hydrolase domain confers glutamine dependency. J. Biol. Chem. 2003;278:10914–10921. doi: 10.1074/jbc.M209203200. [DOI] [PubMed] [Google Scholar]
- 33.Cambronne X.A., Stewart M.L., Kim D., Jones-Brunette A.M., Morgan R.K., Farrens D.L., et al. Biosensor reveals multiple sources for mitochondrial NAD(+) Science. 2016;352:1474–1477. doi: 10.1126/science.aad5168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Girardi E., Agrimi G., Goldmann U., Fiume G., Lindinger S., Sedlyarov V., et al. Epistasis-driven identification of SLC25A51 as a regulator of human mitochondrial NAD import. Nat. Commun. 2020;11:6145. doi: 10.1038/s41467-020-19871-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kory N., Uit de Bos J., van der Rijt S., Jankovic N., Güra M., Arp N., et al. MCART1/SLC25A51 is required for mitochondrial NAD transport. Sci. Adv. 2020;6 doi: 10.1126/sciadv.abe5310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Luongo T.S., Eller J.M., Lu M.J., Niere M., Raith F., Perry C., et al. SLC25A51 is a mammalian mitochondrial NAD(+) transporter. Nature. 2020;588:174–179. doi: 10.1038/s41586-020-2741-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Davies C.A., Perrett D., Zhang Z., Nielsen B.R., Blake D.R., Winyard P.G. Simultaneous analysis of nitrite, nitrate and the nicotinamide nucleotides by capillary electrophoresis: application to biochemical studies and human extracellular fluids. Electrophoresis. 1999;20:2111–2117. doi: 10.1002/(SICI)1522-2683(19990701)20:10<2111::AID-ELPS2111>3.0.CO;2-5. [DOI] [PubMed] [Google Scholar]
- 38.Haag F., Adriouch S., Brass A., Jung C., Moller S., Scheuplein F., et al. Extracellular NAD and ATP: partners in immune cell modulation. Purinergic Signal. 2007;3:71–81. doi: 10.1007/s11302-006-9038-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Billington R.A., Travelli C., Ercolano E., Galli U., Roman C.B., Grolla A.A., et al. Characterization of NAD uptake in mammalian cells. J. Biol. Chem. 2008;283:6367–6374. doi: 10.1074/jbc.M706204200. [DOI] [PubMed] [Google Scholar]
- 40.Pillai V.B., Sundaresan N.R., Kim G., Gupta M., Rajamohan S.B., Pillai J.B., et al. Exogenous NAD blocks cardiac hypertrophic response via activation of the SIRT3-LKB1-AMP-activated kinase pathway. J. Biol. Chem. 2010;285:3133–3144. doi: 10.1074/jbc.M109.077271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Grozio A., Sociali G., Sturla L., Caffa I., Soncini D., Salis A., et al. CD73 protein as a source of extracellular precursors for sustained NAD+ biosynthesis in FK866-treated tumor cells. J. Biol. Chem. 2013;288:25938–25949. doi: 10.1074/jbc.M113.470435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Rechsteiner M., Hillyard D., Olivera B.M. Turnover at nicotinamide adenine dinucleotide in cultures of human cells. J. Cell. Physiol. 1976;88:207–217. doi: 10.1002/jcp.1040880210. [DOI] [PubMed] [Google Scholar]
- 43.Chini C.C.S., Zeidler J.D., Kashyap S., Warner G., Chini E.N. Evolving concepts in NAD(+) metabolism. Cell Metab. 2021;33:1076–1087. doi: 10.1016/j.cmet.2021.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Liu L., Su X., Quinn W.J., Hui S., Krukenberg K., Frederick D.W., et al. Quantitative analysis of NAD synthesis-breakdown fluxes. Cell Metab. 2018;27:1067–1080.e1065. doi: 10.1016/j.cmet.2018.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Xiao W., Wang R.S., Handy D.E., Loscalzo J. NAD(H) and NADP(H) redox couples and cellular energy metabolism. Antioxid. Redox Signal. 2018;28:251–272. doi: 10.1089/ars.2017.7216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Agledal L., Niere M., Ziegler M. The phosphate makes a difference: cellular functions of NADP. Redox Rep. 2010;15:2–10. doi: 10.1179/174329210X12650506623122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Tedeschi P.M., Bansal N., Kerrigan J.E., Abali E.E., Scotto K.W., Bertino J.R. NAD+ kinase as a therapeutic target in cancer. Clin. Cancer Res. 2016;22:5189–5195. doi: 10.1158/1078-0432.CCR-16-1129. [DOI] [PubMed] [Google Scholar]
- 48.Veech R.L., Eggleston L.V., Krebs H.A. The redox state of free nicotinamide-adenine dinucleotide phosphate in the cytoplasm of rat liver. Biochem. J. 1969;115:609–619. doi: 10.1042/bj1150609a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Aredia F., Scovassi A.I. Poly(ADP-ribose): a signaling molecule in different paradigms of cell death. Biochem. Pharmacol. 2014;92:157–163. doi: 10.1016/j.bcp.2014.06.021. [DOI] [PubMed] [Google Scholar]
- 50.Gupte R., Liu Z., Kraus W.L. PARPs and ADP-ribosylation: recent advances linking molecular functions to biological outcomes. Genes Dev. 2017;31:101–126. doi: 10.1101/gad.291518.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Reber J.M., Mangerich A. Why structure and chain length matter: on the biological significance underlying the structural heterogeneity of poly(ADP-ribose) Nucl. Acids Res. 2021;49:8432–8448. doi: 10.1093/nar/gkab618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Azarm K., Smith S. Nuclear PARPs and genome integrity. Genes Dev. 2020;34:285–301. doi: 10.1101/gad.334730.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Roos W.P., Krumm A. The multifaceted influence of histone deacetylases on DNA damage signalling and DNA repair. Nucl. Acids Res. 2016;44:10017–10030. doi: 10.1093/nar/gkw922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kosciuk T., Wang M., Hong J.Y., Lin H. Updates on the epigenetic roles of sirtuins. Curr. Opin. Chem. Biol. 2019;51:18–29. doi: 10.1016/j.cbpa.2019.01.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Aksoy P., White T.A., Thompson M., Chini E.N. Regulation of intracellular levels of NAD: a novel role for CD38. Biochem. Biophys. Res. Commun. 2006;345:1386–1392. doi: 10.1016/j.bbrc.2006.05.042. [DOI] [PubMed] [Google Scholar]
- 56.Lee H.C., Zhao Y.J. Resolving the topological enigma in Ca(2+) signaling by cyclic ADP-ribose and NAADP. J. Biol. Chem. 2019;294:19831–19843. doi: 10.1074/jbc.REV119.009635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Saville K.M., Clark J., Wilk A., Rogers G.D., Andrews J.F., Koczor C.A., et al. NAD(+)-mediated regulation of mammalian base excision repair. DNA Repair. 2020;93:102930. doi: 10.1016/j.dnarep.2020.102930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kiss A., Ráduly A.P., Regdon Z., Polgár Z., Tarapcsák S., Sturniolo I., et al. Targeting nuclear NAD(+) synthesis inhibits DNA repair, impairs metabolic adaptation and increases chemosensitivity of U-2OS osteosarcoma cells. Cancers (Basel) 2020;12:1180. doi: 10.3390/cancers12051180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lagunas-Rangel F.A. Current role of mammalian sirtuins in DNA repair. DNA Repair (Amst) 2019;80:85–92. doi: 10.1016/j.dnarep.2019.06.009. [DOI] [PubMed] [Google Scholar]
- 60.Chen S.H., Yu X. Human DNA ligase IV is able to use NAD+ as an alternative adenylation donor for DNA ends ligation. Nucl. Acids Res. 2019;47:1321–1334. doi: 10.1093/nar/gky1202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Zarkovic G., Belousova E.A., Talhaoui I., Saint-Pierre C., Kutuzov M.M., Matkarimov B.T., et al. Characterization of DNA ADP-ribosyltransferase activities of PARP2 and PARP3: new insights into DNA ADP-ribosylation. Nucl. Acids Res. 2018;46:2417–2431. doi: 10.1093/nar/gkx1318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Matta E., Kiribayeva A., Khassenov B., Matkarimov B.T., Ishchenko A.A. Insight into DNA substrate specificity of PARP1-catalysed DNA poly(ADP-ribosyl)ation. Sci. Rep. 2020;10:3699. doi: 10.1038/s41598-020-60631-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.O'Sullivan J., Tedim Ferreira M., Gagné J.P., Sharma A.K., Hendzel M.J., Masson J.Y., et al. Emerging roles of eraser enzymes in the dynamic control of protein ADP-ribosylation. Nat. Commun. 2019;10:1182. doi: 10.1038/s41467-019-08859-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Eisemann T., Pascal J.M. Poly(ADP-ribose) polymerase enzymes and the maintenance of genome integrity. Cell Mol. Life Sci. 2020;77:19–33. doi: 10.1007/s00018-019-03366-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Burkle A. Poly(ADP-ribose). The most elaborate metabolite of NAD+ FEBS J. 2005;272:4576–4589. doi: 10.1111/j.1742-4658.2005.04864.x. [DOI] [PubMed] [Google Scholar]
- 66.Yu S.W., Wang H., Poitras M.F., Coombs C., Bowers W.J., Federoff H.J., et al. Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor. Science. 2002;297:259–263. doi: 10.1126/science.1072221. [DOI] [PubMed] [Google Scholar]
- 67.Rank L., Veith S., Gwosch E.C., Demgenski J., Ganz M., Jongmans M.C., et al. Analyzing structure-function relationships of artificial and cancer-associated PARP1 variants by reconstituting TALEN-generated HeLa PARP1 knock-out cells. Nucl. Acids Res. 2016;44:10386–10405. doi: 10.1093/nar/gkw859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Krüger A., Bürkle A., Hauser K., Mangerich A. Real-time monitoring of PARP1-dependent PARylation by ATR-FTIR spectroscopy. Nat. Commun. 2020;11:2174. doi: 10.1038/s41467-020-15858-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Pascal J.M. The comings and goings of PARP-1 in response to DNA damage. DNA Repair (Amst) 2018;71:177–182. doi: 10.1016/j.dnarep.2018.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.de Murcia G., Ménissier de Murcia J. Poly(ADP-ribose) polymerase: a molecular nick-sensor. Trends Biochem. Sci. 1994;19:172–176. doi: 10.1016/0968-0004(94)90280-1. [DOI] [PubMed] [Google Scholar]
- 71.El-Khamisy S.F., Masutani M., Suzuki H., Caldecott K.W. A requirement for PARP-1 for the assembly or stability of XRCC1 nuclear foci at sites of oxidative DNA damage. Nucl. Acids Res. 2003;31:5526–5533. doi: 10.1093/nar/gkg761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Li M., Yu X. Function of BRCA1 in the DNA damage response is mediated by ADP-ribosylation. Cancer cell. 2013;23:693–704. doi: 10.1016/j.ccr.2013.03.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Rybanska I., Ishaq O., Chou J., Prakash M., Bakhsheshian J., Huso D.L., et al. PARP1 and DNA-PKcs synergize to suppress p53 mutation and telomere fusions during T-lineage lymphomagenesis. Oncogene. 2013;32:1761–1771. doi: 10.1038/onc.2012.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Haince J.F., McDonald D., Rodrigue A., Dery U., Masson J.Y., Hendzel M.J., et al. PARP1-dependent kinetics of recruitment of MRE11 and NBS1 proteins to multiple DNA damage sites. J. Biol. Chem. 2008;283:1197–1208. doi: 10.1074/jbc.M706734200. [DOI] [PubMed] [Google Scholar]
- 75.Berti M., Ray Chaudhuri A., Thangavel S., Gomathinayagam S., Kenig S., Vujanovic M., et al. Human RECQ1 promotes restart of replication forks reversed by DNA topoisomerase I inhibition. Nat. Struct. Mol. Biol. 2013;20:347–354. doi: 10.1038/nsmb.2501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Smeenk G., Wiegant W.W., Marteijn J.A., Luijsterburg M.S., Sroczynski N., Costelloe T., et al. Poly(ADP-ribosyl)ation links the chromatin remodeler SMARCA5/SNF2H to RNF168-dependent DNA damage signaling. J. Cel. Sci. 2013;126:889–903. doi: 10.1242/jcs.109413. [DOI] [PubMed] [Google Scholar]
- 77.Hassa P.O., Buerki C., Lombardi C., Imhof R., Hottiger M.O. Transcriptional coactivation of nuclear factor-kappaB-dependent gene expression by p300 is regulated by poly(ADP)-ribose polymerase-1. J. Biol. Chem. 2003;278:45145–45153. doi: 10.1074/jbc.M307957200. [DOI] [PubMed] [Google Scholar]
- 78.Hassa P.O., Haenni S.S., Buerki C., Meier N.I., Lane W.S., Owen H., et al. Acetylation of poly(ADP-ribose) polymerase-1 by p300/CREB-binding protein regulates coactivation of NF-kappaB-dependent transcription. J. Biol. Chem. 2005;280:40450–40464. doi: 10.1074/jbc.M507553200. [DOI] [PubMed] [Google Scholar]
- 79.Gibson B.A., Zhang Y., Jiang H., Hussey K.M., Shrimp J.H., Lin H., et al. Chemical genetic discovery of PARP targets reveals a role for PARP-1 in transcription elongation. Science. 2016;353:45–50. doi: 10.1126/science.aaf7865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Wang Y., Luo W., Wang Y. PARP-1 and its associated nucleases in DNA damage response. DNA Repair (Amst) 2019;81:102651. doi: 10.1016/j.dnarep.2019.102651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Martin-Hernandez K., Rodriguez-Vargas J.M., Schreiber V., Dantzer F. Expanding functions of ADP-ribosylation in the maintenance of genome integrity. Semin. Cel. Dev. Biol. 2017;63:92–101. doi: 10.1016/j.semcdb.2016.09.009. [DOI] [PubMed] [Google Scholar]
- 82.Liu C., Vyas A., Kassab M.A., Singh A.K., Yu X. The role of poly ADP-ribosylation in the first wave of DNA damage response. Nucl. Acids Res. 2017;45:8129–8141. doi: 10.1093/nar/gkx565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Burkle A., Virag L. Poly(ADP-ribose): PARadigms and PARadoxes. Mol. Aspects Med. 2013;34:1046–1065. doi: 10.1016/j.mam.2012.12.010. [DOI] [PubMed] [Google Scholar]
- 84.Ray Chaudhuri A., Nussenzweig A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat. Rev. Mol. Cel. Biol. 2017;18:610–621. doi: 10.1038/nrm.2017.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Szanto M., Brunyanszki A., Kiss B., Nagy L., Gergely P., Virag L., et al. Poly(ADP-ribose) polymerase-2: emerging transcriptional roles of a DNA-repair protein. Cell Mol. Life Sci. 2012;69:4079–4092. doi: 10.1007/s00018-012-1003-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Pahi Z.G., Borsos B.N., Pantazi V., Ujfaludi Z., Pankotai T. PARylation during transcription: insights into the fine-tuning mechanism and regulation. Cancers (Basel) 2020;12:183. doi: 10.3390/cancers12010183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Kim D.S., Challa S., Jones A., Kraus W.L. PARPs and ADP-ribosylation in RNA biology: from RNA expression and processing to protein translation and proteostasis. Genes Dev. 2020;34:302–320. doi: 10.1101/gad.334433.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Bock F.J., Todorova T.T., Chang P. RNA regulation by poly(ADP-ribose) polymerases. Mol. Cell. 2015;58:959–969. doi: 10.1016/j.molcel.2015.01.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Munnur D., Bartlett E., Mikolcevic P., Kirby I.T., Rack J.G.M., Mikoc A., et al. Reversible ADP-ribosylation of RNA. Nucl. Acids Res. 2019;47:5658–5669. doi: 10.1093/nar/gkz305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Slade D. Mitotic functions of poly(ADP-ribose) polymerases. Biochem. Pharmacol. 2019;167:33–43. doi: 10.1016/j.bcp.2019.03.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Berger N.A. Poly(ADP-ribose) in the cellular response to DNA damage. Radiat. Res. 1985;101:4–15. [PubMed] [Google Scholar]
- 92.Curtin N.J., Szabo C. Poly(ADP-ribose) polymerase inhibition: past, present and future. Nat. Rev. Drug Discov. 2020;19:711–736. doi: 10.1038/s41573-020-0076-6. [DOI] [PubMed] [Google Scholar]
- 93.Zong W.X., Ditsworth D., Bauer D.E., Wang Z.Q., Thompson C.B. Alkylating DNA damage stimulates a regulated form of necrotic cell death. Genes Dev. 2004;18:1272–1282. doi: 10.1101/gad.1199904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Fischbach A., Kruger A., Hampp S., Assmann G., Rank L., Hufnagel M., et al. The C-terminal domain of p53 orchestrates the interplay between non-covalent and covalent poly(ADP-ribosyl)ation of p53 by PARP1. Nucl. Acids Res. 2018;46:804–822. doi: 10.1093/nar/gkx1205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Ivana Scovassi A., Diederich M. Modulation of poly(ADP-ribosylation) in apoptotic cells. Biochem. Pharmacol. 2004;68:1041–1047. doi: 10.1016/j.bcp.2004.04.023. [DOI] [PubMed] [Google Scholar]
- 96.Wang Y., Dawson V.L., Dawson T.M. Poly(ADP-ribose) signals to mitochondrial AIF: a key event in parthanatos. Exp. Neurol. 2009;218:193–202. doi: 10.1016/j.expneurol.2009.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Schuhwerk H., Bruhn C., Siniuk K., Min W., Erener S., Grigaravicius P., et al. Kinetics of poly(ADP-ribosyl)ation, but not PARP1 itself, determines the cell fate in response to DNA damage in vitro and in vivo. Nucl. Acids Res. 2017;45:11174–11192. doi: 10.1093/nar/gkx717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Cohen M.S. Interplay between compartmentalized NAD(+) synthesis and consumption: a focus on the PARP family. Genes Dev. 2020;34:254–262. doi: 10.1101/gad.335109.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Langelier M.F., Zandarashvili L., Aguiar P.M., Black B.E., Pascal J.M. NAD(+) analog reveals PARP-1 substrate-blocking mechanism and allosteric communication from catalytic center to DNA-binding domains. Nat. Commun. 2018;9:844. doi: 10.1038/s41467-018-03234-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Zhang T., Berrocal J.G., Yao J., DuMond M.E., Krishnakumar R., Ruhl D.D., et al. Regulation of poly(ADP-ribose) polymerase-1-dependent gene expression through promoter-directed recruitment of a nuclear NAD+ synthase. J. Biol. Chem. 2012;287:12405–12416. doi: 10.1074/jbc.M111.304469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Berger F., Lau C., Ziegler M. Regulation of poly(ADP-ribose) polymerase 1 activity by the phosphorylation state of the nuclear NAD biosynthetic enzyme NMN adenylyl transferase 1. Proc. Natl. Acad. Sci. U. S. A. 2007;104:3765–3770. doi: 10.1073/pnas.0609211104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Cameron A.M., Castoldi A., Sanin D.E., Flachsmann L.J., Field C.S., Puleston D.J., et al. Inflammatory macrophage dependence on NAD(+) salvage is a consequence of reactive oxygen species-mediated DNA damage. Nat. Immunol. 2019;20:420–432. doi: 10.1038/s41590-019-0336-y. [DOI] [PubMed] [Google Scholar]
- 103.Li J., Bonkowski M.S., Moniot S., Zhang D., Hubbard B.P., Ling A.J., et al. A conserved NAD(+) binding pocket that regulates protein-protein interactions during aging. Science. 2017;355:1312–1317. doi: 10.1126/science.aad8242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Hurtado-Bagès S., Knobloch G., Ladurner A.G., Buschbeck M. The taming of PARP1 and its impact on NAD+ metabolism. Mol. Metab. 2020;38:100950. doi: 10.1016/j.molmet.2020.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Hopp A.K., Teloni F., Bisceglie L., Gondrand C., Raith F., Nowak K., et al. Mitochondrial NAD(+) controls nuclear ARTD1-induced ADP-ribosylation. Mol. Cell. 2021;81:340–354.e345. doi: 10.1016/j.molcel.2020.12.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Radak Z., Koltai E., Taylor A.W., Higuchi M., Kumagai S., Ohno H., et al. Redox-regulating sirtuins in aging, caloric restriction, and exercise. Free Radic. Biol. Med. 2013;58:87–97. doi: 10.1016/j.freeradbiomed.2013.01.004. [DOI] [PubMed] [Google Scholar]
- 107.van de Ven R.A.H., Santos D., Haigis M.C. Mitochondrial sirtuins and molecular mechanisms of aging. Trends Mol. Med. 2017;23:320–331. doi: 10.1016/j.molmed.2017.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Chalkiadaki A., Guarente L. The multifaceted functions of sirtuins in cancer. Nat. Rev. Cancer. 2015;15:608–624. doi: 10.1038/nrc3985. [DOI] [PubMed] [Google Scholar]
- 109.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. Mol. Biol. Cel. 2005;16:4623–4635. doi: 10.1091/mbc.E05-01-0033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Yanagisawa S., Baker J.R., Vuppusetty C., Koga T., Colley T., Fenwick P., et al. The dynamic shuttling of SIRT1 between cytoplasm and nuclei in bronchial epithelial cells by single and repeated cigarette smoke exposure. PLoS One. 2018;13 doi: 10.1371/journal.pone.0193921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Inoue T., Hiratsuka M., Osaki M., Yamada H., Kishimoto I., Yamaguchi S., et al. SIRT2, a tubulin deacetylase, acts to block the entry to chromosome condensation in response to mitotic stress. Oncogene. 2007;26:945–957. doi: 10.1038/sj.onc.1209857. [DOI] [PubMed] [Google Scholar]
- 112.Du J., Zhou Y., Su X., Yu J.J., Khan S., Jiang H., et al. Sirt5 is a NAD-dependent protein lysine demalonylase and desuccinylase. Science. 2011;334:806–809. doi: 10.1126/science.1207861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Jiang H., Khan S., Wang Y., Charron G., He B., Sebastian C., et al. SIRT6 regulates TNF-alpha secretion through hydrolysis of long-chain fatty acyl lysine. Nature. 2013;496:110–113. doi: 10.1038/nature12038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Imai S.I., Guarente L. It takes two to tango: NAD(+) and sirtuins in aging/longevity control. NPJ Aging Mech. Dis. 2016;2:16017. doi: 10.1038/npjamd.2016.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Houtkooper R.H., Pirinen E., Auwerx J. Sirtuins as regulators of metabolism and healthspan. Nat. Rev. Mol. Cell Biol. 2012;13:225–238. doi: 10.1038/nrm3293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Vachharajani V.T., Liu T., Wang X., Hoth J.J., Yoza B.K., McCall C.E. Sirtuins link inflammation and metabolism. J. Immunol. Res. 2016;2016:8167273. doi: 10.1155/2016/8167273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Masri S., Sassone-Corsi P. Sirtuins and the circadian clock: bridging chromatin and metabolism. Sci. Signal. 2014;7:re6. doi: 10.1126/scisignal.2005685. [DOI] [PubMed] [Google Scholar]
- 118.Singh C.K., Chhabra G., Ndiaye M.A., Garcia-Peterson L.M., Mack N.J., Ahmad N. The role of sirtuins in antioxidant and redox signaling. Antioxid. Redox Signal. 2018;28:643–661. doi: 10.1089/ars.2017.7290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Lee I.H. Mechanisms and disease implications of sirtuin-mediated autophagic regulation. Exp. Mol. Med. 2019;51:1–11. doi: 10.1038/s12276-019-0302-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Verdin E., Hirschey M.D., Finley L.W., Haigis M.C. Sirtuin regulation of mitochondria: energy production, apoptosis, and signaling. Trends Biochem. Sci. 2010;35:669–675. doi: 10.1016/j.tibs.2010.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Vaquero A., Scher M., Lee D., Erdjument-Bromage H., Tempst P., Reinberg D. Human SirT1 interacts with histone H1 and promotes formation of facultative heterochromatin. Mol. Cell. 2004;16:93–105. doi: 10.1016/j.molcel.2004.08.031. [DOI] [PubMed] [Google Scholar]
- 122.Vaquero A., Scher M., Erdjument-Bromage H., Tempst P., Serrano L., Reinberg D. SIRT1 regulates the histone methyl-transferase SUV39H1 during heterochromatin formation. Nature. 2007;450:440–444. doi: 10.1038/nature06268. [DOI] [PubMed] [Google Scholar]
- 123.Kawahara T.L., Michishita E., Adler A.S., Damian M., Berber E., Lin M., et al. SIRT6 links histone H3 lysine 9 deacetylation to NF-kappaB-dependent gene expression and organismal life span. Cell. 2009;136:62–74. doi: 10.1016/j.cell.2008.10.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Barber M.F., Michishita-Kioi E., Xi Y., Tasselli L., Kioi M., Moqtaderi Z., et al. SIRT7 links H3K18 deacetylation to maintenance of oncogenic transformation. Nature. 2012;487:114–118. doi: 10.1038/nature11043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Vaziri H., Dessain S.K., Ng Eaton E., Imai S.I., Frye R.A., Pandita T.K., et al. hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell. 2001;107:149–159. doi: 10.1016/s0092-8674(01)00527-x. [DOI] [PubMed] [Google Scholar]
- 126.Yeung F., Hoberg J.E., Ramsey C.S., Keller M.D., Jones D.R., Frye R.A., et al. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 2004;23:2369–2380. doi: 10.1038/sj.emboj.7600244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Qiang L., Lin H.V., Kim-Muller J.Y., Welch C.L., Gu W., Accili D. Proatherogenic abnormalities of lipid metabolism in SirT1 transgenic mice are mediated through Creb deacetylation. Cell Metab. 2011;14:758–767. doi: 10.1016/j.cmet.2011.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Utani K., Aladjem M.I. Extra view: sirt1 acts as a gatekeeper of replication initiation to preserve genomic stability. Nucleus (Austin, Tex.) 2018;9:261–267. doi: 10.1080/19491034.2018.1456218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Zhang T., Kraus W.L. SIRT1-dependent regulation of chromatin and transcription: linking NAD(+) metabolism and signaling to the control of cellular functions. Biochim. Biophys. Acta. 2010;1804:1666–1675. doi: 10.1016/j.bbapap.2009.10.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Mostoslavsky R., Chua K.F., Lombard D.B., Pang W.W., Fischer M.R., Gellon L., et al. Genomic instability and aging-like phenotype in the absence of mammalian SIRT6. Cell. 2006;124:315–329. doi: 10.1016/j.cell.2005.11.044. [DOI] [PubMed] [Google Scholar]
- 131.Hou T., Cao Z., Zhang J., Tang M., Tian Y., Li Y., et al. SIRT6 coordinates with CHD4 to promote chromatin relaxation and DNA repair. Nucl. Acids Res. 2020;48:2982–3000. doi: 10.1093/nar/gkaa006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Onn L., Portillo M., Ilic S., Cleitman G., Stein D., Kaluski S., et al. SIRT6 is a DNA double-strand break sensor. eLife. 2020;9 doi: 10.7554/eLife.51636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Van Meter M., Simon M., Tombline G., May A., Morello T.D., Hubbard B.P., et al. JNK phosphorylates SIRT6 to stimulate DNA double-strand break repair in response to oxidative stress by recruiting PARP1 to DNA breaks. Cell Rep. 2016;16:2641–2650. doi: 10.1016/j.celrep.2016.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Vazquez B.N., Thackray J.K., Serrano L. Sirtuins and DNA damage repair: SIRT7 comes to play. Nucleus (Austin, Tex.) 2017;8:107–115. doi: 10.1080/19491034.2016.1264552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Li L., Shi L., Yang S., Yan R., Zhang D., Yang J., et al. SIRT7 is a histone desuccinylase that functionally links to chromatin compaction and genome stability. Nat. Commun. 2016;7:12235. doi: 10.1038/ncomms12235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Tang M., Li Z., Zhang C., Lu X., Tu B., Cao Z., et al. SIRT7-mediated ATM deacetylation is essential for its deactivation and DNA damage repair. Sci. Adv. 2019;5 doi: 10.1126/sciadv.aav1118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Tsai Y.C., Greco T.M., Cristea I.M. Sirtuin 7 plays a role in ribosome biogenesis and protein synthesis. Mol. Cell. Proteomics. 2014;13:73–83. doi: 10.1074/mcp.M113.031377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Chen S., Blank M.F., Iyer A., Huang B., Wang L., Grummt I., et al. SIRT7-dependent deacetylation of the U3-55k protein controls pre-rRNA processing. Nat. Commun. 2016;7:10734. doi: 10.1038/ncomms10734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Blank M.F., Chen S., Poetz F., Schnolzer M., Voit R., Grummt I. SIRT7-dependent deacetylation of CDK9 activates RNA polymerase II transcription. Nucl. Acids Res. 2017;45:2675–2686. doi: 10.1093/nar/gkx053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Michishita E., McCord R.A., Berber E., Kioi M., Padilla-Nash H., Damian M., et al. SIRT6 is a histone H3 lysine 9 deacetylase that modulates telomeric chromatin. Nature. 2008;452:492–496. doi: 10.1038/nature06736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Gomes P., Fleming Outeiro T., Cavadas C. Emerging role of sirtuin 2 in the regulation of mammalian metabolism. Trends Pharmacol. Sci. 2015;36:756–768. doi: 10.1016/j.tips.2015.08.001. [DOI] [PubMed] [Google Scholar]
- 142.Pereira J.M., Chevalier C., Chaze T., Gianetto Q., Impens F., Matondo M., et al. Infection reveals a modification of SIRT2 critical for chromatin association. Cell Rep. 2018;23:1124–1137. doi: 10.1016/j.celrep.2018.03.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Head P.E., Zhang H., Bastien A.J., Koyen A.E., Withers A.E., Daddacha W.B., et al. Sirtuin 2 mutations in human cancers impair its function in genome maintenance. J. Biol. Chem. 2017;292:9919–9931. doi: 10.1074/jbc.M116.772566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Zhang H., Head P.E., Yu D.S. SIRT2 orchestrates the DNA damage response. Cell Cycle (Georgetown, Tex.) 2016;15:2089–2090. doi: 10.1080/15384101.2016.1184517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Zhang H., Head P.E., Daddacha W., Park S.H., Li X., Pan Y., et al. ATRIP deacetylation by SIRT2 drives ATR checkpoint activation by promoting binding to RPA-ssDNA. Cell Rep. 2016;14:1435–1447. doi: 10.1016/j.celrep.2016.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Sundaresan N.R., Samant S.A., Pillai V.B., Rajamohan S.B., Gupta M.P. SIRT3 is a stress-responsive deacetylase in cardiomyocytes that protects cells from stress-mediated cell death by deacetylation of Ku70. Mol. Cell. Biol. 2008;28:6384–6401. doi: 10.1128/MCB.00426-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Yasuda T., Kagawa W., Ogi T., Kato T.A., Suzuki T., Dohmae N., et al. Novel function of HATs and HDACs in homologous recombination through acetylation of human RAD52 at double-strand break sites. PLoS Genet. 2018;14 doi: 10.1371/journal.pgen.1007277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Cheng Y., Ren X., Gowda A.S., Shan Y., Zhang L., Yuan Y.S., et al. Interaction of Sirt3 with OGG1 contributes to repair of mitochondrial DNA and protects from apoptotic cell death under oxidative stress. Cell Death Dis. 2013;4:e731. doi: 10.1038/cddis.2013.254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Blank M.F., Grummt I. The seven faces of SIRT7. Transcription. 2017;8:67–74. doi: 10.1080/21541264.2016.1276658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Kugel S., Mostoslavsky R. Chromatin and beyond: the multitasking roles for SIRT6. Trends Biochem. Sci. 2014;39:72–81. doi: 10.1016/j.tibs.2013.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Bruzzone S., Fruscione F., Morando S., Ferrando T., Poggi A., Garuti A., et al. Catastrophic NAD+ depletion in activated T lymphocytes through Nampt inhibition reduces demyelination and disability in EAE. PLoS One. 2009;4 doi: 10.1371/journal.pone.0007897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Belenky P., Racette F.G., Bogan K.L., McClure J.M., Smith J.S., Brenner C. Nicotinamide riboside promotes Sir2 silencing and extends lifespan via Nrk and Urh1/Pnp1/Meu1 pathways to NAD+ Cell. 2007;129:473–484. doi: 10.1016/j.cell.2007.03.024. [DOI] [PubMed] [Google Scholar]
- 153.Revollo J.R., Grimm A.A., Imai S. The NAD biosynthesis pathway mediated by nicotinamide phosphoribosyltransferase regulates Sir2 activity in mammalian cells. J. Biol. Chem. 2004;279:50754–50763. doi: 10.1074/jbc.M408388200. [DOI] [PubMed] [Google Scholar]
- 154.Zhang T., Berrocal J.G., Frizzell K.M., Gamble M.J., DuMond M.E., Krishnakumar R., et al. Enzymes in the NAD+ salvage pathway regulate SIRT1 activity at target gene promoters. J. Biol. Chem. 2009;284:20408–20417. doi: 10.1074/jbc.M109.016469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Zhan T., Xiong H., Pang J., Zhang W., Ye Y., Liang Z., et al. Modulation of NAD(+) biosynthesis activates SIRT1 and resists cisplatin-induced ototoxicity. Toxicol. Lett. 2021;349:115–123. doi: 10.1016/j.toxlet.2021.05.013. [DOI] [PubMed] [Google Scholar]
- 156.Sociali G., Grozio A., Caffa I., Schuster S., Becherini P., Damonte P., et al. SIRT6 deacetylase activity regulates NAMPT activity and NAD(P)(H) pools in cancer cells. FASEB J. 2019;33:3704–3717. doi: 10.1096/fj.201800321R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Buonvicino D., Ranieri G., Pittelli M., Lapucci A., Bragliola S., Chiarugi A. SIRT1-dependent restoration of NAD+ homeostasis after increased extracellular NAD+ exposure. J. Biol. Chem. 2021;297:100855. doi: 10.1016/j.jbc.2021.100855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Cantó C., Sauve A.A., Bai P. Crosstalk between poly(ADP-ribose) polymerase and sirtuin enzymes. Mol. Aspects Med. 2013;34:1168–1201. doi: 10.1016/j.mam.2013.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Luna A., Aladjem M.I., Kohn K.W. SIRT1/PARP1 crosstalk: connecting DNA damage and metabolism. Genome Integr. 2013;4:6. doi: 10.1186/2041-9414-4-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Kolthur-Seetharam U., Dantzer F., McBurney M.W., Murcia G.d., Sassone-Corsi P. Control of AIF-mediated cell death by the functional interplay of SIRT1 and PARP-1 in response to DNA damage. Cell Cycle (Georgetown, Tex.) 2006;5:873–877. doi: 10.4161/cc.5.8.2690. [DOI] [PubMed] [Google Scholar]
- 161.Rajamohan S.B., Pillai V.B., Gupta M., Sundaresan N.R., Birukov K.G., Samant S., et al. SIRT1 promotes cell survival under stress by deacetylation-dependent deactivation of poly(ADP-ribose) polymerase 1. Mol. Cell. Biol. 2009;29:4116–4129. doi: 10.1128/MCB.00121-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Feng X., Wang Y., Chen W., Xu S., Li L., Geng Y., et al. SIRT3 inhibits cardiac hypertrophy by regulating PARP-1 activity. Aging (Albany NY) 2020;12:4178–4192. doi: 10.18632/aging.102862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Mao Z., Hine C., Tian X., Van Meter M., Au M., Vaidya A., et al. SIRT6 promotes DNA repair under stress by activating PARP1. Science (New York, N.Y.) 2011;332:1443–1446. doi: 10.1126/science.1202723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Bai P., Cantó C., Oudart H., Brunyánszki A., Cen Y., Thomas C., et al. PARP-1 inhibition increases mitochondrial metabolism through SIRT1 activation. Cell Metab. 2011;13:461–468. doi: 10.1016/j.cmet.2011.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Bai P., Canto C., Brunyánszki A., Huber A., Szántó M., Cen Y., et al. PARP-2 regulates SIRT1 expression and whole-body energy expenditure. Cell Metab. 2011;13:450–460. doi: 10.1016/j.cmet.2011.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Xu Z., Zhang L., Zhang W., Meng D., Zhang H., Jiang Y., et al. SIRT6 rescues the age related decline in base excision repair in a PARP1-dependent manner. Cell Cycle (Georgetown, Tex.) 2015;14:269–276. doi: 10.4161/15384101.2014.980641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Lehman I.R. DNA ligase: structure, mechanism, and function. Science. 1974;186:790–797. doi: 10.1126/science.186.4166.790. [DOI] [PubMed] [Google Scholar]
- 168.Shuman S. DNA ligases: progress and prospects. J. Biol. Chem. 2009;284:17365–17369. doi: 10.1074/jbc.R900017200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Doherty A.J., Suh S.W. Structural and mechanistic conservation in DNA ligases. Nucl. Acids Res. 2000;28:4051–4058. doi: 10.1093/nar/28.21.4051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Barnes D.E., Tomkinson A.E., Lehmann A.R., Webster A.D., Lindahl T. Mutations in the DNA ligase I gene of an individual with immunodeficiencies and cellular hypersensitivity to DNA-damaging agents. Cell. 1992;69:495–503. doi: 10.1016/0092-8674(92)90450-q. [DOI] [PubMed] [Google Scholar]
- 171.Akbari M., Keijzers G., Maynard S., Scheibye-Knudsen M., Desler C., Hickson I.D., et al. Overexpression of DNA ligase III in mitochondria protects cells against oxidative stress and improves mitochondrial DNA base excision repair. DNA Repair (Amst) 2014;16:44–53. doi: 10.1016/j.dnarep.2014.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Altmann T., Gennery A.R. DNA ligase IV syndrome; a review. Orphanet J. Rare Dis. 2016;11:137. doi: 10.1186/s13023-016-0520-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Ellenberger T., Tomkinson A.E. Eukaryotic DNA ligases: structural and functional insights. Annu. Rev. Biochem. 2008;77:313–338. doi: 10.1146/annurev.biochem.77.061306.123941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Zhao B., Naila T., Lieber M.R., Tomkinson A.E. NAD+ is not utilized as a co-factor for DNA ligation by human DNA ligase IV. Nucl. Acids Res. 2020;48:12746–12750. doi: 10.1093/nar/gkaa1118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Bi F., Ma R., Ma S. Discovery and optimization of NAD+-dependent DNA ligase inhibitors as novel antibacterial compounds. Curr. Pharm. Des. 2017;23:2117–2130. doi: 10.2174/1381612822666161025145639. [DOI] [PubMed] [Google Scholar]
- 176.Narlikar G.J., Sundaramoorthy R., Owen-Hughes T. Mechanisms and functions of ATP-dependent chromatin-remodeling enzymes. Cell. 2013;154:490–503. doi: 10.1016/j.cell.2013.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Clapier C.R., Iwasa J., Cairns B.R., Peterson C.L. Mechanisms of action and regulation of ATP-dependent chromatin-remodelling complexes. Nat. Rev. Mol. Cell Biol. 2017;18:407–422. doi: 10.1038/nrm.2017.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Martin I.V., MacNeill S.A. ATP-dependent DNA ligases. Genome Biol. 2002;3 doi: 10.1186/gb-2002-3-4-reviews3005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Shiloh Y., Ziv Y. The ATM protein kinase: regulating the cellular response to genotoxic stress, and more. Nat. Rev. Mol. Cell Biol. 2013;14:197–210. [PubMed] [Google Scholar]
- 180.Liu S., Opiyo S.O., Manthey K., Glanzer J.G., Ashley A.K., Amerin C., et al. Distinct roles for DNA-PK, ATM and ATR in RPA phosphorylation and checkpoint activation in response to replication stress. Nucl. Acids Res. 2012;40:10780–10794. doi: 10.1093/nar/gks849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Deshpande R.A., Lee J.H., Paull T.T. Rad50 ATPase activity is regulated by DNA ends and requires coordination of both active sites. Nucl. Acids Res. 2017;45:5255–5268. doi: 10.1093/nar/gkx173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Yount A.L., Zong H., Walczak C.E. Regulatory mechanisms that control mitotic kinesins. Exp. Cel. Res. 2015;334:70–77. doi: 10.1016/j.yexcr.2014.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Lynch M., Marinov G.K. The bioenergetic costs of a gene. Proc. Natl. Acad. Sci. U. S. A. 2015;112:15690–15695. doi: 10.1073/pnas.1514974112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Sies H., Jones D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cel. Biol. 2020;21:363–383. doi: 10.1038/s41580-020-0230-3. [DOI] [PubMed] [Google Scholar]
- 185.Perillo B., Di Donato M., Pezone A., Di Zazzo E., Giovannelli P., Galasso G., et al. ROS in cancer therapy: the bright side of the moon. Exp. Mol. Med. 2020;52:192–203. doi: 10.1038/s12276-020-0384-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Kreuz S., Fischle W. Oxidative stress signaling to chromatin in health and disease. Epigenomics. 2016;8:843–862. doi: 10.2217/epi-2016-0002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Sirover M.A. Pleiotropic effects of moonlighting glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in cancer progression, invasiveness, and metastases. Cancer Met. Rev. 2018;37:665–676. doi: 10.1007/s10555-018-9764-7. [DOI] [PubMed] [Google Scholar]
- 188.Wang J.Q., Chen Q., Wang X., Wang Q.C., Wang Y., Cheng H.P., et al. Dysregulation of mitochondrial calcium signaling and superoxide flashes cause mitochondrial genomic DNA damage in Huntington disease. J. Biol. Chem. 2013;288:3070–3084. doi: 10.1074/jbc.M112.407726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Bugreev D.V., Mazin A.V. Ca2+ activates human homologous recombination protein Rad51 by modulating its ATPase activity. Proc. Natl. Acad. Sci. U. S. A. 2004;101:9988–9993. doi: 10.1073/pnas.0402105101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Dantas T.J., Wang Y., Lalor P., Dockery P., Morrison C.G. Defective nucleotide excision repair with normal centrosome structures and functions in the absence of all vertebrate centrins. J. Cel. Biol. 2011;193:307–318. doi: 10.1083/jcb.201012093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Misquitta C.M., Chen T., Grover A.K. Control of protein expression through mRNA stability in calcium signalling. Cell Calcium. 2006;40:329–346. doi: 10.1016/j.ceca.2006.04.004. [DOI] [PubMed] [Google Scholar]
- 192.Salisbury J.L., Suino K.M., Busby R., Springett M. Centrin-2 is required for centriole duplication in mammalian cells. Curr. Biol. 2002;12:1287–1292. doi: 10.1016/s0960-9822(02)01019-9. [DOI] [PubMed] [Google Scholar]
- 193.Helassa N., Nugues C., Rajamanoharan D., Burgoyne R.D., Haynes L.P. A centrosome-localized calcium signal is essential for mammalian cell mitosis. FASEB J. 2019;33:14602–14610. doi: 10.1096/fj.201901662R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Stewart T.A., Yapa K.T., Monteith G.R. Altered calcium signaling in cancer cells. Biochim. Biophys. Acta. 2015;1848:2502–2511. doi: 10.1016/j.bbamem.2014.08.016. [DOI] [PubMed] [Google Scholar]
- 195.Martinez-Outschoorn U.E., Peiris-Pages M., Pestell R.G., Sotgia F., Lisanti M.P. Cancer metabolism: a therapeutic perspective. Nat. Rev. Clin. Oncol. 2017;14:11–31. doi: 10.1038/nrclinonc.2016.60. [DOI] [PubMed] [Google Scholar]
- 196.Spronck J.C., Nickerson J.L., Kirkland J.B. Niacin deficiency alters p53 expression and impairs etoposide-induced cell cycle arrest and apoptosis in rat bone marrow cells. Nutr. Cancer. 2007;57:88–99. doi: 10.1080/01635580701268337. [DOI] [PubMed] [Google Scholar]
- 197.Spronck J.C., Bartleman A.P., Boyonoski A.C., Kirkland J.B. Chronic DNA damage and niacin deficiency enhance cell injury and cause unusual interactions in NAD and poly(ADP-ribose) metabolism in rat bone marrow. Nutr. Cancer. 2003;45:124–131. doi: 10.1207/S15327914NC4501_14. [DOI] [PubMed] [Google Scholar]
- 198.Boyonoski A.C., Spronck J.C., Gallacher L.M., Jacobs R.M., Shah G.M., Poirier G.G., et al. Niacin deficiency decreases bone marrow poly(ADP-ribose) and the latency of ethylnitrosourea-induced carcinogenesis in rats. J. Nutr. 2002;132:108–114. doi: 10.1093/jn/132.1.108. [DOI] [PubMed] [Google Scholar]
- 199.Boyonoski A.C., Spronck J.C., Jacobs R.M., Shah G.M., Poirier G.G., Kirkland J.B. Pharmacological intakes of niacin increase bone marrow poly(ADP-ribose) and the latency of ethylnitrosourea-induced carcinogenesis in rats. J. Nutr. 2002;132:115–120. doi: 10.1093/jn/132.1.115. [DOI] [PubMed] [Google Scholar]
- 200.Kirkland J.B. Niacin status and genomic instability in bone marrow cells; mechanisms favoring the progression of leukemogenesis. Sub-cellular Biochem. 2012;56:21–36. doi: 10.1007/978-94-007-2199-9_2. [DOI] [PubMed] [Google Scholar]
- 201.Berger N.A., Sikorski G.W. Nicotinamide stimulates repair of DNA damage in human lymphocytes. Biochem. Biophys. Res. Commun. 1980;95:67–72. doi: 10.1016/0006-291x(80)90705-6. [DOI] [PubMed] [Google Scholar]
- 202.Fang E.F., Scheibye-Knudsen M., Brace L.E., Kassahun H., SenGupta T., Nilsen H., et al. Defective mitophagy in XPA via PARP-1 hyperactivation and NAD(+)/SIRT1 reduction. Cell. 2014;157:882–896. doi: 10.1016/j.cell.2014.03.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Fang E.F., Kassahun H., Croteau D.L., Scheibye-Knudsen M., Marosi K., Lu H., et al. NAD(+) replenishment improves lifespan and healthspan in ataxia telangiectasia models via mitophagy and DNA repair. Cell Metab. 2016;24:566–581. doi: 10.1016/j.cmet.2016.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Scheibye-Knudsen M., Mitchell S.J., Fang E.F., Iyama T., Ward T., Wang J., et al. A high-fat diet and NAD(+) activate Sirt1 to rescue premature aging in cockayne syndrome. Cell Metab. 2014;20:840–855. doi: 10.1016/j.cmet.2014.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Hou Y., Lautrup S., Cordonnier S., Wang Y., Croteau D.L., Zavala E., et al. NAD(+) supplementation normalizes key Alzheimer's features and DNA damage responses in a new AD mouse model with introduced DNA repair deficiency. Proc. Natl. Acad. Sci. U. S. A. 2018;115:E1876–E1885. doi: 10.1073/pnas.1718819115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Okur M.N., Mao B., Kimura R., Haraczy S., Fitzgerald T., Edwards-Hollingsworth K., et al. Short-term NAD(+) supplementation prevents hearing loss in mouse models of Cockayne syndrome. NPJ Aging Mech. Dis. 2020;6:1. doi: 10.1038/s41514-019-0040-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Fang E.F., Hou Y., Lautrup S., Jensen M.B., Yang B., SenGupta T., et al. NAD(+) augmentation restores mitophagy and limits accelerated aging in Werner syndrome. Nat. Commun. 2019;10:5284. doi: 10.1038/s41467-019-13172-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Sun X., Cao B., Naval-Sanchez M., Pham T., Sun Y.B.Y., Williams B., et al. Nicotinamide riboside attenuates age-associated metabolic and functional changes in hematopoietic stem cells. Nat. Commun. 2021;12:2665. doi: 10.1038/s41467-021-22863-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Sun C., Wang K., Stock A.J., Gong Y., Demarest T.G., Yang B., et al. Re-equilibration of imbalanced NAD metabolism ameliorates the impact of telomere dysfunction. EMBO J. 2020;39 doi: 10.15252/embj.2019103420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Chen A.C., Martin A.J., Choy B., Fernández-Peñas P., Dalziell R.A., McKenzie C.A., et al. A phase 3 randomized trial of nicotinamide for skin-cancer chemoprevention. New Engl. J. Med. 2015;373:1618–1626. doi: 10.1056/NEJMoa1506197. [DOI] [PubMed] [Google Scholar]
- 211.Surjana D., Halliday G.M., Martin A.J., Moloney F.J., Damian D.L. Oral nicotinamide reduces actinic keratoses in phase II double-blinded randomized controlled trials. J. Invest. Dermatol. 2012;132:1497–1500. doi: 10.1038/jid.2011.459. [DOI] [PubMed] [Google Scholar]
- 212.Xie N., Zhang L., Gao W., Huang C., Huber P.E., Zhou X., et al. NAD(+) metabolism: pathophysiologic mechanisms and therapeutic potential. Signal. Transduct. Target Ther. 2020;5:227. doi: 10.1038/s41392-020-00311-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Yoshino J., Baur J.A., Imai S.I. NAD(+) intermediates: the biology and therapeutic potential of NMN and NR. Cell Metab. 2018;27:513–528. doi: 10.1016/j.cmet.2017.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Zhang H., Ryu D., Wu Y., Gariani K., Wang X., Luan P., et al. NAD(+) repletion improves mitochondrial and stem cell function and enhances life span in mice. Science. 2016;352:1436–1443. doi: 10.1126/science.aaf2693. [DOI] [PubMed] [Google Scholar]
- 215.Dollerup O.L., Trammell S.A.J., Hartmann B., Holst J.J., Christensen B., Moller N., et al. Effects of nicotinamide riboside on endocrine Pancreatic function and incretin hormones in nondiabetic men with obesity. J. Clin. Endocrinol. Metab. 2019;104:5703–5714. doi: 10.1210/jc.2019-01081. [DOI] [PubMed] [Google Scholar]
- 216.DeBerardinis R.J., Lum J.J., Hatzivassiliou G., Thompson C.B. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 2008;7:11–20. doi: 10.1016/j.cmet.2007.10.002. [DOI] [PubMed] [Google Scholar]
- 217.Warburg O., Wind F., Negelein E. The metabolism of tumors in the body. J. Gen. Physiol. 1927;8:519–530. doi: 10.1085/jgp.8.6.519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Nacarelli T., Lau L., Fukumoto T., Zundell J., Fatkhutdinov N., Wu S., et al. NAD(+) metabolism governs the proinflammatory senescence-associated secretome. Nat. Cel. Biol. 2019;21:397–407. doi: 10.1038/s41556-019-0287-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Sampath D., Zabka T.S., Misner D.L., O'Brien T., Dragovich P.S. Inhibition of nicotinamide phosphoribosyltransferase (NAMPT) as a therapeutic strategy in cancer. Pharmacol. Ther. 2015;151:16–31. doi: 10.1016/j.pharmthera.2015.02.004. [DOI] [PubMed] [Google Scholar]
- 220.Gujar A.D., Le S., Mao D.D., Dadey D.Y., Turski A., Sasaki Y., et al. An NAD+-dependent transcriptional program governs self-renewal and radiation resistance in glioblastoma. Proc. Natl. Acad. Sci. U. S. A. 2016;113:E8247–E8256. doi: 10.1073/pnas.1610921114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Fons N.R., Sundaram R.K., Breuer G.A., Peng S., McLean R.L., Kalathil A.N., et al. PPM1D mutations silence NAPRT gene expression and confer NAMPT inhibitor sensitivity in glioma. Nat. Commun. 2019;10:3790. doi: 10.1038/s41467-019-11732-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Chowdhry S., Zanca C., Rajkumar U., Koga T., Diao Y., Raviram R., et al. NAD metabolic dependency in cancer is shaped by gene amplification and enhancer remodelling. Nature. 2019;569:570–575. doi: 10.1038/s41586-019-1150-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Tummala K.S., Gomes A.L., Yilmaz M., Grana O., Bakiri L., Ruppen I., et al. Inhibition of de novo NAD(+) synthesis by oncogenic URI causes liver tumorigenesis through DNA damage. Cancer cell. 2014;26:826–839. doi: 10.1016/j.ccell.2014.10.002. [DOI] [PubMed] [Google Scholar]
- 224.Gensler H.L., Williams T., Huang A.C., Jacobson E.L. Oral niacin prevents photocarcinogenesis and photoimmunosuppression in mice. Nutr. Cancer. 1999;34:36–41. doi: 10.1207/S15327914NC340105. [DOI] [PubMed] [Google Scholar]
- 225.Zhu Y., Liu J., Park J., Rai P., Zhai R.G. Subcellular compartmentalization of NAD(+) and its role in cancer: a sereNADe of metabolic melodies. Pharmacol. Ther. 2019;200:27–41. doi: 10.1016/j.pharmthera.2019.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Haince J.F., Kozlov S., Dawson V.L., Dawson T.M., Hendzel M.J., Lavin M.F., et al. Ataxia telangiectasia mutated (ATM) signaling network is modulated by a novel poly(ADP-ribose)-dependent pathway in the early response to DNA-damaging agents. J. Biol. Chem. 2007;282:16441–16453. doi: 10.1074/jbc.M608406200. [DOI] [PubMed] [Google Scholar]
- 227.Gottschalk A.J., Timinszky G., Kong S.E., Jin J., Cai Y., Swanson S.K., et al. Poly(ADP-ribosyl)ation directs recruitment and activation of an ATP-dependent chromatin remodeler. Proc. Natl. Acad. Sci. U. S. A. 2009;106:13770–13774. doi: 10.1073/pnas.0906920106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Pines A., Vrouwe M.G., Marteijn J.A., Typas D., Luijsterburg M.S., Cansoy M., et al. PARP1 promotes nucleotide excision repair through DDB2 stabilization and recruitment of ALC1. J. Cell Biol. 2012;199:235–249. doi: 10.1083/jcb.201112132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Mastrocola A.S., Kim S.H., Trinh A.T., Rodenkirch L.A., Tibbetts R.S. The RNA-binding protein fused in sarcoma (FUS) functions downstream of poly(ADP-ribose) polymerase (PARP) in response to DNA damage. J. Biol. Chem. 2013;288:24731–24741. doi: 10.1074/jbc.M113.497974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Leppard J.B., Dong Z., Mackey Z.B., Tomkinson A.E. Physical and functional interaction between DNA ligase IIIalpha and poly(ADP-Ribose) polymerase 1 in DNA single-strand break repair. Mol. Cell. Biol. 2003;23:5919–5927. doi: 10.1128/MCB.23.16.5919-5927.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Altmeyer M., Toledo L., Gudjonsson T., Grofte M., Rask M.B., Lukas C., et al. The chromatin scaffold protein SAFB1 renders chromatin permissive for DNA damage signaling. Mol. Cell. 2013;52:206–220. doi: 10.1016/j.molcel.2013.08.025. [DOI] [PubMed] [Google Scholar]
- 232.Das B.B., Huang S.Y., Murai J., Rehman I., Ame J.C., Sengupta S., et al. PARP1-TDP1 coupling for the repair of topoisomerase I-induced DNA damage. Nucl. Acids Res. 2014;42:4435–4449. doi: 10.1093/nar/gku088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Xie S., Mortusewicz O., Ma H.T., Herr P., Poon R.Y., Helleday T., et al. Timeless interacts with PARP-1 to promote homologous recombination repair. Mol. Cell. 2015;60:163–176. doi: 10.1016/j.molcel.2015.07.031. [DOI] [PubMed] [Google Scholar]
- 234.King B.S., Cooper K.L., Liu K.J., Hudson L.G. Poly(ADP-ribose) contributes to an association between poly(ADP-ribose) polymerase-1 and xeroderma pigmentosum complementation group A in nucleotide excision repair. J. Biol. Chem. 2012;287:39824–39833. doi: 10.1074/jbc.M112.393504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Quenet D., Gasser V., Fouillen L., Cammas F., Sanglier-Cianferani S., Losson R., et al. The histone subcode: poly(ADP-ribose) polymerase-1 (Parp-1) and Parp-2 control cell differentiation by regulating the transcriptional intermediary factor TIF1beta and the heterochromatin protein HP1alpha. FASEB J. 2008;22:3853–3865. doi: 10.1096/fj.08-113464. [DOI] [PubMed] [Google Scholar]
- 236.Schreiber V., Ame J.C., Dolle P., Schultz I., Rinaldi B., Fraulob V., et al. Poly(ADP-ribose) polymerase-2 (PARP-2) is required for efficient base excision DNA repair in association with PARP-1 and XRCC1. J. Biol. Chem. 2002;277:23028–23036. doi: 10.1074/jbc.M202390200. [DOI] [PubMed] [Google Scholar]
- 237.Rulten S.L., Fisher A.E., Robert I., Zuma M.C., Rouleau M., Ju L., et al. PARP-3 and APLF function together to accelerate nonhomologous end-joining. Mol. Cel. 2011;41:33–45. doi: 10.1016/j.molcel.2010.12.006. [DOI] [PubMed] [Google Scholar]
- 238.Rouleau M., McDonald D., Gagne P., Ouellet M.E., Droit A., Hunter J.M., et al. PARP-3 associates with polycomb group bodies and with components of the DNA damage repair machinery. J. Cell. Biochem. 2007;100:385–401. doi: 10.1002/jcb.21051. [DOI] [PubMed] [Google Scholar]
- 239.Boehler C., Gauthier L.R., Mortusewicz O., Biard D.S., Saliou J.M., Bresson A., et al. Poly(ADP-ribose) polymerase 3 (PARP3), a newcomer in cellular response to DNA damage and mitotic progression. Proc. Natl. Acad. Sci. U. S. A. 2011;108:2783–2788. doi: 10.1073/pnas.1016574108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Yan Q., Xu R., Zhu L., Cheng X., Wang Z., Manis J., et al. BAL1 and its partner E3 ligase, BBAP, link Poly(ADP-ribose) activation, ubiquitylation, and double-strand DNA repair independent of ATM, MDC1, and RNF8. Mol. Cell. Biol. 2013;33:845–857. doi: 10.1128/MCB.00990-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Nicolae C.M., Aho E.R., Vlahos A.H., Choe K.N., De S., Karras G.I., et al. The ADP-ribosyltransferase PARP10/ARTD10 interacts with proliferating cell nuclear antigen (PCNA) and is required for DNA damage tolerance. J. Biol. Chem. 2014;289:13627–13637. doi: 10.1074/jbc.M114.556340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Nicolae C.M., Aho E.R., Choe K.N., Constantin D., Hu H.J., Lee D., et al. A novel role for the mono-ADP-ribosyltransferase PARP14/ARTD8 in promoting homologous recombination and protecting against replication stress. Nucl. Acids Res. 2015;43:3143–3153. doi: 10.1093/nar/gkv147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Nagy Z., Kalousi A., Furst A., Koch M., Fischer B., Soutoglou E. Tankyrases promote homologous recombination and check point activation in response to DSBs. PLoS Genet. 2016;12 doi: 10.1371/journal.pgen.1005791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Okamoto K., Ohishi T., Kuroiwa M., Iemura S.I., Natsume T., Seimiya H. MERIT40-dependent recruitment of tankyrase to damaged DNA and its implication for cell sensitivity to DNA-damaging anticancer drugs. Oncotarget. 2018;9:35844–35855. doi: 10.18632/oncotarget.26312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Yamamori T., DeRicco J., Naqvi A., Hoffman T.A., Mattagajasingh I., Kasuno K., et al. SIRT1 deacetylates APE1 and regulates cellular base excision repair. Nucl. Acids Res. 2010;38:832–845. doi: 10.1093/nar/gkp1039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Jeong J., Juhn K., Lee H., Kim S.H., Min B.H., Lee K.M., et al. SIRT1 promotes DNA repair activity and deacetylation of Ku70. Exp. Mol. Med. 2007;39:8–13. doi: 10.1038/emm.2007.2. [DOI] [PubMed] [Google Scholar]
- 247.Yuan Z., Zhang X., Sengupta N., Lane W.S., Seto E. SIRT1 regulates the function of the Nijmegen breakage syndrome protein. Mol. Cell. 2007;27:149–162. doi: 10.1016/j.molcel.2007.05.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Madabushi A., Hwang B.J., Jin J., Lu A.L. Histone deacetylase SIRT1 modulates and deacetylates DNA base excision repair enzyme thymine DNA glycosylase. Biochem. J. 2013;456:89–98. doi: 10.1042/BJ20130670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Li K., Casta A., Wang R., Lozada E., Fan W., Kane S., et al. Regulation of WRN protein cellular localization and enzymatic activities by SIRT1-mediated deacetylation. J. Biol. Chem. 2008;283:7590–7598. doi: 10.1074/jbc.M709707200. [DOI] [PubMed] [Google Scholar]
- 250.Fan W., Luo J. SIRT1 regulates UV-induced DNA repair through deacetylating XPA. Mol. Cell. 2010;39:247–258. doi: 10.1016/j.molcel.2010.07.006. [DOI] [PubMed] [Google Scholar]
- 251.Zhang H., Park S.H., Pantazides B.G., Karpiuk O., Warren M.D., Hardy C.W., et al. SIRT2 directs the replication stress response through CDK9 deacetylation. Proc. Natl. Acad. Sci. U. S. A. 2013;110:13546–13551. doi: 10.1073/pnas.1301463110. [DOI] [PMC free article] [PubMed] [Google Scholar]