• Open Access
    Review

    Sirtuin activators as an anti-aging intervention for longevity

    Puja Sah
    Anita K. Rai
    Donkupar Syiem *

    Explor Drug Sci. 2025;3:100881 DOI: https://doi.org/10.37349/eds.2025.100881

    Received: August 01, 2024 Accepted: November 15, 2024 Published: January 12, 2025

    Academic Editor: Ricardo P. Garay, French National Council of Scientific Research, France

    This article belongs to the special issue Recent advances with investigational compounds and strategies to delay or reverse normal aging processes

    Abstract

    Sirtuins are a family of NAD+-dependent class III histone deacetylases that regulate histones and other proteins. The mammalian sirtuins comprise seven members that have a role in energy metabolism, DNA repair, inflammation, cell survival, apoptosis, cellular senescence, oxidative stress, and mitochondrial production. Sirtuin modulation may have beneficial effects on aging and age-related diseases; thus, attracting a growing interest in discovering small molecules modifying their activity. A class of compounds both natural and chemically synthesized has emerged as sirtuin activators. This review discusses mammalian sirtuins in aging, the small molecules that activate sirtuins, modulation of sirtuin activity, and its impact in alleviating the effects of aging.

    Impact of activators on sirtuins and its role in alleviating the effects of aging. PGC-1α: peroxisome proliferator-activated receptor-γ co-activator 1α; AMPK: AMP-activated protein kinase; Pol I: polymerase I; SOD: superoxide dismutase

    Keywords

    Sirtuins, activators, aging, resveratrol, NAD+

    Introduction

    The world population is expected to peak around 2084, rising to 10.3 billion [1]. However, the rate of population aging is faster than it ever was in the past. By the end of this decade, in 2030, one in six individuals will be 60 or older, increasing the population of people aged 60 to 22% by the year 2050 [2]. In the upcoming decades, this will lead to aging being one of the major socio-economic and psychological burdens for individuals and society globally [3]. As an individual’s age progresses, its organ function declines, increasing its susceptibility to many chronic diseases, and putting immense pressure on the monetary power of countries in healthcare and pension. The lifespan of humans has increased rapidly, with the advancement in medical technology and improved living conditions; but it has not led to an increase in the health span. This is the primary reason to study, investigate, and understand the various aspects of aging and devise interventions to potentially benefit individuals, and play a part in reducing the socio-economic burden of countries with high percentages of an aged population.

    Aging is a time-dependent and irreversible pathophysiological phenomenon, accompanied by the development of numerous age-related diseases, such as type 2 diabetes, cardiovascular disease, cancer, Alzheimer’s disease, Parkinson’s disease, musculoskeletal problems, etc. [4]. Aging can also be referred to as an inclusive term for a number of molecular, biochemical, physiological, genetic, and epigenetic processes that have been described as hallmarks of aging [5, 6]. Interestingly, these hallmarks of aging are decentralized and interdependent on one another. Over the years, as our understanding of the biochemical, morphological, and functional decline of various processes related to aging has increased, efforts are underway to come up with strategies and interventions to ameliorate the effects of age-related diseases. Dietary interventions such as intermittent fasting, calorie restriction (CR), incorporation of CR mimetics, with physical exercise, etc., have been used in animal models and humans to slow down aging by alleviating the effects of the processes falling under the category of hallmarks of aging. This review focuses on one emerging field of research, i.e., sirtuin activators, a group of small molecules that activate sirtuin proteins, leading to sirtuins being considered therapeutic targets for anti-aging and age-related diseases.

    Sirtuins in aging

    Sirtuins are a family of nicotine adenine dinucleotide (NAD+)-dependent class III histone deacetylases [7], which catalyze post-translational modification of histones and other proteins and play a prominent role in aging. Sirtuins are dependent on NAD+ for their activity and, thus, are activated during conditions of low energy, like fasting or exercise. In 1999, it was reported that a sirtuin protein could extend the lifespan of Saccharomyces cerevisiae [8]. Sirtuins play a major role in energy metabolism, DNA repair, inflammation, cell survival, apoptosis, cellular senescence [9], oxidative stress, and mitochondrial production [10] and can be modulated by diet and exercise.

    In mammals, seven sirtuin family members have been detected, namely Sirt1–Sirt7. Although, the seven members have an identical highly conserved catalytic core domain of 275 amino acids, their localization within the cellular compartments varies, with Sirt1, 2, 6, and 7 localized in the nucleus, Sirt3, 4, and 5 in mitochondria, and Sirt1, 2 in the cytoplasm [11]. In addition to deacetylase, sirtuins also possess different enzymatic activity [12]. Some of the target proteins of sirtuins include NF-κB, phosphoglycerate mutase-1 (PGAM-1), sterol regulatory element-binding protein (SREBP), p53, peroxisome proliferator-activated receptor-γ co-activator 1α (PGC-1α), eNOS, mTORC2, and forkhead box protein O3a (FOXO3a).

    Sirt1

    Sirt1 is the most widely studied mammalian sirtuin as it is closely related to S. cerevisiae Sir2 and also for its role in aging, inflammation, and metabolic dysfunction. It is localized in the nucleus; however, it translocates to the cytoplasm, in response to certain stimuli [13]. It plays an important role in DNA damage recognition and response, and metabolism via deacetylation of target proteins. It deacetylates histones like H3, H4, and H1, along with modification of more than fifty non-histone proteins, including transcriptional factors p53, p65, PGC-1α, NF-κB, SREBP, and repair proteins. Sirt1, although its overexpression in transgenic mice leads to an increase in lifespan, the longevity aspect of the role of Sirt1 in aging, is implicated by the process of CR [14]. However, overexpression of Sirt1 specifically in the brain of both male and female mice, leads to an increase in lifespan [15]. This suggests that overexpression of Sirt1 in the brain will also have an impact on neurodegenerative diseases like Parkinson’s. For example, it has been observed that overexpression of Sirt1 causes a decline in amyloid plaques in several models of Alzheimer’s disease [16]. Although, most research for Sirt1 has been conducted in non-human model organisms, increased Sirt1 levels were found in muscle tissue of humans participating in CR [17].

    Sirt2

    Sirt2 is mainly localized in the cytoplasm, but it translocates to the nucleus. It moves to the nucleus during mitosis and plays a major role in the cell cycle. It regulates mitosis by controlling the activity of the anaphase-promoting complex/cyclosome. Sirt2 delays cell cycle progression, deacetylates p300 and tubulin, and also plays a role in reducing inflammation [18, 19]. Sirt2 promotes DNA repair and acts as a tumor suppressor by transcriptionally activating glycosylase OGG1 in an ATM/ATR-dependent manner [20]. It also contributes to the stabilization of p53 in the nucleus by regulating the acetylation state of p53 at lysine 382 and regulating the DNA damage response [21]. The kinetic and crystal structure study of Sirt2 also demonstrates a demyristoylase activity [22]. In kidneys, activated Sirt2 deacetylates FOXO3a during renal ischemia, promoting its nuclear shuffle and activating caspase-8 and caspase-3, thus inducing death [23].

    Sirt3

    Sirt3 is a mitochondrial sirtuin that promotes the transcription of superoxide dismutase 2 (SOD2) by directly deacetylating and activating SOD2, promoting the transcription of SOD2 and peroxisomes [24]. It induces PGC-1α and FOXO3a upregulation, leading to the restoration of manganese superoxide dismutase (MnSOD) activity and levels [25]. Sirt3 regulates glutamate dehydrogenase (GDH) [26] and acetyl-CoA synthetase 2 (ACS2), whose activity is regulated by reversible acetylation, as deacetylation activates the enzyme [27]. During prolonged fasting, Sirt3 deacetylates long-chain acyl-CoA dehydrogenase (LCAD), a protein involved in fatty acid oxidation, which activates fatty acid oxidation [28]. Sirt3, during CR, contributes to mitochondrial physiology by reprogramming the mitochondrial protein acetylome [29].

    Sirt4

    Sirt4 is a mitochondrial protein, lacking in in vitro deacetylase activity. In pancreatic beta cells, Sirt4 has been found to oppose the effects of CR, by actively inhibiting GDH [30]. Sirt4 also regulates ATP homeostasis by improving the efficacy of ATP via the adenine nucleotide translocator 2 (ANT2) and maintains retrograde signaling. Sirt4 overexpression is associated with increased ATP levels, and loss of Sirt4 expression leads to decreased cellular ATP levels in vitro and in vivo [31]. Overexpression of Sirt4 protects the organism by reducing the expression of proteins, such as Bax and phosphorylated p38, and upregulated Bcl-2 expression. It causes a decline in inflammatory factors, such as tumor necrosis factor alpha (TNF-α), interleukin (IL)-1β, and IL-6 [32].

    Sirt5

    Sirt5 is a mitochondrial enzyme, involved in metabolism and stress response, regulated largely by two proteins PGC-1α and AMP-activated protein kinase (AMPK). Overexpression of PGC-1α elevates cellular Sirt5 levels, while activation of AMPK down-regulates Sirt5 levels, playing a role in mitochondrial energy metabolism [33]. Sirt5-deficient mice exhibit defective energy metabolism and a decline in ATP production [34]. Overexpression of Sirt5 enhances AMPK phosphorylation and attenuates mitochondrial dysfunction, by alleviation of mitochondrial structural damage and restoration of ATP molecules [35]. In mice, Sirt5 desuccinylates metabolic enzymes, maintaining energy homeostasis and protecting mitochondrial metabolism [36].

    Sirt6

    Sirt6 is a nuclear sirtuin, with three catalytic enzyme activities—ADP-ribosyltransferase, deacetylase, and deacylase activity. Mice deficient in Sirt6 protein depict genomic instability and premature aging [37]. Sirt6 maintains the chromatin structure at telomeres by deacetylation of histone H3 lysine 9, preventing telomeric DNA damage and cellular senescence [38]. Sirt6 and Sirt1 act synergistically, where Sirt1 deacetylates Sirt6 on residue K33, and deacetylated Sirt6 remodels the chromatin [39]. CR and nutrient availability have a positive impact on Sirt6, as it is upregulated during CR, resulting in the expression of genes involved in energy metabolism, oxidative stress, autophagy, and inflammation [40]. Sirt6 also deacetylates non-histone proteins in the nucleus and cytoplasm, including members of the FOXO family, p53, and nicotinamide phosphoribosyl transferase (NAMPT) [41]. Genetic overexpression of Sirt6, in mice partially prolongs lifespan by restoring energy homeostasis and blocking insulin-like growth factor-1 (IGF-1)/AKT signaling [4244]. Sirt6 also represses LINE1 retrotransposons by mono-ADP ribosylating the nuclear corepressor protein KAP1; however, with the increase in age of the mice and DNA damage, Sirt6 levels decrease, and the silenced retroelements are activated [45]. In a study by Pan et al. [46], it was shown that Sirt6 also plays a protective role by reducing oxidative stress.

    Sirt7

    Sirt7 is a nuclear sirtuin, involved in the regulation of RNA polymerase I (Pol I) transcriptional machinery in mammalian cells [47]. It plays an active role in ribosome biogenesis, metabolic homeostasis, mitochondrial biogenesis, DNA repair, inflammation, and apoptosis [48]. Mice deficient in Sirt7 have a decrease in lifespan and develop inflammatory cardiomyopathy [49, 50]. Sirt7 mediates DNA damage repair by deacetylation of ATM protein and inhibiting it from activation [51]. Overexpression of Sirt7 in hypertensive mice promotes Krüppel-like factor 15/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling, indicating that Sirt7 is a promising target for the treatment of injured kidneys [52]. It has been reported that overexpression of Sirt7 in the hippocampal region in aged mice improves neurogenesis and cognitive functions [53].

    The cellular localization, a few examples of target proteins, and their functions are summarized in Table 1.

    Summary of cellular localization, target proteins, and functions of sirtuins in aging

    SirtuinCellular localizationMolecular weight (kDa)Enzymatic activityTarget proteinsFunctionReferences
    Sirt1Nuclear
    Cytoplasmic
    81.7Deacetylation
    Decrotonylation
    Delactylation
    H1, H3, H4, LKB1, AMPK, MnSOD, NF-κB, FOXO, PGC-1α, mTORDNA repair, insulin secretion, glucose metabolism, neuroprotection[7, 54, 55]
    Sirt2Cytoplasmic
    Nuclear
    43.2Deacetylation
    Demyristoylation
    H4K16, α-tubulin, FOXO, p53Cell cycle regulation, DNA repair[22, 56]
    Sirt3Mitochondrial
    Nuclear
    Cytoplasmic
    43.6Deacetylation
    Decrotonylation
    Delactylation
    H3, H4, FOXO, MnSOD, catalase, p53
    GDH
    ATP production, regulation of mitochondrial metabolism[26, 27, 54, 55]
    Sirt4Mitochondrial35.2ADP-ribosylation
    Deacylation
    Deacetylation
    AMPK, Glutamate dehydrogenase malonyl-CoA decarboxylaseInsulin secretion, fatty acid oxidation, apoptosis[30, 57, 58]
    Sirt5Mitochondrial33.9Deacetylation
    Demalonyation
    Desuccinylation
    Deglutarylation
    SOD, Succinate dehydrogenaseFatty acid oxidation, oxidative stress[36, 5961]
    Sirt6Nuclear39.1ADP-ribosylation Demyristoylation
    Deacetylation
    Depalmitoylation
    H2B, H3, FOXO, PARP1, NF-κB, IGF-1/AKTDNA repair, genome stability, telomere maintenance, stress response, energy homeostasis[38, 45, 62, 63]
    Sirt7Nucleolar44.8Deacetylation
    Desuccinylation
    ADP-ribosylation
    H2A, H2B, H3, FOXO3a, RNA polymerase-1, PARP-1, p53, ELK-4Regulation of rRNA transcription, stress resistance, DNA repair[6466]
    Display full size

    AMPK: AMP-activated protein kinase; MnSOD: manganese superoxide dismutase; FOXO: forkhead box protein O; PGC-1α: peroxisome proliferator-activated receptor-γ co-activator-1α; GDH: glutamate dehydrogenase; SOD: superoxide dismutase; IGF-1: insulin-like growth factor-1

    Sirtuin activating compounds

    The mammalian sirtuins are involved in several biological processes and overexpression of sirtuins plays an important role in increasing lifespan in model organisms. Sirtuins possess the potential to become markers and therapeutic targets for the management of aging and other age-related diseases. Dietary restriction is the only intervention known to increase lifespan in model organisms [67] and primates to a certain extent. The beneficial effects of CR are mediated through the nutrient sensing proteins, and sirtuins being one of the proteins, its activators can play an enormous role in therapeutics. Other than targeting the core catalytic subunit, sirtuins can be modulated by targeting the N- and C-terminal of the proteins, to enhance their enzymatic activity. With the advancement in research in this field, various small molecules, both natural and chemically synthesized compounds, have emerged as activators of sirtuins that can alleviate the conditions associated with aging and age-related diseases.

    Resveratrol

    Resveratrol is one of the first compounds discovered to mimic CR and activate sirtuins. Resveratrol (3,5,4ʹ-trihydroxy-trans-stilbene) is a naturally occurring plant polyphenol and a phytoalexin, of the stilbene family. It is found in plants such as grapes, blueberries, apples, plums, peanuts, and grape products such as red wine. It interacts with a wide variety of proteins, including Nrf2 and sirtuins [68]. Resveratrol and related polyphenolic compounds have antioxidative properties, thus possessing health-enhancing effects [69]. In the year 2003, it was reported that resveratrol, as a direct activator of sirtuins, extended the lifespan of S. cerevisiae [70]. Subsequent studies were also performed in other model organisms like Drosophila melanogaster, mice, etc. Resveratrol also modulates Sirt3 and Sirt5’s deacetylase activity [71]. Resveratrol upregulates the expression of antioxidant encoding genes and neuroprotection factors, Sirt1 and Sirt3 in immortalized lymphocytes of Alzheimer’s patients [72]. In the hippocampal area, Sirt1 activation by resveratrol produces neural malleability [73]. In 2020, Ma et al. [74] showed that resveratrol significantly increased Sirt1 expression in a rat model of diabetes with concomitant Alzheimer’s disease. The study also demonstrated increased acetylcholinesterase, malondialdehyde, and reduced SOD and glutathione levels.

    A close interaction occurs between the energy sensors, AMPK, and Sirt1 [75]. It regulates AMPK and Sirt1 in a dose-dependent and reciprocal manner. Resveratrol, in low doses, activates Sirt1, leading to deacetylation of liver kinase B1, which acts as an upstream kinase of AMPK, thus activating it. An analysis of gene expression in streptozotocin-induced diabetic rats showed that resveratrol supplementation balanced the hippocampal gene expression, involved in the neuronal origin and synaptic malleability (Hdac4, Hat1, Wnt7a, ApoE) and decline in expression of Jak-Stat pro-inflammatory signaling (IL-15, IL-22) [76]. The effect of resveratrol pretreatment in rats on NLRP3 inflammasome and ROS may also be Sirt1 dependent [77].

    However, the role of resveratrol in activating Sirt1 is ambiguous and needs further investigation [66]. In initial studies, activation of Sirt1 by resveratrol is dependent on the presence of a fluorophore, Fluor-de-Lys peptide substrate, as removal of the fluorophore failed to deacetylate the same peptide [78, 79].

    Curcumin

    Curcumin is a bright yellow compound, derived from the rhizome of the turmeric plant. It is a hydrophobic polyphenol with low bioavailability in humans [80]. Its supplementation in lower concentrations shows anti-aging effects, however, higher concentrations can trigger senescence [81]. It also acts as an activator of Sirt1 and is known to stimulate sirtuins [82, 83]. The down-regulation of Sirt1 after myocardial infarction was attenuated by curcumin pretreatment, which indicated that the activation of Sirt1 might be activated by curcumin [84]. Curcumin pretreatment also protects against a range of neurological disorders like stroke by activation of Sirt1 [85, 86]. Increased level/activity of AMPK and Sirt1 was observed in muscles of mice and rats, post curcumin supplementation [87, 88] enhancing the effect of exercise. Activation of Sirt1 by curcumin blocks the neurotoxicity of amyloid‐β25–35 in rat cortical neurons [85]. In a rat model of chronic obstructive pulmonary disease, curcumin supplementation upregulated Sirt3 expression and attenuated inflammatory markers and oxidative stress [89]. Curcumin reduced oxidative stress and protected the kidneys by activating Sirt1, Sirt3, and Sirt4 in cisplatin-induced toxicity in male rats [90]. The protective role of curcumin in aging and age-related pathologies is mediated by activated sirtuins, along with its anti-inflammatory and antioxidant effects.

    Pterostilbene

    Pterostilbene is a dimethyl ether resveratrol analogue, safe for human consumption with a high bioavailability [91]. It has been reported to increase lifespan in model organisms due to its anti-oxidative and anti-inflammatory properties [92, 93]. In recent studies, pterostilbene was found to activate sirtuins, AMPK and Nrf2 [94, 95]. In an obesity mice model, pterostilbene enhances thermogenesis and mitochondrial biogenesis by activating the Sirt1/PGC-1α/Sirt3 pathway [96]. It has also been reported that pterostilbene activates the Sirt1-FOXO1/p53 signaling pathway and decreases the apoptotic ratio in skeletal muscle cells induced by ischemia-reperfusion injury [97].

    Other natural compounds activating sirtuins

    There are some other natural compounds that are known to activate sirtuins. These include flavonols like fisetin, quercetin, apigenin, epigallocatechin gallate (EGCG), etc., along with chalconesbutein and isoliquiritigenin, the stilbene piceatannol. The chemical structures of some of the natural compounds activating sirtuins are shown in Figure 1. These compounds increase the lifespan of yeast by activating sirtuins [98]. Fisetin also activates sirtuin deacetylase activity in Drosophila melanogaster S2 cells, and a 23% extension of the lifespan was observed when the Drosophila melanogaster was fed on a fisetin-rich diet [99]. In mice, honokiol was found to reverse cardiac hypertrophy by activating Sirt3 [100]. Quercetin and its derivatives at high concentrations activate Sirt6 by binding to the Sirt6-selective acyl binding channel [101]. Polyphenols like cyanidin and luteolin also activate Sirt6 [102]. Fisetin and luteolin suppresses oxidative stress by activating sirtuins like Sirt1, Sirt3, and Sirt6 and reducing expression of FOXO3a [103].

    Chemical structures of naturally occurring sirtuin activators

    Synthetic sirtuin activators

    The natural activators of sirtuins like resveratrol, although they gained popularity initially, did not show promising results in clinical trials as they accompanied a significant amount of side effects [104]. This led researchers to explore effective and alternative compounds as sirtuin activators. A number of compounds were established as the first generation of synthetic Sirt1 activators, including SRT1720, SRT2183, and SRT1460, and are derived from the imidazo[1,2-b]thiazole core structure. However, further studies based on these molecules led to a diversion from the original idea [105].

    The second generation of synthetic Sirt1 activators was represented by two molecules SRT2104 and SRT3025. Out of the two, SRT2104 was identified via screening of 29,000 compounds. On the basis of its biochemical activity and pharmacokinetic characteristics, SRT2104 was selected as a potential drug candidate. SRT2104 was picked up by GlaxoSmithKline undergoing clinical trials. The results of the trials and its role as a potential agonist is described in the paper reviewed by Chang et al. [106]. The structures of synthetic Sirt1 activators are shown in Figure 2.

    Chemical structures of first and second generation of synthetic activators of Sirt1

    Synthetic activators of Sirt1 are widely studied, however, activation of the other mammalian sirtuins is not much reported. Interestingly, with the development in research, studies are being performed to find compounds that can activate and modulate other sirtuins. A prominent example of this is MDL-800, which has been found to activate Sirt6 by binding to an allosteric site and promoting histones H3K9 and H3K56 deacetylation in human hepatocellular carcinoma cells [107]. MDL-800 is reported to counteract age-related decline in DNA repair in murine-derived cells [108]. A series of novel pyrrolo[1,2-a]quinoxaline-based derivatives has been identified as potent selective activators of Sirt6 by repressing LPS-induced proinflammatory cytokine/chemokine secretion [109].

    NAD+ boosting molecules

    Sirtuins can be activated by regulating the cellular level of NAD+. Two approaches can be followed to modulate NAD+ levels; direct restoration of NAD+ levels by NAD+ precursor supplementation, and overexpression of enzymes related to NAD+ synthesis, NAMPT, and nicotinamide mononucleotide adenyltransferase (NMNAT), to enhance the rate of NAD+ synthesis or by inhibiting CD38 NAD+ hydrolase [110]. The NAD+ precursor supplements include nicotinamide mononucleotide (NMN) and nicotinamide riboside (NR) [111, 112]. A recent class of sirtuin activators called NAD+ boosting molecules is gaining popularity as a technique to restore NAD+ levels in aged individuals and potentially activate all seven sirtuins by a single molecule. NMN supplementation for a long time mitigates physiological age-associated decline in mice by improving energy metabolism and physical activity [113]. NMN supplementation has partially restored skeletal muscle loss and, also protects against β-amyloid oligomer-induced cognitive impairment and neuronal death in an Alzheimer’s model of mice [114, 115].

    Molecular docking as a tool for studying sirtuin activators

    Much insight into the mechanism of action and drug discovery today comes about due to the use of computational methods. Molecular docking and dynamics simulations are increasingly used to predict how molecules interact with their targets and predict ligand-target interactions [116, 117]. These techniques reduce the need for extensive laboratory testing, accelerating the drug development process. Docking studies have highlighted the potential of bioactive compounds from Curcuma zanthorrhiza, such as curcumin, as promising candidates for reducing age-related diseases through the Sirt1/NF-κB signaling pathway [118]. Additionally, research on resveratrol has demonstrated its role in stabilizing Sirt1/peptide interactions, offering insights for designing new substrate-specific Sirt1 modulators [119]. In silico approaches to discover bioactive compounds such as Sirt1 activators and NF-κB inhibitors, evaluating their toxicity and potential therapeutic benefits, have also been shown for metformin an important anti-diabetic drug. Analysis of metformin’s interaction with Sirt1 through molecular docking and dynamic simulation provides insights into its binding patterns and potential as a Sirt1 agonist [120].

    Conclusions

    With the advancement in studies related to sirtuin activators, the field of biogerontology is stepping into a new world of possibilities. It has great potential for future pharmacological interventions to slow aging. The family of mammalian sirtuins has risen to prominence in the field of aging research since the discovery of the yeast Sir2. Studies accumulating over the years have revealed that sirtuins exert protective functions against aging and age-related pathologies. Although the localization of sirtuins varies at the cellular level, their unique functions lead them to a common goal of promoting and preventing aging, marking them as promising targets for developing interventions for several age-related diseases. Since the discovery of resveratrol as an activator of sirtuins, efforts have been made by researchers to put forward certain molecules, which can activate all seven members of the sirtuin family and alleviate the features associated with aging. A small number of natural compounds like curcumin, fisetin, etc., have proved to be beneficial only up to a limited extent. Chemically synthesized molecules like SRT2104 showed promising results initially, and are undergoing clinical trials for various age-related diseases like diabetes, neurodegeneration, Parkinson’s, etc. However, an area of concern arises in the form of safety for humans. There are conflicting data in vitro, leading to assumptions that Sirt1 may act as an oncogene, despite the other health benefits. Also, a thorough understanding of the role of all isoforms of sirtuins is necessary before designing activators that target them, as a lapse in understanding the role of sirtuins may have a negative impact on other related molecules and processes.

    Abbreviations

    AMPK:

    AMP-activated protein kinase

    CR:

    calorie restriction

    FOXO3a:

    forkhead box protein O3a

    GDH:

    glutamate dehydrogenase

    IGF-1:

    insulin-like growth factor-1

    IL:

    interleukin

    NAD+:

    nicotine adenine dinucleotide

    NAMPT:

    nicotinamide phosphoribosyl transferase

    NMN:

    nicotinamide mononucleotide

    Nrf2:

    nuclear factor erythroid 2-related factor 2

    PGC-1α:

    peroxisome proliferator-activated receptor-γ co-activator 1α

    SOD2:

    superoxide dismutase 2

    SREBP:

    sterol regulatory element-binding protein

    Declarations

    Acknowledgments

    The authors thank the Department of Biochemistry, North-Eastern Hill University, Shillong, for support of research facilities under UGC-SAP, DBT, and DST-FIST, New Delhi.

    Author contributions

    PS: Conceptualization, Investigation, Writing—original draft, Writing—review & editing, Visualization. AKR: Investigation, Writing—original draft, Writing—review & editing. DS: Conceptualization, Validation, Writing—review & editing, Supervision. The authors read and approved the submitted version.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    The study was supported by the Department of Biochemistry, North-Eastern Hill University, Shillong, under UGC-SAP, DBT, and DST-FIST, New Delhi. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

    Copyright

    © The Author(s) 2025.

    Publisher’s note

    Open Exploration maintains a neutral stance on jurisdictional claims in published institutional affiliations and maps. All opinions expressed in this article are the personal views of the author(s) and do not represent the stance of the editorial team or the publisher.

    References

    Peak global population and other key findings from the 2024 UN World Population Prospects [Internet]. Our World in Data; c2024 [cited 2024 Jul 29]. Available from: https://ourworldindata.org/un-population-2024-revision
    Ageing and health [Internet]. WHO; c2024 [cited 2024 Oct 1]. Available from: https://www.who.int/news-room/fact-sheets/detail/ageing-and-health
    de Magalhães JP, Stevens M, Thornton D. The Business of Anti-Aging Science. Trends Biotechnol. 2017;35:106273. [DOI] [PubMed]
    Li Z, Zhang Z, Ren Y, Wang Y, Fang J, Yue H, et al. Aging and age-related diseases: from mechanisms to therapeutic strategies. Biogerontology. 2021;22:16587. [DOI] [PubMed] [PMC]
    López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The Hallmarks of Aging. Cell. 2013;153:1194217. [DOI] [PubMed] [PMC]
    López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: An expanding universe. Cell. 2023;186:24378. [DOI] [PubMed]
    Imai S, Armstrong CM, Kaeberlein M, Guarente L. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature. 2000;403:795800. [DOI] [PubMed]
    Kaeberlein M, McVey M, Guarente L. The SIR2/3/4 complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms. Genes Dev. 1999;13:257080. [DOI] [PubMed] [PMC]
    Bi S, Jiang X, Ji Q, Wang Z, Ren J, Wang S, et al. The sirtuin-associated human senescence program converges on the activation of placenta-specific gene PAPPA. Dev Cell. 2024;59:9911009.e12. [DOI] [PubMed]
    Bonkowski MS, Sinclair DA. Slowing ageing by design: the rise of NAD+ and sirtuin-activating compounds. Nat Rev Mol Cell Biol. 2016;17:67990. [DOI] [PubMed] [PMC]
    Frye RA. Phylogenetic Classification of Prokaryotic and Eukaryotic Sir2-like Proteins. Biochem Biophys Res Commun. 2000;273:7938. [DOI] [PubMed]
    Zhao L, Cao J, Hu K, He X, Yun D, Tong T, et al. Sirtuins and their Biological Relevance in Aging and Age-Related Diseases. Aging Dis. 2020;11:92745. [DOI] [PubMed] [PMC]
    Manjula R, Anuja K, Alcain FJ. SIRT1 and SIRT2 Activity Control in Neurodegenerative Diseases. Front Pharmacol. 2021;11:585821. [DOI] [PubMed] [PMC]
    Chen D, Steele AD, Lindquist S, Guarente L. Increase in Activity During Calorie Restriction Requires Sirt1. Science. 2005;310:1641. [DOI] [PubMed]
    Satoh A, Brace CS, Rensing N, Cliften P, Wozniak DF, Herzog ED, et al. Sirt1 Extends Life Span and Delays Aging in Mice through the Regulation of Nk2 Homeobox 1 in the DMH and LH. Cell Metab. 2013;18:41630. [DOI] [PubMed] [PMC]
    Donmez G, Outeiro TF. SIRT1 and SIRT2: emerging targets in neurodegeneration. EMBO Mol Med. 2013;5:34452. [DOI] [PubMed] [PMC]
    Civitarese AE, Carling S, Heilbronn LK, Hulver MH, Ukropcova B, Deutsch WA, et al.; CALERIE Pennington Team. Calorie Restriction Increases Muscle Mitochondrial Biogenesis in Healthy Humans. PLoS Med. 2007;4:e76. [DOI] [PubMed] [PMC]
    Serrano L, Martínez-Redondo P, Marazuela-Duque A, Vazquez BN, Dooley SJ, Voigt P, et al. The tumor suppressor SirT2 regulates cell cycle progression and genome stability by modulating the mitotic deposition of H4K20 methylation. Genes Dev. 2013;27:63953. [DOI] [PubMed] [PMC]
    Rothgiesser KM, Erener S, Waibel S, Lüscher B, Hottiger MO. SIRT2 regulates NF-κB-dependent gene expression through deacetylation of p65 Lys310. J Cell Sci. 2010;123:42518. [DOI] [PubMed]
    Geng A, Sun J, Tang H, Yu Y, Wang X, Zhang J, et al. SIRT2 promotes base excision repair by transcriptionally activating OGG1 in an ATM/ATR-dependent manner. Nucleic Acids Res. 2024;52:510720. [DOI] [PubMed] [PMC]
    Agborbesong E, Zhou JX, Li LX, Harris PC, Calvet JP, Li X. Prdx5 regulates DNA damage response through autophagy-dependent Sirt2-p53 axis. Hum Mol Genet. 2023;32:56779. [DOI] [PubMed] [PMC]
    Teng YB, Jing H, Aramsangtienchai P, He B, Khan S, Hu J, et al. Efficient Demyristoylase Activity of SIRT2 Revealed by Kinetic and Structural Studies. Sci Rep. 2015;5:8529. [DOI] [PubMed] [PMC]
    Wang Y, Mu Y, Zhou X, Ji H, Gao X, Cai WW, et al. SIRT2-mediated FOXO3a deacetylation drives its nuclear translocation triggering FasL-induced cell apoptosis during renal ischemia reperfusion. Apoptosis. 2017;22:51930. [DOI] [PubMed]
    Zhang Y, Liu Y, Hou M, Xia X, Liu J, Xu Y, et al. Reprogramming of Mitochondrial Respiratory Chain Complex by Targeting SIRT3-COX4I2 Axis Attenuates Osteoarthritis Progression. Adv Sci (Weinh). 2023;10:e2206144. [DOI] [PubMed] [PMC]
    Anamika, Roy A, Trigun SK. Hippocampus mitochondrial MnSOD activation by a SIRT3 activator, honokiol, correlates with its deacetylation and upregulation of FoxO3a and PGC1α in a rat model of ammonia neurotoxicity. J Cell Biochem. 2023;124:60618. [DOI] [PubMed]
    Schlicker C, Gertz M, Papatheodorou P, Kachholz B, Becker CF, Steegborn C. Substrates and Regulation Mechanisms for the Human Mitochondrial Sirtuins Sirt3 and Sirt5. J Mol Biol. 2008;382:790801. [DOI] [PubMed]
    Hallows WC, Lee S, Denu JM. Sirtuins deacetylate and activate mammalian acetyl-CoA synthetases. Proc Natl Acad Sci U S A. 2006;103:102305. [DOI] [PubMed] [PMC]
    Hirschey MD, Shimazu T, Goetzman E, Jing E, Schwer B, Lombard DB, et al. SIRT3 regulates mitochondrial fatty-acid oxidation by reversible enzyme deacetylation. Nature. 2010;464:1215. [DOI] [PubMed] [PMC]
    Hebert AS, Dittenhafer-Reed KE, Yu W, Bailey DJ, Selen ES, Boersma MD, et al. Calorie Restriction and SIRT3 Trigger Global Reprogramming of the Mitochondrial Protein Acetylome. Mol Cell. 2013;49:18699. [DOI] [PubMed] [PMC]
    Haigis MC, Mostoslavsky R, Haigis KM, Fahie K, Christodoulou DC, Murphy AJ, et al. SIRT4 Inhibits Glutamate Dehydrogenase and Opposes the Effects of Calorie Restriction in Pancreatic β Cells. Cell. 2006;126:94154. [DOI] [PubMed]
    Ho L, Titus AS, Banerjee KK, George S, Lin W, Deota S, et al. SIRT4 regulates ATP homeostasis and mediates a retrograde signaling via AMPK. Aging (Albany NY). 2013;5:83549. [DOI] [PubMed] [PMC]
    Shi JX, Wang QJ, Li H, Huang Q. SIRT4 overexpression protects against diabetic nephropathy by inhibiting podocyte apoptosis. Exp Ther Med. 2017;13:3428. [DOI] [PubMed] [PMC]
    Buler M, Aatsinki SM, Izzi V, Uusimaa J, Hakkola J. SIRT5 is under the control of PGC-1α and AMPK and is involved in regulation of mitochondrial energy metabolism. FASEB J. 2014;28:322537. [DOI] [PubMed]
    Zhang Y, Bharathi SS, Rardin MJ, Lu J, Maringer KV, Sims-Lucas S, et al. Lysine desuccinylase SIRT5 binds to cardiolipin and regulates the electron transport chain. J Biol Chem. 2017;292:1023949. [DOI] [PubMed] [PMC]
    Wang T, Lin B, Qiu W, Yu B, Li J, An S, et al. Adenosine monophosphate-activated protein kinase phosphorylation mediated by sirtuin 5 alleviates septic acute kidney injury. Shock. 2023;59:47785. [DOI] [PubMed]
    Xiao ZP, Lv T, Hou PP, Manaenko A, Liu Y, Jin Y, et al. Sirtuin 5-Mediated Lysine Desuccinylation Protects Mitochondrial Metabolism Following Subarachnoid Hemorrhage in Mice. Stroke. 2021;52:404353. [DOI] [PubMed]
    Mostoslavsky R, Chua KF, Lombard DB, Pang WW, Fischer MR, Gellon L, et al. Genomic Instability and Aging-like Phenotype in the Absence of Mammalian SIRT6. Cell. 2006;124:31529. [DOI] [PubMed]
    Michishita E, McCord RA, 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;452:4926. [DOI] [PubMed] [PMC]
    Meng F, Qian M, Peng B, Peng L, Wang X, Zheng K, et al. Synergy between SIRT1 and SIRT6 helps recognize DNA breaks and potentiates the DNA damage response and repair in humans and mice. Elife. 2020;9:e55828. [DOI] [PubMed] [PMC]
    Kanfi Y, Shalman R, Peshti V, Pilosof SN, Gozlan YM, Pearson KJ, et al. Regulation of SIRT6 protein levels by nutrient availability. FEBS Lett. 2008;582:5438. [DOI] [PubMed] [PMC]
    Ren SC, Chen X, Gong H, Wang H, Wu C, Li PH, et al. SIRT6 in Vascular Diseases, from Bench to Bedside. Aging Dis. 2022;13:101529. [DOI] [PubMed] [PMC]
    Roichman A, Elhanati S, Aon MA, Abramovich I, Di Francesco A, Shahar Y, et al. Restoration of energy homeostasis by SIRT6 extends healthy lifespan. Nat Commun. 2021;12:3208. [DOI] [PubMed] [PMC]
    Sundaresan NR, Vasudevan P, Zhong L, Kim G, Samant S, Parekh V, et al. The sirtuin SIRT6 blocks IGF-Akt signaling and development of cardiac hypertrophy by targeting c-Jun. Nat Med. 2012;18:164350. [DOI] [PubMed] [PMC]
    Kanfi Y, Naiman S, Amir G, Peshti V, Zinman G, Nahum L, et al. The sirtuin SIRT6 regulates lifespan in male mice. Nature. 2012;483:21821. [DOI] [PubMed]
    Van Meter M, Kashyap M, Rezazadeh S, Geneva AJ, Morello TD, Seluanov A, et al. SIRT6 represses LINE1 retrotransposons by ribosylating KAP1 but this repression fails with stress and age. Nat Commun. 2014;5:5011. [DOI] [PubMed] [PMC]
    Pan H, Guan D, Liu X, Li J, Wang L, Wu J, et al. SIRT6 safeguards human mesenchymal stem cells from oxidative stress by coactivating NRF2. Cell Res. 2016;26:190205. [DOI] [PubMed] [PMC]
    Grummt I, Pikaard CS. Epigenetic silencing of RNA polymerase I transcription. Nat Rev Mol Cell Biol. 2003;4:6419. [DOI] [PubMed]
    Yi X, Wang H, Yang Y, Wang H, Zhang H, Guo S, et al. SIRT7 orchestrates melanoma progression by simultaneously promoting cell survival and immune evasion via UPR activation. Signal Transduct Target Ther. 2023;8:107. [DOI] [PubMed] [PMC]
    Vakhrusheva O, Braeuer D, Liu Z, Braun T, Bober E. Sirt7-dependent inhibition of cell growth and proliferation might be instrumental to mediate tissue integrity during aging. J Physiol Pharmacol. 2008;59:20112. [PubMed]
    Vakhrusheva O, Smolka C, Gajawada P, Kostin S, Boettger T, Kubin T, et al. Sirt7 Increases Stress Resistance of Cardiomyocytes and Prevents Apoptosis and Inflammatory Cardiomyopathy in Mice. Circ Res. 2008;102:70310. [DOI] [PubMed]
    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:eaav1118. [DOI] [PubMed] [PMC]
    Li XT, Song JW, Zhang ZZ, Zhang MW, Liang LR, Miao R, et al. Sirtuin 7 mitigates renal ferroptosis, fibrosis and injury in hypertensive mice by facilitating the KLF15/Nrf2 signaling. Free Radic Biol Med. 2022;193:45973. [DOI] [PubMed]
    Wang CL, Ohkubo R, Mu WC, Chen W, Fan JL, Song Z, et al. The mitochondrial unfolded protein response regulates hippocampal neural stem cell aging. Cell Metab. 2023;35:9961008.e7. [DOI] [PubMed] [PMC]
    Bao X, Wang Y, Li X, Li XM, Liu Z, Yang T, et al. Identification of ‘erasers’ for lysine crotonylated histone marks using a chemical proteomics approach. Elife. 2014;3:e02999. [DOI] [PubMed] [PMC]
    Moreno-Yruela C, Zhang D, Wei W, Bæk M, Liu W, Gao J, et al. Class I histone deacetylases (HDAC1–3) are histone lysine delactylases. Sci Adv. 2022;8:eabi6696. [DOI] [PubMed] [PMC]
    Feldman JL, Baeza J, Denu JM. Activation of the Protein Deacetylase SIRT6 by Long-chain Fatty Acids and Widespread Deacylation by Mammalian Sirtuins. J Biol Chem. 2013;288:313506. [DOI] [PubMed] [PMC]
    Anderson KA, Green MF, Huynh FK, Wagner GR, Hirschey MD. SnapShot: Mammalian Sirtuins. Cell. 2014;159:956.e1. [DOI] [PubMed] [PMC]
    Laurent G, German NJ, Saha AK, de Boer VC, Davies M, Koves TR, et al. SIRT4 Coordinates the Balance between Lipid Synthesis and Catabolism by Repressing Malonyl CoA Decarboxylase. Mol Cell. 2013;50:68698. [DOI] [PubMed] [PMC]
    Roessler C, Nowak T, Pannek M, Gertz M, Nguyen GT, Scharfe M, et al. Chemical Probing of the Human Sirtuin 5 Active Site Reveals Its Substrate Acyl Specificity and Peptide-Based Inhibitors. Angew Chem Int Ed Engl. 2014;53:1072832. [DOI] [PubMed]
    Du J, Zhou Y, Su X, Yu JJ, Khan S, Jiang H, et al. Sirt5 Is a NAD-Dependent Protein Lysine Demalonylase and Desuccinylase. Science. 2011;334:8069. [DOI] [PubMed] [PMC]
    Tan M, Peng C, Anderson KA, Chhoy P, Xie Z, Dai L, et al. Lysine Glutarylation Is a Protein Posttranslational Modification Regulated by SIRT5. Cell Metab. 2014;19:60517. [DOI] [PubMed] [PMC]
    Tasselli L, Zheng W, Chua KF. SIRT6: Novel Mechanisms and Links to Aging and Disease. Trends Endocrinol Metab. 2017;28:16885. [DOI] [PubMed] [PMC]
    Jiang H, Khan S, Wang Y, Charron G, He B, Sebastian C, et al. SIRT6 regulates TNF-α secretion through hydrolysis of long-chain fatty acyl lysine. Nature. 2013;496:1103. [DOI] [PubMed] [PMC]
    Barber MF, Michishita-Kioi E, Xi Y, Tasselli L, Kioi M, Moqtaderi Z, et al. SIRT7 links H3K18 deacetylation to maintenance of oncogenic transformation. Nature. 2012;487:1148. [DOI] [PubMed] [PMC]
    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] [PubMed] [PMC]
    Mitra N, Dey S. Biochemical characterization of mono ADP ribosyl transferase activity of human sirtuin SIRT7 and its regulation. Arch Biochem Biophys. 2020;680:108226. [DOI] [PubMed]
    Ingram DK, Roth GS. Calorie restriction mimetics: Can you have your cake and eat it, too? Ageing Res Rev. 2015;20:4662. [DOI] [PubMed]
    Britton RG, Kovoor C, Brown K. Direct molecular targets of resveratrol: identifying key interactions to unlock complex mechanisms. Ann N Y Acad Sci. 2015;1348:12433. [DOI] [PubMed]
    Baur JA, Sinclair DA. Therapeutic potential of resveratrol: the in vivo evidence. Nat Rev Drug Discov. 2006;5:493506. [DOI] [PubMed]
    Howitz KT, Bitterman KJ, Cohen HY, Lamming DW, Lavu S, Wood JG, et al. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature. 2003;425:1916. [DOI] [PubMed]
    Gertz M, Nguyen GT, Fischer F, Suenkel B, Schlicker C, Fränzel B, et al. A Molecular Mechanism for Direct Sirtuin Activation by Resveratrol. PLoS One. 2012;7:e49761. [DOI] [PubMed] [PMC]
    Cosín-Tomàs M, Senserrich J, Arumí-Planas M, Alquézar C, Pallàs M, Martín-Requero Á, et al. Role of Resveratrol and Selenium on Oxidative Stress and Expression of Antioxidant and Anti-Aging Genes in Immortalized Lymphocytes from Alzheimer’s Disease Patients. Nutrients. 2019;11:1764. [DOI] [PubMed] [PMC]
    Hasegawa K, Yoshikawa K. Necdin Regulates p53 Acetylation via Sirtuin1 to Modulate DNA Damage Response in Cortical Neurons. J Neurosci. 2008;28:877284. [DOI] [PubMed] [PMC]
    Ma X, Sun Z, Han X, Li S, Jiang X, Chen S, et al. Neuroprotective Effect of Resveratrol via Activation of Sirt1 Signaling in a Rat Model of Combined Diabetes and Alzheimer’s Disease. Front Neurosci. 2020;13:1400. [DOI] [PubMed] [PMC]
    Cantó C, Gerhart-Hines Z, Feige JN, Lagouge M, Noriega L, Milne JC, et al. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature. 2009;458:105660. [DOI] [PubMed] [PMC]
    Thomas J, Garg ML, Smith DW. Dietary resveratrol supplementation normalizes gene expression in the hippocampus of streptozotocin-induced diabetic C57Bl/6 mice. J Nutr Biochem. 2014;25:3138. [DOI] [PubMed]
    Zou P, Liu X, Li G, Wang Y. Resveratrol pretreatment attenuates traumatic brain injury in rats by suppressing NLRP3 inflammasome activation via SIRT1. Mol Med Rep. 2018;17:32127. [DOI] [PubMed]
    Kaeberlein M, McDonagh T, Heltweg B, Hixon J, Westman EA, Caldwell SD, et al. Substrate-specific Activation of Sirtuins by Resveratrol. J Biol Chem. 2005;280:1703845. [DOI] [PubMed]
    Borra MT, Smith BC, Denu JM. Mechanism of Human SIRT1 Activation by Resveratrol. J Biol Chem. 2005;280:1718795. [DOI] [PubMed]
    Sahebkar A, Saboni N, Pirro M, Banach M. Curcumin: An effective adjunct in patients with statin-associated muscle symptoms? J Cachexia Sarcopenia Muscle. 2017;8:1924. [DOI] [PubMed] [PMC]
    Grabowska W, Mosieniak G, Achtabowska N, Czochara R, Litwinienko G, Bojko A, et al. Curcumin induces multiple signaling pathways leading to vascular smooth muscle cell senescence. Biogerontology. 2019;20:78398. [DOI] [PubMed] [PMC]
    Yang Y, Duan W, Lin Y, Yi W, Liang Z, Yan J, et al. SIRT1 activation by curcumin pretreatment attenuates mitochondrial oxidative damage induced by myocardial ischemia reperfusion injury. Free Radic Biol Med. 2013;65:66779. [DOI] [PubMed]
    Zendedel E, Butler AE, Atkin SL, Sahebkar A. Impact of curcumin on sirtuins: A review. J Cell Biochem. 2018;119:10291300. [DOI] [PubMed]
    Xiao J, Sheng X, Zhang X, Guo M, Ji X. Curcumin protects against myocardial infarction-induced cardiac fibrosis via SIRT1 activation in vivo and in vitro. Drug Des Devel Ther. 2016;10:126777. [DOI] [PubMed] [PMC]
    Sun Q, Jia N, Wang W, Jin H, Xu J, Hu H. Activation of SIRT1 by curcumin blocks the neurotoxicity of amyloid-β25–35 in rat cortical neurons. Biochem Biophys Res Commun. 2014;448:8994. [DOI] [PubMed]
    Miao Y, Zhao S, Gao Y, Wang R, Wu Q, Wu H, et al. Curcumin pretreatment attenuates inflammation and mitochondrial dysfunction in experimental stroke: The possible role of Sirt1 signaling. Brain Res Bull. 2016;121:915. [DOI] [PubMed]
    Huang WC, Chiu WC, Chuang HL, Tang DW, Lee ZM, Wei L, et al. Effect of Curcumin Supplementation on Physiological Fatigue and Physical Performance in Mice. Nutrients. 2015;7:90521. [DOI] [PubMed] [PMC]
    Ray Hamidie RD, Yamada T, Ishizawa R, Saito Y, Masuda K. Curcumin treatment enhances the effect of exercise on mitochondrial biogenesis in skeletal muscle by increasing cAMP levels. Metabolism. 2015;64:133447. [DOI] [PubMed]
    Zhang M, Tang J, Li Y, Xie Y, Shan H, Chen M, et al. Curcumin attenuates skeletal muscle mitochondrial impairment in COPD rats: PGC-1α/SIRT3 pathway involved. Chem Biol Interact. 2017;277:16875. [DOI] [PubMed]
    Ugur S, Ulu R, Dogukan A, Gurel A, Yigit IP, Gozel N, et al. The renoprotective effect of curcumin in cisplatin-induced nephrotoxicity. Ren Fail. 2015;37:3326. [DOI] [PubMed]
    Wang P, Sang S. Metabolism and pharmacokinetics of resveratrol and pterostilbene. Biofactors. 2018;44:1625. [DOI] [PubMed]
    Lange KW, Li S. Resveratrol, pterostilbene, and dementia. Biofactors. 2018;44:8390. [DOI] [PubMed]
    Li YR, Li S, Lin CC. Effect of resveratrol and pterostilbene on aging and longevity. Biofactors. 2018;44:6982. [DOI] [PubMed]
    Malik SA, Acharya JD, Mehendale NK, Kamat SS, Ghaskadbi SS. Pterostilbene reverses palmitic acid mediated insulin resistance in HepG2 cells by reducing oxidative stress and triglyceride accumulation. Free Radic Res. 2019;53:81527. [DOI] [PubMed] [PMC]
    Zhou J, Ci X, Ma X, Yu Q, Cui Y, Zhen Y, et al. Pterostilbene Activates the Nrf2-Dependent Antioxidant Response to Ameliorate Arsenic-Induced Intracellular Damage and Apoptosis in Human Keratinocytes. Front Pharmacol. 2019;10:497. [DOI] [PubMed] [PMC]
    Koh YC, Lin SJ, Hsu KY, Nagabhushanam K, Ho CT, Pan MH. Pterostilbene Enhances Thermogenesis and Mitochondrial Biogenesis by Activating the SIRT1/PGC-1α/SIRT3 Pathway to Prevent Western Diet-Induced Obesity. Mol Nutr Food Res. 2023;67:e2300370. [DOI] [PubMed]
    Cheng Y, Di S, Fan C, Cai L, Gao C, Jiang P, et al. SIRT1 activation by pterostilbene attenuates the skeletal muscle oxidative stress injury and mitochondrial dysfunction induced by ischemia reperfusion injury. Apoptosis. 2016;21:90516. [DOI] [PubMed]
    Davis JM, Murphy EA, Carmichael MD, Davis B. Quercetin increases brain and muscle mitochondrial biogenesis and exercise tolerance. Am J Physiol Regul Integr Comp Physiol. 2009;296:R10717. [DOI] [PubMed]
    Wood JG, Rogina B, Lavu S, Howitz K, Helfand SL, Tatar M, et al. Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Nature. 2004;430:6869. [DOI] [PubMed]
    Pillai VB, Samant S, Sundaresan NR, Raghuraman H, Kim G, Bonner MY, et al. Honokiol blocks and reverses cardiac hypertrophy in mice by activating mitochondrial Sirt3. Nat Commun. 2015;6:6656. [DOI] [PubMed] [PMC]
    You W, Zheng W, Weiss S, Chua KF, Steegborn C. Structural basis for the activation and inhibition of Sirtuin 6 by quercetin and its derivatives. Sci Rep. 2019;9:19176. [DOI] [PubMed] [PMC]
    Rahnasto-Rilla M, Tyni J, Huovinen M, Jarho E, Kulikowicz T, Ravichandran S, et al. Natural polyphenols as sirtuin 6 modulators. Sci Rep. 2018;8:4163. [DOI] [PubMed] [PMC]
    Kim A, Lee W, Yun JM. Luteolin and fisetin suppress oxidative stress by modulating sirtuins and forkhead box O3a expression under in vitro diabetic conditions. Nutr Res Pract. 2017;11:4304. [DOI] [PubMed] [PMC]
    Singh AP, Singh R, Verma SS, Rai V, Kaschula CH, Maiti P, et al. Health benefits of resveratrol: Evidence from clinical studies. Med Res Rev. 2019;39:185191. [DOI] [PubMed]
    Pacholec M, Bleasdale JE, Chrunyk B, Cunningham D, Flynn D, Garofalo RS, et al. SRT1720, SRT2183, SRT1460, and Resveratrol Are Not Direct Activators of SIRT1. J Biol Chem. 2010;285:834051. [DOI] [PubMed] [PMC]
    Chang N, Li J, Lin S, Zhang J, Zeng W, Ma G, et al. Emerging roles of SIRT1 activator, SRT2104, in disease treatment. Sci Rep. 2024;14:5521. [DOI] [PubMed] [PMC]
    Huang Z, Zhao J, Deng W, Chen Y, Shang J, Song K, et al. Identification of a cellularly active SIRT6 allosteric activator. Nat Chem Biol. 2018;14:111826. [DOI] [PubMed]
    Chen Y, Chen J, Sun X, Yu J, Qian Z, Wu L, et al. The SIRT6 activator MDL-800 improves genomic stability and pluripotency of old murine-derived iPS cells. Aging Cell. 2020;19:e13185. [DOI] [PubMed] [PMC]
    Xu J, Shi S, Liu G, Xie X, Li J, Bolinger AA, et al. Design, synthesis, and pharmacological evaluations of pyrrolo[1,2-a]quinoxaline-based derivatives as potent and selective sirt6 activators. Eur J Med Chem. 2023;246:114998. [DOI] [PubMed] [PMC]
    Escande C, Nin V, Price NL, Capellini V, Gomes AP, Barbosa MT, et al. Flavonoid Apigenin Is an Inhibitor of the NAD+ase CD38: Implications for Cellular NAD+ Metabolism, Protein Acetylation, and Treatment of Metabolic Syndrome. Diabetes. 2013;62:108493. [DOI] [PubMed] [PMC]
    Gong B, Pan Y, Vempati P, Zhao W, Knable L, Ho L, et al. Nicotinamide riboside restores cognition through an upregulation of proliferator-activated receptor-γ coactivator 1α regulated β-secretase 1 degradation and mitochondrial gene expression in Alzheimer’s mouse models. Neurobiol Aging. 2013;34:15818. [DOI] [PubMed] [PMC]
    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;352:143643. [DOI] [PubMed]
    Mills KF, Yoshida S, Stein LR, Grozio A, Kubota S, Sasaki Y, et al. Long-Term Administration of Nicotinamide Mononucleotide Mitigates Age-Associated Physiological Decline in Mice. Cell Metab. 2016;24:795806. [DOI] [PubMed] [PMC]
    Mendelsohn AR, Larrick JW. Partial Reversal of Skeletal Muscle Aging by Restoration of Normal NAD⁺ Levels. Rejuvenation Res. 2014;17:629. [DOI] [PubMed]
    Wang X, Hu X, Yang Y, Takata T, Sakurai T. Nicotinamide mononucleotide protects against β-amyloid oligomer-induced cognitive impairment and neuronal death. Brain Res. 2016;1643:19. [DOI] [PubMed]
    Muhammed MT, Kuyucuklu G, Kaynak-Onurdag F, Aki-Yalcin E. Synthesis, Antimicrobial Activity, and Molecular Modeling Studies of Some Benzoxazole Derivatives. Lett Drug Des Discov. 2022;19:75768. [DOI]
    Dar AM, Mir S. Molecular Docking: Approaches, Types, Applications and Basic Challenges. J Anal Bioanal Tech. 2017;8:13. [DOI]
    Prasetyawan S, Safitri A, Atho’illah MF, Rahayu S. Computational evaluation of bioactive compounds in Curcuma zanthorrhiza targeting SIRT1 and NFκB. BioTechnologia (Pozn). 2023;104:17182. [DOI] [PubMed] [PMC]
    Hou X, Rooklin D, Fang H, Zhang Y. Resveratrol serves as a protein-substrate interaction stabilizer in human SIRT1 activation. Sci Rep. 2016;6:38186. [DOI] [PubMed] [PMC]
    Cuyàs E, Verdura S, Llorach-Parés L, Fernández-Arroyo S, Joven J, Martin-Castillo B, et al. Metformin Is a Direct SIRT1-Activating Compound: Computational Modeling and Experimental Validation. Front Endocrinol (Lausanne). 2018;9:657. [DOI] [PubMed] [PMC]