Open access peer-reviewed chapter

Resveratrol and SIRT1 Activators for the Treatment of Aging and Age-Related Diseases

Written By

Alessandra Stacchiotti, Gaia Favero and Rita Rezzani

Submitted: 14 February 2018 Reviewed: 21 May 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.78977

From the Edited Volume

Resveratrol - Adding Life to Years, Not Adding Years to Life

Edited by Farid A. Badria

Chapter metrics overview

2,756 Chapter Downloads

View Full Metrics

Abstract

Reduced calorie intake is a religious and medical practice known since very old times, but its direct influence on life span in all organisms, included humans, has been demonstrated in the modern era. Not only periodic fasting, but also natural or synthetic compounds that mimic this phenomenon are growing to slow aging and the onset of chronic morbidities. Resveratrol (RSV), a plant polyphenol, is an elixir of longevity for simple organisms and preclinical rodent models even if a beneficial role in humans is still debated. Its main rejuvenating mechanism copes with the activation of specific longevity genes called sirtuins. Among seven known mammalian sirtuins, sirtuin 1 is the most studied. This pleiotropic nicotinamide adenine dinucleotide (NAD)-based deacetylase maintains longevity by removing acetyl group in nuclear histones, transcription factors, and other DNA repairing proteins. Actually, an exciting challenge is to discover and test novel sirtuin 1 activators to extend life span and to treat age-associated disabilities. This chapter updates on the antiaging effect of RSV and sirtuin 1 activators in experimental animals and in humans. Finally, pros and cons on RSV analogues and sirtuin 1 activators tested in preclinical and clinical trials to hamper neurological deficit, cardiovascular complications, diabetes, bone and muscle deterioration, and cancer are discussed.

Keywords

  • sirtuin 1
  • resveratrol
  • aging
  • neurodegeneration
  • diabetes
  • myopathy

1. Introduction

The increase in average life expectancy is a global consequence of sanitary welfare, proper nutrition, and healthy life style [1, 2], but unfortunately, longevity is linked to the onset of chronic irreversible diseases like neurological decline, cardiovascular damage, bone and muscle frailty, diabetes, and metabolic diseases with high economic and social costs [3, 4, 5]. Specific international clinical objectives are to maintain a “healthy aging” defined by World Health Organization (WHO) as the “functional ability that enables wellbeing in old age” by firstly preserving the quality of life together with its duration [6, 7]. Besides unchangeable genetic background, in this context, great credit is due to the quality but also the quantity of dietary nutrients [8, 9, 10].

Historically, since fifth century BC, the famous Greek physician Hippocrates declared that fasting was the best practice to block sickness progression, and, on the contrary, too much feeding gave rise to a disease. This old and simple medical concept, i.e., caloric restriction without malnutrition, has maintained for centuries but without a direct impact on life extension. Only in modern times since early 1930s, a pioneering Mc Cay discovery confirmed that undernutrition caused prolonged life span in laboratory rats [11]. By 2000s, Longo and coworkers systematically studied dietary restriction as a therapy for slowing aging and related diseases in human settings [12, 13, 14, 15]. Periodic reduction of dietary calorie intake or an intermittent fasting, under medical control, appears alternative tools to pharmacology to delay the onset of aging comorbidities, like metabolic diseases, and to improve health outcomes [16, 17, 18, 19]. “Geroscience,” a definition coined by Burch et al. [20], identifies the branch of aging biology aimed to reduce human frailty and the impact of associated chronic diseases. However, considering the difference in the duration of life between lower organisms versus humans, to fill this gap, caloric limitation has been used also in nonhuman primates like monkeys. Anyway, significant translational value has been attributed to dietary studies in short-life animals versus nonhuman primates and humans that enhanced their life span under reduced nutrients intake.

This chapter is focused on a natural dietary polyphenol, resveratrol (RSV), able to mimic calorie restriction and to prolong life duration in simple organisms and experimental animal models, but controversial in humans [21, 22]. RSV has been firstly identified as a modulator of conserved survival genes family called sirtuins [23]. Sirtuins, crucial modulators of life span extension and metabolism, have been firstly discovered by Guarente team as product of silent information regulator 2 (Sir 2) gene in yeast, Saccharomyces cerevisiae [24], worm, Caenorhabditis elegans, and fly fruit, Drosophila melanogaster [25]. In mammals, among seven known members, sirtuin 1 (SIRT1) enzyme was most beneficial against aging by mimicking calorie reduction [26, 27]. Moreover, other natural or synthetic compounds, more effective than RSV, like stimulate molecular longevity pathways via SIRT1 or pan-sirtuin activation, are currently under study [28, 29, 30]. Therefore, here major attention has been focused on newest preclinical experimental studies on laboratory animals, mainly rodents, and clinical trials based on RSV and other SIRT1 activators published over last 10 years. Due to the impressive wide scientific literature on longevity, we select only more recent articles or reviews and apologize in advance for unintentional omissions. We believe that a primary scope of this chapter is to steer readers, even if not expert in the field, to study in detail if interested, the newest antiaging therapeutic perspectives. Intriguingly, we stressed on the role of SIRT1 activators in cancer but a dichotomy exists based on specific type of cancer and clinical outcome. Indeed, SIRT1 activators (STACs) that will promote benefits in selected tumors may obtain opposite detrimental effects in others. Therefore, to date, particular caution must be taken in the pharmacological use of SIRT1 modulators in oncology. So best understanding on SIRT1 activation and maintenance by specific modulators is essential to fight age-derived inevitable disorders.

Advertisement

2. Sirtuin 1 history

The history of sirtuin family (silent information regulator 2—Sir 2) as antiaging proteins started in yeast, Saccharomyces cerevisiae, where they prolonged life span and regulated the number of replications from a mother cell [31]. Later, it was demonstrated in yeast that Sir 2 was further able to block the formation of extrachromosomal DNA linked to aging and genetic “toxicity.” Indeed, an excess of Sir 2 gene ameliorated reproductive cycle and sustained longevity in yeast budding [32]. Moreover, sirtuins were able to influence longevity in other lower organisms like worms, Caenorhabditis elegans [33], and dose-dependently in fruit fly, Drosophila melanogaster [34, 35]. Guarente team in 2000 demonstrated that the main action of sirtuins was to remove, as deacetylase enzymes, acetyl groups from specific lysine sites in nuclear histones, so allowing DNA silencing and chromosome stability [36]. This peculiar role is basic for an epigenetic regulation of DNA and telomere health and stabilization. Besides on class III DNA histones, sirtuin deacetylation activity has been extended to a plethora of other nonhistone proteins, transcription factors, and cytoplasmic proteins, which, by this posttranslational event, changed their structure and consequently function or signaling [37]. Recently, sirtuins have been considered not only deacetylases but also able to perform other posttranslational changes in their targets and for this reason defined “deacylases” [38]. Moreover, for their enzymatic activity, sirtuins used nicotinamide dinucleotide (NAD+) as a specific substrate, so their role has been related to NAD+ availability in the cell and to the NAD+/NADH ratio. In particular, it is known that during dietary caloric limitation and regular physical exercise, abundant NAD+ is produced and sirtuins are more active. There are two different ways to produce NAD+: one ex novo and another by conversion of nicotinamide (NAM) into nicotinamide mononucleotide (NMN) then charged with adenine nucleotide to become NAD. For vertebrates, the limiting enzyme necessary for the final step in the NAD synthesis is nicotinamide phosphoribosyltransferase (NAMPT) that is regulated by circadian rhythm regulator and clock genes (CLOCK and BMALI) [39]. The strict connection between NAD+ availability and sirtuin activity has been recently demonstrated and implied that competition for NAD+ substrate by different enzymes may affect sirtuin level, so contributing to age-associated diseases in mice [40, 41, 42]. In particular, NAMPT-mediated NAD synthesis is associated to the transcription of circadian-regulated genes and sirtuin in metabolically active tissues [43]. However, another crucial step in the long sirtuin history was the characterization of mammalian sirtuins, seven different isoforms (sirtuin 1–7) [44]. SIRT1 is the most studied nuclear member, pleiotropic transcriptional factor that drives many cellular activities, like energy metabolism, cell survival, DNA stability, inflammation, and circadian rhythms [45, 46]. SIRT1 is involved not only in deacetylation of a specific histone but in complex chemical reactions for different pathways involved in metabolism, like target of rapamycin (mTOR) [47], insulin signaling [48], and forkhead box O (FOXO) [49] (Figure 1). Moreover, different mice overexpressing SIRT1 have been characterized that presented better metabolism, less inflammation, and cancer but a sex-dependent longevity (longer mean life span in males vs females) [50]. In these last 5 years, the involvement of SIRT1 in crucial cellular pathways has been demonstrated and the research of new drugs acting as SIRT1 modulators and relative patents exploded [28, 30, 51, 52]. A common intriguing idea is that if SIRT1 acts as a therapeutic target, specific drugs able to activate its signaling might be effective in specific age-related pathologies and in cancer. Herein, we resumed and discuss recent scientific data on SIRT1 modulators (STACs) and their potentiality assessed in vivo, in preclinical rodent models or in clinical settings.

Figure 1.

Sirtuin 1 deacetylase activity and its downstream substrates involved in longevity. The red square indicates the inhibited pathways and the green square the activated pathways. Stars indicate acetyl groups; (AMPK): 5′ AMP-activated protein kinase; (eNOS): endothelial nitric oxide synthase; (FOXO1/3): forkhead box protein O 1/3; (NAM): nicotinamide; (NAD+): nicotinamide dinucleotide; (NAMPT): nicotinamide phosphoribosyltransferase; (NF-kB): nuclear factor k B; (p53): protein 53; (PGC1α): peroxisome proliferatoractivated receptor G coactivator 1α; (mTOR): mammalian target of rapamycin.

Advertisement

3. Resveratrol and its derivatives as sirtuin 1 activators in aging

Since 2006 Sinclair’s team in Harvard University reported that RSV, 3,5,4′-trihydroxystilbene, has a therapeutic potential to extend life span by miming caloric restriction to limit metabolic alterations in rodents in more than 140 genic pathways [23, 53]. RSV was added to the rodent diet at concentrations similar to human use (5.2 and 22.4 mg/kg/day) for 6 months. RSV was firstly isolated in late 1930s in leaves of white hellebore Veratrum grandiflorum and characterized as a phytoalexin [54], but later in 1990s, it was found in red wine and grapevines [55], in traditional herbs in Asia [56, 57, 58] in response to adverse conditions, in over 70 different plants and fruits like blueberry, raspberry, and mulberry [59]. Its first function was to reduce inflammation and to limit oxidant damage in cardiovascular diseases, the so-called “French paradox.” Intriguingly, the use of a glass of red wine reduced the extent of platelet aggregation and cardiovascular side effects in French people despite a high fat diet as reviewed in [60]. However, RSV is a photosensitive molecule, chemically composed by two aromatic rings linked by a methylene bridge, existing in two isomeric configurations, called trans-RSV and cis-RSV (Figure 2). Intriguingly, the most beneficial therapeutic properties are linked to trans-RSV even if, when exposed to light and high temperature, more than 80% of trans-RSV changes into cis-RSV with low solubility and stability [61].

Figure 2.

Resveratrol, 3′,5, 4′-trihydroxystilbene, isomers adapted from Nawaz et al. [78].

Nevertheless, it has been calculated that more than 110 glasses of wine should be drunk to achieve an anticancer effect in humans [62]. Unfortunately, lipophilicity of RSV conditioned its bioavailability, low intestinal absorption, and rapid clearance from the plasma, making necessary a high dosage in preclinical and clinical trials [63, 64]. In humans, RSV was administered as a dose of 25 mg, then grew in the range from 25 to 1000 mg, up to 5 g daily for almost 1 month still well tolerated [65]. However, when administered in healthy volunteers in the morning, RSV bioavailability and pharmacokinetic ameliorated [66] and tolerability was maintained if associated with other drugs, like quercetin and alcohol [67]. To ameliorate half-life in the plasma, a modified version of RSV was produced, called Longevinex, able to prevent isomerization from trans to cis and it is rich of vitamin D3 (at a dose 1200 IU) and quercetin very effective in the metabolic syndrome [68] and cardiac health in mice [69]. Interestingly, also in humans, RSV mimicked caloric restriction [70] and attenuated obesogenic changes in the metabolic syndrome but had no effects under normal weight patients [71]. Herein, the RSV ability to sustain SIRT1 expression/activity is effective when the deacetylase is scarce, but unnecessary if SIRT1 level/activity is normal like in postmenopausal women with normal glucose tolerance or in slightly obese men and women (trans-RSV was taken at a dose 150 mg daily for 4 weeks) [72]. However, despite extensive data on efficacy of RSV in rodent preclinical trials, actually its beneficial role in humans is still debated, greatly depending from doses and time of administration [73]. Antiaging properties of RSV via SIRT1 and mitochondrial health are addressed on amelioration of oxidative metabolism in crucial organs like heart and vessels, muscles, kidney but the mechanism is strictly cell-dependent [74, 75, 76]. Remarkably, at high doses, RSV has been reported to induce mild toxicity in humans where somnolence, headache, rash, and myalgia occurred [77]. Herein, to overcome these problems, in the past few years, different RSV derivatives have been synthesized ex novo by substituting hydroxyl with methoxy groups, so enhancing lipophilicity, or by adding a 4-hydroxy group in trans-RSV or a halogen group to potentiate the therapeutic efficacy [78], although different promising antimicrobial, antioxidant, and cardioprotective effects have been obtained in vitro. Moreover, to improve oral bioavailability of RSV-specific complexes with liposomes, lipid or synthetic nanoparticles have been made, even if they resulted with low therapeutic potential [79, 80]. In respiratory diseases reproduced in rodent like pulmonary hypertension, RSV ameliorates asthma and fibrosis via SIRT1 activation [81, 82] and also its derivative, trimethoxystilbene, demonstrated antioxidant and anti-inflammatory properties in rats exposed to hypoxia [83]. Recently, Bastin and Djouadi [84] reviewed RSV potentiality against mitochondrial damage and myopathies, and promising results in an animal model of a fatal genetic disease called Duchenne muscular dystrophy were reported. In dystrophic mice (mdx model), RSV supplementation (0.5% in the diet for 3 weeks) ameliorated muscle atrophy and prevented sciatic denervation signaling [85]. However, caution must be taken to extrapolate successful data obtained in animal models to humans where RSV might modulate different pathways in a dose-dependent manner. Autoimmune diseases represent an emerging type of chronic diseases often associated to aging that affect specific or multiple organs at the same time like rheumatoid arthritis, inflammatory bowel disease, systemic lupus erythematosus, and type I diabetes [86]. RSV inhibited transcription factors crucial in autoinflammation, like nuclear factor k B (NF-kB) [87], and curiously in these pathologies, stopped SIRT1 activity. Intriguingly, RSV behaves as a potent drug mainly in animal models, although actually poor patient outcome and reduced number of clinical trials made necessary further studies in humans to exclude side effects or competition with other drugs [88]. RSV has been tested successfully in neuronal inflammation, psychotic mice (up to 30 mg/kg/daily) [89, 90], depressed menopausal women (orally 25 mg/day for 12 weeks) [91], and in patients with minimal hepatic encephalopathy, a frequent complication of cirrhosis [92]. However, the oral dosing of RSV seemed to cross the blood-brain barrier but more potent polyphenols specifically addressed on brain with promising preventive and not only therapeutic properties are under research [93]. Moreover, RSV administered 48 h prior the induction of a subarachnoid hemorrhage activated SIRT1 thereby reducing mortality and improving neurological functions in rats [94]. Pterostilbene is a natural phenolic drug found in sandalwood and some fruits like grapes and blueberries that ameliorated neuronal aging and memory deficit in rats [95]. Chemically, it is a dimethylether analogue of RSV with better pharmacokinetic and oral bioavailability in rats (80% in comparison to 20% RSV) [96]. Recently, pterostilbene demonstrated better efficacy than RSV in the modulation of behavior and functional improving in SAMP8 mouse, a rodent model of accelerated aging validated to study Alzheimer’s disease [97]. Moreover, recent evidences indicated that pterostilbene worked well as cardioprotective and anti-inflammatory drug in rodents with ischemia-reperfusion [98]. However, both pterostilbene and RSV efficacy on aging and longevity have been reviewed recently by Li et al. [99], but conclusive data are lacking due to poor water solubility, low bioavailability, and rapid elimination (half-life only 9 h in humans), the main obstacles to work in clinical trials. However, different strategies have been also undertaken to optimize the delivery of RSV to strengthen its clinical utility: one strategy was to complex it to controlled release devices, another avenue was to use micronized powder (called SRT501) [100]. A crucial year in the history of RSV pharmacology was 1997 when Jang et al. [101] firstly reported an antitumoral effect in vitro and in vivo in a murine model of skin cancer. However, SIRT1 played a double opposite role in cancer linked to severity grade. Indeed, in early cancer phases, when cellular mutations are scarce, to activate SIRT1 is a good strategy, but in advanced stages with many mutations, SIRT1 may potentiate and accelerate oncogenesis, as demonstrated in breast cancer [102]. Remarkably, Li et al. [103] provided evidences that dietary RSV for 4–5 weeks reduced prostate neoplastic lesions by about 50% in mice through SIRT1-mediated autophagy. More recently in chondrosarcoma cells, xenografted in mice, RSV via SIRT1 stimulated apoptosis and decreased tumor growth [104].

Advertisement

4. Synthetic sirtuin 1 activators (STACs) in aging

RSV and natural polyphenols, like quercetin and butein, have been included into the first generation of SIRT1 activators (STACs). However, to overcome limitations in RSV pharmacology, its aspecific interaction with several sirtuins like human sirtuin 5 and the competition with SIRT1 for the catalytic site [105], Sinclair laboratory since 2000s produced novel synthetic STACs [28] to fight aging-associated disabilities. The screening of potential STACS began in vitro looking for more than 18,000 drugs and resulted in 21 compounds able to stimulate the catalytic site in SIRT1 deacetylase enzyme [106]. Using a fluorescence polarization assay verified by mass spectrometry, novel synthetic STACs were obtained but a strong scientific controversy occurred on the efficacy of different drugs on SIRT1 activation. To date, the research on synthetic STACs (came to the fifth generation) has produced more soluble and specific compounds [30] with an in vitro 1000-fold greater potency than RSV to mimic calorie restriction. Intriguingly, STACs extended life span in obese mice [107] but also in mice fed a standard diet. [108]. Interestingly, SRT2104 was able to preserve muscle mass, strength, and bone integrity in muscle-atrophy induced by hind limb suspension and fasting [109]. Moreover, also SRT1720 behaved as an effective SIRT1 agonist in vivo in two independent rodent models of osteoporosis, where after 1 month or 3 months of oral treatment, femoral bone mass grew about 30% [110]. These studies have great translational implications considering the high frequency of fractures in human osteoporosis. Oral administration of SRT3025 (50 or 100 mg/kg/day) for 6 weeks, starting 6 weeks after ovariectomy, successfully reversed scarcity of bone mass and osteoporosis in mice [111]. Remarkably, SRT1720 intraperitoneally injected daily in female obese mice for 6 weeks improved the vitality of follicles via sustained SIRT1 able to reduce atresia and the abnormal primordial follicles activation [112]. In human plasmacytoma xenografted mice, a model utilized to validate new therapies against multiple myeloma, oral treatment with SRT1720 (200 mg/kg) on five consecutive days/week schedule for 4 weeks reduced tumor growth in combination with other drugs, like bortezomib, potentiated antimyeloma effects [113]. Several STACS have been tested as SIRT1 modulators in preclinical rodent model to fight diabetes, obesity, neurodegeneration, atherosclerosis, bone and muscle mass [114], and most relevant diseases investigated in the preclinical trials have been resumed in Figure 3.

Figure 3.

Synthetic SIRT1 activators (STACs) effective in rodent models.

Later, since 2012, an STAC phase I clinical trials started in humans [115]. SRT2104 in doses ranged from 0.03 to 3 g was well tolerated with a bioavailability about 14% in male and female volunteers. Remarkably, the same drug was also tested in elderly volunteers with no side effects [116]. In these last years, various human clinical trials with STACs have been started but to date the only ended with SRT2104 in patients with moderate to severe psoriasis demonstrated a promising efficacy [117]. Despite some encouraging evidences, in other clinical trials, SRT2104 administration for 28 days to diabetic patients (n = 15) was ineffective on insulin resistance and endothelial function but induced a striking weight reduction not observed in placebo [118]. Moreover, patients with mild to moderate ulcerative colitis were treated with SRT2104 at 50–500 mg daily for 8 weeks, and they not presented any clinical remission so the clinical trials stopped [119]. However, the times are ripe to start further clinical trials and to test novel STACs for longer times, hoping in new exciting results.

Advertisement

5. Recommendation

RSV, the main member of the first generation STACs, is not found in meat or dairy but is present in vegetables and herbs. Renisalo et al. [120] have indicated main sources of RSV in vegetables and seeds like cocoa, grape, hop, peanuts, pistachios, tomato, and berries. However, RSV is also present in common Asian herbs like Polygonum cuspidatum, known as Japanese knotweed. Its dried roots have been infused to produce “Itadori” tea, which means “well-being” in Japanese, a folk beverage largely used for the treatment of heart disease and stroke [59].

To date, pleiotropic effects of trans-RSV in degenerative and metabolic diseases in elderly are recognized, but despite a lot of evidences in vitro and in rodent model, their therapeutic role in humans is still debated. Firstly, its beneficial effect is strictly dose-dependent, and to potentiate mitochondria health, it is required at least an oral dose of RSV of 1 g/day in Alzheimer’s patients and generally from 0.5 g up to 5 g/day to reach a therapeutic level in plasma (5 μM) [121]. Furthermore, RSV efficacy is also time-dependent, and an oral daily intake for 2 months is effective in patients with angina pectoris only if trans-RSV (at 20 mg) is associated with calcium fructoborate [122]. Moreover, controversial RSV effects reported on nonalcoholic fatty liver (NAFLD) are probably linked to different oral doses (from 500 mg/day up to 3000 mg/day), time of administration (from 56 days up to 6 months), and number of patients considered in clinical trials [123]. On the contrary, promising results have been recently obtained on 119 patients with mild to moderate Alzheimer’s disease orally supplemented with RSV at 1 g twice a day for 52 weeks, which presented reduced inflammatory markers in plasma and cerebrospinal fluid [124]. Herein, there is still a lot of research to do on the RSV and other STACs drug and caution must be taken to sustain their definitive clinical therapeutic effects in humans.

Advertisement

6. Conclusions

A constant effort has been made to synthesize novel drugs to extend life span, to prolong the quality of the life in elderly, and to limit age-associated diseases. Unfortunately, to date, there is no drug that could be an elixir of longevity for humans even if more than a hundred clinical trials on natural or synthetic SIRT1 activators are ongoing worldwide. However, there are still many challenges to discover and test the efficacy of antiaging drugs in clinics together with the necessity to get funds for a long time. The main final message from this chapter may be that a big scientific awareness is placed in the aging research, as indicated by the tremendous number of published articles on this topic. Indeed, all actors in the biogerontology scenario agree that not only the extension of the life is important in elderly, but it is fundamental its quality, maintaining a good health. To reach this aim, dietary intervention with caloric restriction mimetics is crucial, but remember to start early during adult age to best adapt your metabolism to counteract aging.

Advertisement

Acknowledgments

This chapter was supported by FFARB 2017 and ex 60% grants from the University of Brescia, Italy.

Advertisement

Conflict of interest

Authors declare no conflict of interest.

References

  1. 1. Andersen A, Sebastiani P, Dworkis D, Feldman L, Perls T. Health span approximates life span among many supercentenarians: Compression of morbidity at the approximate limit of life span. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 2012;67A:395-405. DOI: 10.1093/gerona/gls010
  2. 2. Lopez-Otin C, Blasco M, Partridge L, Serrano L, Kroemer G. The hallmarks of aging. Cell. 2013;153:1194-1217. DOI: 10.1016/j.cell.2013.05.039
  3. 3. Fabbri E, Zoli M, Gonzalez-Freire M, Salive M, Studentski S, Ferrucci L. Aging and multimorbidity: New tasks, priorities, and frontiers for integrated gerontological and clinical research. Journal of the American Medical Directors Association. 2015;16:640-647. DOI: 10.1016/j.jamda.2015.03.013
  4. 4. Kennedy B, Berger S, Brunet A, Campisi J, Cuervo AM, Epel E, Franceschi C, Lithgow G, Morimoto R, Pessin J, et al. Geroscience: Linking aging to chronic diseases. Cell. 2014;159:709-713. DOI: 10.1016/j.cell.2014.10.039
  5. 5. Bloom D, Chatterji S, Kowal P, Lloyd-Sherlock P, Mc Kee M, Rechel B, Rosenberg L, Smith J. Macroeconomic implications of population ageing and selected policy responses. Lancet. 2015;385:649-657. DOI: 10.1016/S0140-6736(14)61464-1
  6. 6. World Health Organization. World Report on Ageing and Health. Geneva: World Health Organization; 2015. p. 40
  7. 7. Mc Laughlin S, Jette A, Connell C. An examination of healthy aging across a conceptual continuum: Prevalence estimates, demographic patterns, and validity. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 2012;67(7):783-789. DOI: 10.1093/gerona/glr234
  8. 8. Kenyon C. The genetics of ageing. Nature. 2010;464:502-512. DOI: 10.1038/nature08980
  9. 9. Finkel T. The metabolic regulation of aging. Nature Medicine. 2015;21:1416-1423. DOI: 10.1038/nm.3998
  10. 10. Balasubramanian P, Howell P, Anderson R. Aging and caloric restriction research: A biological perspective with translational potential. eBioMedicine. 2017;21:37-44. DOI: 10.1016/j.ebiom.2017.06.015
  11. 11. Mc Cay C, Crowell M, Maynard L. The effect of retarded growth upon the length of life span and upon the ultimate body size: One figure. Journal of Nutrition. 1935;10:63-79. DOI: 10.1093/jn /10.1.63
  12. 12. Lee C, Longo V. Fasting vs dietary restriction in cellular protection and cancer treatment: From model organisms to patients. Oncogene. 2011;30:23305-23316. DOI: 10.1038/onc.2011.91
  13. 13. Brandhorst S, Choi I, Wei M, Cheng C, Sedrakyan S, Navarrete G, Dubeau L, Yap L, Park R, Viniguerra M, Di Biase S, Mirzaei H, Mirisola M, Childress P, Ji L, Groshen S, Penna F, Odetti F, Perin L, Conti P, Ikeno Y, Kennedy B, Cohen P, Morgan T, Dorff T, Longo V. A periodic diet that mimics fasting promotes multi-system regeneration, enhanced cognitive performance, and healthspan. Cell Metabolism. 2015;22:86-99. DOI: 10.1016/j.cmet.2015.05.012
  14. 14. Lee C, Longo V. Dietary restriction with and without caloric restriction for healthy aging. F1000Research. 2016;5:117. DOI: 10.12688/f1000research.7136.1
  15. 15. Mattson M, Longo V, Harvie M. Impact of intermittent fasting on health and disease processes. Ageing Research Reviews. 2017;39:46-58. DOI: 10.1016/j.arr.2016.10.005
  16. 16. Guarente L. Sirtuins, aging, and medicine. The New England Journal of Medicine. 2011;364:2235-2244. DOI: 10.1056/NEJMra1100831
  17. 17. Mattson M, Allison D, Fontana L, Harvie M, Longo V, Malaisse W, Mosley M, Notterpek L, Ravussin E, Scheer F, et al. Meal frequency and timing in health and disease. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:16647-16653. DOI: 10.1073/pnas.1413965111
  18. 18. Golbidi S, Daiber A, Korac B, Li H, Essop M, Laher I. Health benefits of fasting and calorie restriction. Current Diabetes Reports. 2017;17:123. DOI: 10.1007/s11892-017-0951-7
  19. 19. Weiss E, Fontana L. Caloric restriction: Powerful protection for the aging heart and vasculature. American Journal of Physiology. Heart and Circulatory Physiology. 2011;301:H1205-H1219. DOI: 10.1152/ajpheart.00685.2011
  20. 20. Burch J, Dechut A, Frieden L, Hadley E, Howcroft T, Johnson R, Khalsa P, Kohanski R, Li X, Macchiarini F, Niederehe G, et al. Advances in geroscience: Impact on healthspan and chronic disease. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 2014;69:S1-S3. DOI: 10.1093/Gerona/glu041
  21. 21. Rascon B, Hubbard B, Sinclair D, Amdam G. The lifespan extension effects of resveratrol are conserved in the honeybee and may be driven by a mechanism related to caloric restriction. Aging (Albany NY);4:499-508. DOI: 10.18632/aging.100474
  22. 22. Agarwal B, Baur J. Resveratrol and life extension. Annals of the New York Academy of Sciences. 2011;1215:138-143. DOI: 10.1111/j.1749-6632.2010.05850.x
  23. 23. Baur J, Sinclair D. Therapeutic potential of resveratrol: The in vivo evidence. Nature Reviews. Drug Discovery. 2006;5:493-506. DOI: 10.1038/nrd2060
  24. 24. Lin S, Defossez P, Guarente L. Requirement of NAD and SIR2 for life-span extension by calorie restriction in Saccharomyces cerevisiae. Science. 2000;289:2126-2128
  25. 25. Guarente L, Picard F. Calorie restriction-the SIR2 connection. Cell. 2005;120:473-482. DOI: 10.1016/j.cell.2005.01.029
  26. 26. Cohen H, Miller C, Bitterman K, Wall N, Hekking B, Kessler B, Howitz K, Gorospe M, de Cabo R, Sinclair D. Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science. 2004;305:390-392. DOI: 10.1016/j.cell.2005.01.029
  27. 27. Gillum M, Erion D, Shulman G. Sirtuin 1 regulation of mammalian metabolism. Trends in Molecular Medicine. 2011;17:8-13. DOI: 10.1016/j.molmed.2010.09.005
  28. 28. Sinclair D, Guarente L. Small-molecule allosteric activators of sirtuins. Annual Review of Pharmacology and Toxicology. 2014;54:363-380. DOI: 10.1146/annurev-pharmtox-010611-134657
  29. 29. Dai H, Kustigian L, Carney D, Case A, Considine T, Hubbard B, Perni R, Riera T, Szczepankiewicz B, Vlasuk G, Stein R. SIRT1 activation by small molecules: Kinetic and biophysical evidence for direct interaction of enzyme and activator. The Journal of Biological Chemistry. 2010;285:32695-32703. DOI: 10.1074/jbc.M110.133892
  30. 30. Bonkowski M, Sinclair D. Slowing ageing by design: The rise of NAD+ and sirtuin-activating compounds. Nature Reviews. Molecular Cell Biology. 2016;17:679-690. DOI: 10.1038/nrm.2016.93
  31. 31. Kaeberlein M, Mc Vey M, Guarente L. The SIR 2/3/4 complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms. Genes & Development. 1999;13:2570-2580
  32. 32. Sinclair D, Guarente L. Extrachromosomal rDNA circles—A cause of aging in yeast. Cell. 1997;91:1033-1042
  33. 33. Wood J, Rogin B, Laun S, Howitz K, Helfand S, Tatar M, Sinclair D. Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Nature. 2004;430:686-689. DOI: 10.1038/nature02789
  34. 34. Burnett C, Valentini S, Cabreiro F, Gass M, et al. Absence of effects of Sir2 overexpression on lifespan in C. elegans and Drosophila. Nature. 2011;477:482-485. DOI: 10.1038/nature10296
  35. 35. Whitaker R, Faulkner S, Miyokama R, Burham L, Henniksen M, Wood J, Helfand S. Increased expression of Drosophila sir2 extends life span in a dose-dependent manner. Aging (Albany NY). 2013;5:682-691. DOI: 10.18632/aging.100599
  36. 36. Imai S, Armstrong C, Kaeberlein M, Guarente L. Transcriptional silencing and longevity protein Sir2 is a NAD-dependent histone deacetylase. Nature. 2000;403:795-800
  37. 37. Grabowska W, Sikora E, Bielak-Zmjiewska A. Sirtuins, a promising target in slowing down the ageing process. Biogerontology. 2017;18:447-476. DOI: 10.1007/s10522-017-9685-9
  38. 38. Jiang H, Khan S, Wang Y, Charron G, He B, Sebastian C, Du J, Kim R, Ge E, et al. SIRT6 regulates TNF-alpha secretion through hydrolysis of long-chain fatty acyl lysine. Nature. 2013;496:110-113. DOI: 10.1036/nature12038
  39. 39. Nakagawa T, Guarente L. Sirtuins at a glance. Journal of Cell Science. 2011;124:833-838
  40. 40. Bai P, Cantò C, Oudart H, Brunyanzki A, Cen Y, Thomas C, Yamamoto H, Huber A, Kiss B, et al. PARP1 inhibition increases mitochondrial metabolism through SIRT1 activation. Cell Metabolism. 2011;13:461-468. DOI: 10.1016/j.cmet.2011.03.004
  41. 41. Schmeisser K, Mansfeld J, Kuhlow D, Weimer S, Prebe S, Heiland I, Birrinnger M, Groth M, Begree A, et al. Role of sirtuins in lifespan regulation is linked to methylation of nicotinamide. Nature Chemical Biology. 2013;9:693-700. DOI: 10.1038/nchembio.1352
  42. 42. Imai S, Guarente L. It takes two to tango: NAD+ and sirtuins in aging/longevity control. npj Aging and Mechanisms of Disease. 2016;2:16017. DOI: 10.1038/npjamd.2016.17
  43. 43. Polisak B. NAMPT-mediated NAD biosynthesis as the internal timing mechanism: In NAD+ world, time is running in its own way. Rejuvenation Research. 2017. DOI: 10.1089/rej2017.1975
  44. 44. Haigis M, Sinclair D. Mammalian sirtuins: Biological insights and disease relevance. Annual Review of Pathology. 2010;5:253-295. DOI: 10.1146/annurev.pathol.4.110807.092250
  45. 45. Boily G, Seifert E, Bevilacqua L, He X, Sabourin G, et al. SirT1 regulates energy metabolism and response to caloric restriction in mice. PLoS One. 2008;3:e1759. DOI: 10.1371/journalpone.0001759
  46. 46. Satoh A, Brace C, Ben-Josef G, West T, Wozniak D, et al. SIRT1 promotes the central adaptive response to diet restriction through activation of the dorsomedial and lateral nuclei of the hypothalamus. The Journal of Neuroscience. 2010;30:10220-10232. DOI: 10.1523/JNEUROSCI.1385-10.2010
  47. 47. Ghosh H, Mc Burney M, Robbins P. SIRT1 negatively regulates the mammalian target of rapamycin. PLoS One. 2010;5:e9199. DOI: 10.1371/journalpone.0009199
  48. 48. Longo V. Linking sirtuins IGF1-signaling and starvation. Experimental Gerontology. 2009;44:70-74. DOI: 10.1016/j.exger.2008.06.005
  49. 49. Mouchiroud L, Houtkooper R, Moullan N, Katsyuba E, Ryu D, Cantó C, Mottis A, Jo Y, Viswanathan M, Schoonjans K, Guarente L, Auwerx J. The NAD+/sirtuin pathway modulates longevity through activation of mitochondrial UPR and FOXO signaling. Cell. 2013;154:430-441. DOI: 10.1016/j.cell.2013.06.016
  50. 50. Boutant M, Cantò C. SIRT1 metabolic actions: Integrating recent advances from mouse models. Molecular Metabolism. 2014;3:5-18. DOI: 10.1016/jmomet.2013.10.006
  51. 51. Hubbard B, Gomes A, Dai H, Li J, Case A, Considine T, Riera T, Lee J, E SY, Lamming D, Pentelute B, Schuman E, Stevens L, Ling A, Armour S, Michan S, Zhao H, Jiang Y, Sweitzer S, Blum C, Disch J, Ng P, Howitz K, Rolo A, Hamuro Y, Moss J, Perni R, Ellis J, Vlasuk G, Sinclair D. Evidence for a common mechanism of SIRT1 regulation by allosteric activators. Science. 2013;339:1216-1219. DOI: 10.1126/science.1231097
  52. 52. Graziadio A, Locatelli A, Morigi R, Rambaldi M. Small molecules targeting sirtuin 1: A patent review (2012-2015). Medicinal Chemistry. 2016;6:6. DOI: 10.4172/2161-0444.1000378
  53. 53. Baur J, Pearson K, Price N, Jamieson H, Lerin C, Kalra A, Prabhu V, Allard J, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006;444:337-342. DOI: 10.1038/nature05354
  54. 54. Takaoka M. Resveratrol, a new phenolic compound from Veratrum grandiflorum. Nippon Kagaku Kaishi. 1939;60:1090-1100
  55. 55. Renaud S, de Lonrgeri M. Wine, alcohol, platelets, and the French paradox for coronary heart disease. Lancet. 1992;339:1523-1526
  56. 56. Nonomura S, Kanagawa H, Makimoto A. Chemical constituents of polygonaceous plants. I. Studies on the components of Ko-J-Kon (Polygonum cuspidatum). Yakugaku Zasshi. 1963;83:988-990
  57. 57. Hanawa F, Tahara S, Mizutani J. Antifungal stress compounds from Veratrum grandiflorum leaves treated with cupric chloride. Phytochemistry. 1992;31:3005-3007
  58. 58. Chung M, Teng C, Cheng K, Ko F, Lin C. An antiplatelet principle of Veratrum formosanum. Planta Medica. 1992;58:274-276
  59. 59. Burns J, Yokota T, Ashihara H, Lean M, Crozier A. Plant foods and herbal sources of resveratrol. Journal of Agricultural and Food Chemistry. 2002;50:3337-3340
  60. 60. Catalgol B, Batirel S, Taga Y, Ozer N. Resveratrol: French paradox revisited. Frontiers in Pharmacology. 2012;3:1-18. DOI: 10.3389/fphar.2012.00141
  61. 61. Zupancic S, Lavric Z, Kristl J. Stability and solubility of trans-resveratrol are strongly influenced by pH and temperature. European Journal of Pharmaceutics and Biopharmaceutics. 2015;93:196-204. DOI: 10.1016/j.ejpb.2015.04.002
  62. 62. Lachenmeier D, Godelmann R, Witt B, Riedel K, Rehm J. Can resveratrol in wine protect against the carcinogenicity of ethanol? A probabilistic dose-response assessment. International Journal of Cancer. 2014;134:144-153. DOI: 10.1002/ijc.28336
  63. 63. Gambini J, Ingles M, Olaso G, Lopez-Grueso R, Bonet-Costa V, Gimeno-Mallench L, Mas-Barues C, et al. Properties of resveratrol: In vitro and in vivo studies about metabolism, bioavailability, and biological effects in animal models and humans. Oxidative Medicine and Cellular Longevity. 2015;2015:837042. DOI: 10.1155/2015/837042
  64. 64. Cottarrt C, Nivet-Antoine V, Beaudeux J. Review of recent data on the metabolism, biological effects and toxicity of resveratrol in humans. Molecular Nutrition & Food Research. 2014;58:7-21. DOI: 110.1002/mnfr.201200589
  65. 65. Yang T, Wang L, Zhu M, Zhang L, Yan L. Properties and molecular mechanisms of resveratrol: A review. Die Pharmazie. 2015;70:501-506
  66. 66. Almeida L, Vaz-da Silva M, Falcao A, et al. Pharmacokinetic and safety profile of trans-resveratrol in a rising multiple dose study in healthy volunteers. Molecular Nutrition & Food Research. 2009;53:S7-S15
  67. 67. La Porte C, Voduc N, Zhang G, Seguin I, Tardiff D, Singhal N, Cameron D. Steady-state pharmacokinetics and tolerability of trans-resveratrol 2000 mg twice daily with food, quercetin and alcohol (ethanol) in healthy human subjects. Clinical Pharmacokinetics. 2010;49:449-454. DOI: 10.2165/11531820-000000000-00000
  68. 68. Fujitaka K, Otani H, Jo F, Jo H, Nomura E, Iwasaki M, Nishikawa M, Iwasaka T, Das D. Modified resveratrol Longevinex improves endothelial function in adults with metabolic syndrome receiving standard treatment. Nutrition Research. 2011;31:842-847. DOI: 10.1016/j.nutres.2011.09.028
  69. 69. Das S, Mitrovsky G, Vasanthi H, Das D. Antiaging properties of a grape-derived antioxidant are regulated by mitochondrial balance of fusion and fission leading to mitophagy triggered by a signalling network of Sirt1-Sirt3-Foxo3-PINK1-PARKIN. Oxidative Medicine and Cellular Longevity. 2014;2014:345105. DOI: 10.1155/2014/345105
  70. 70. Li J, Zhang C, Liu Y, Chen K, Chen G. A comparative study of anti-aging properties and mechanism: Resveratrol and caloric restriction. Oncotarget. 2017;8:65717-65729. DOI: 10.18632/oncotarget.20084
  71. 71. Yoshino J, Conte C, Fontana L, Mittendorfer B, Imai S, Schechtman K, Gu C, Kunz I, Rossi Fanelli F, Patterson B, Klein S. Resveratrol supplementation does not improve metabolic function in nonobese women with normal glucose tolerance. Cell Metabolism. 2012;16:658-664. DOI: 10.1016/j.cmet.2012.09.015
  72. 72. van der Made S, Plat J, Mensink R. Trans-resveratrol supplementation and endothelial function during the fasting and postprandial phase: A randomized placebo-controlled trial in overweight and slightly obese participants. Nutrients. 2017;9:596. DOI: 10.3390/nu9060596
  73. 73. Berman A, Motechin R, Wiesenfeld M, Holz M. The therapeutic potential of resveratrol: A review of clinical trials. npj Precision Oncology. 2017;35:1-9. DOI: 10.1038/s41698-017-0038-6
  74. 74. Trevino-Saldana N, Garcia-Rivas. Regulation of sirtuin-mediated protein deacetylation by cardioprotective phytochemicals. Oxidative Medicine and Cellular Longevity. 2017;2017:1750306. DOI: 10.1155/2017/1750306
  75. 75. Lan F, Weikel K, Caceido J, Ido Y. Resveratrol-induced AMP-activated protein kinase activation is cell-type dependent: Lessons from basic research for clinical application. Nutrients. 2017;9:751. DOI: 10.33390/nu9070751
  76. 76. Kim E, Lim J, Kim M, Ban T, Jang I, Yoon H, Park C, Chang Y, Choi B. Resveratrol, an Nrf2 activator, ameliorates aging-related progressive renal injury. Aging. 2018;10:83-99. DOI: 10.18632/agibg.101361
  77. 77. Cottart C, Nivet-Antoine V, Laguiller-Morizot C, Beaudex J. Resveratrol bioavailability and toxicity in humans. Molecular Nutrition & Food Research. 2010;54:7-16. DOI: 10.1002/mnfr.200900437
  78. 78. Nawaz W, Zhou Z, Dend S, Ma X, Li C, Shu X. Therapeutic versatility of resveratrol derivatives. Nutrients. 2017;9:1188. DOI: 10.3390/nu9111188
  79. 79. Wegiel L, Mauer L, Edgar K. Taylor l: Crystallization of amorphous solid dispersions of resveratrol during preparation and storage-impact of different polymers. Journal of Pharmaceutical Sciences. 2013;102:171-184. DOI: 10.1002/jps.23358
  80. 80. Bu L, Gan L, Guo X, et al. Trans-resveratrol loaded chitosan nanoparticles modified with biotin and avidin to target hepatic carcinoma. International Journal of Pharmaceutics. 2013;452:355-362. DOI: 10.1016/j.ijpharm.2013.05.007
  81. 81. Yu L, Tu Y, Jia X, et al. Resveratrol protects against pulmonary arterial hypertension in rats via activation of silent information regulator 1. Cellular Physiology and Biochemistry. 2017;42:55-67. DOI: 10.1159/000477115
  82. 82. Zhu X, Lei X, Dong W. Resveratrol as a potential therapeutic drug for respiratory system diseases. Drug Design, Development and Therapy. 2017;11:3591-3598. DOI: 10.2147/DDDt.S148868
  83. 83. Liu B, Lon X, Yang Z, et al. Inhibition of NOX/VPO1 pathway and inflammatory reaction by trimethoxystilbene in prevention of cardiovascular remodeling in hypoxia-induced pulmonary hypertensive rats. Journal of Cardiovascular Pharmacology. 2014;63:567-576. DOI: 10.1097/FJC.0000000000000082
  84. 84. Bastin J, Djouadi F. Resveratrol and myopathy. Nutrients. 2016;8:254. DOI: 10.3390/nu8050254
  85. 85. Asami Y, Aizawa M, Kinoshita M, Ishikawa J, Sakuma K. Resveratrol attenuates denervation-induced muscle atrophy due to the blockade of atrogin-1 and p62 accumulation. International Journal of Medical Sciences. 2018;15:628-637. DOI: 10.7150/ijms.22723
  86. 86. Rosenblum M, Remedios K, Abbas A. Mechanisms of human autoimmunity. The Journal of Clinical Investigation. 2015;125:2228-2233. DOI: 10.1172/JCI78088
  87. 87. Lee S, Yang H, Tartar D, Gao B, Luo X, Ye S, Zaghouani H, Fang D. Prevention and treatment of diabetes with resveratrol in a non-obese mouse model of type 1 diabetes. Diabetologia. 2011;54:1136-1146. DOI: 10.1007/s00125-011-2064-1
  88. 88. de Brito Oliveira A, Monteiro S, Navegantes-Lima K, Ferreira Reis J, de Souza Gomes R, Rodrigues D, Gaspar S, Monteiro M. Resveratrol role in autoimmune disease—A mini review. Nutrients. 2017;9:1306. DOI: 10.3390/nu9121306
  89. 89. Magaji M, Iniaghe L, Abolarin M, Abdullahi O, Magaji R. Neurobehavioural evaluation of resveratrol in murine models of anxiety and schizophrenia. Metabolic Brain Disease. 2017;32:437-442
  90. 90. Finnell J, Lombard C, Melson M, Singh N, Nagarkatti M, Nagarkatti P, Fadel J, Wood C, Wood S. The protective effects of resveratrol on social stress-induced cytokine release and depressive-like behaviour. Brain, Behavior, and Immunity. 2017;59:147-157. DOI: 110.1016/j.bbi.2016.08.019
  91. 91. Davinelli S, Scapagnini G, Marzatico F, Nobile V, Ferrara N, Corbi G. Influence of equol and resveratrol supplementation on health-related quality of life in menopausal women: A randomized, placebo-controlled study. Maturitas. 2017;96:77-83. DOI: 10.1016/j.maturitas.2016.11.016
  92. 92. Malaguarnera G, Pennisi M, Bertino G, Motta M, Borzì A, Vicari E, Bella R, Drago F, Malaguarnera M. Resveratrol in patients with minimal hepatic encephalopathy. Nutrients. 2018;10:329. DOI: 10.3390/nu10030329
  93. 93. Pasinetti G, Wang J, Ho L, Zhao W, Dubner L. Roles of resveratrol and other grape-derived polyphenols in Alzheimer’s diseases prevention and treatment. Biochimica et Biophysica Acta. 2015;1852:1202-1208. DOI: 10.1016/j.bbadis.2014.10.006
  94. 94. Qian C, Jin J, Chen J, Li J, Yu Y, Mo H, Chen G. SIRT 1 activation by resveratrol reduces brain edema and neuronal apoptosis in an experimental rat subarachnoid hemorrhage model. Molecular Medicine Reports. 2017;16:9627-9635. DOI: 10.3892/mmr.2017.7773
  95. 95. Casadesus G, Shukitt-Hale B, Stellwagen H, Zhu X, Lee H, Smith M, Joseph J. Modulation of hippocampal plasticity and cognitive behaviour by short-term blueberry supplementation in aged rats. Nutritional Neuroscience. 2004;7:309-316. DOI: 10.1080/10284150400020482
  96. 96. Kapetanovic I, Muzzio M, Huang Z, Thompson T, Mc Cormyk P. Pharmacokinetics, oral bioavailability and metabolic profile of resveratrol and its dimethylether analog, pterostilbene in rat. Cancer Chemotherapy and Pharmacology. 2011;68:593-601. DOI: 10.1007/s00280-010-1525-4
  97. 97. Chang J, Rimando A, Pallas M, Camins A, Porquet D, Reeves J, Shukitt-Hale B, Smith M, Joseph J, Casadesus G. Low-dose pterostilbene, but not resveratrol, is a potent neuromodulator in aging and Alzheimer’s disease. Neurobiology of Aging. 2012;33:2062-2071. DOI: 10.1016/j.neurobiolaging.2011.08.015
  98. 98. Wu M, Lu S, Zhong J, Huang K, Zhang S. Protective effects of pterostilbene against myocardial ischemia/reperfusion injury in rats. Inflammation. 2017;40:578-588. DOI: 10.1007/s10753-016-0504-2
  99. 99. Li Y, Li S, Lin C. Effect of resveratrol and pterostilbene on aging and longevity. BioFactors. 2018;44:69-82. DOI: 10.1002/biof.1400
  100. 100. Smoliga J, Blanchard O. Enhancing the delivery of resveratrol in humans: If low bioavailability is the problem, what is the solution? Molecules. 2014;19:17154-17172. DOI: 10.3390/molecules191117154
  101. 101. Jang M, Cai L, Udeani G, Slowing K, Thomas C, Beecher C, Fong H, Farnsworth N, Kinghorn A, Meht A, Moon R, Pezzuto J. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science. 1997;275:218-220. DOI: 10.1126/science.275.5297.218
  102. 102. Rifai K, Judes G, Idrissou M, Daures M, Bignon Y, Penault-Liorca F, Bernard-Gallon D. Dual SIRT1 expression patterns strongly suggests its bivalent role in human breast cancer. Oncotarget. 2017;8:110922-110930. DOI: 10.18632/oncotarget.23006
  103. 103. Li G, Rivas P, Bedolla R, Thapa D, Reddick R, Ghosh R, Kumar A. Dietary resveratrol prevents development of high-grade prostatic intraepithelial neoplastic lesions: Involvement of SIRT1/S6K axis. Cancer Prevention Research. 2013;6:27-39. DOI: 10.1158/1940-6207.CAPR-12-0349
  104. 104. Chao S, Chen Y, Huang K, Kuo K, Yang T, Huang K, Wang C, Tan Q, Yang R, Liu S. Induction of sirtuin 1 signaling by resveratrol induces human chondrosarcoma cells apoptosis and exhibits antitumor activity. Scientific Reports. 2017;7:3180. DOI: 10.1038/s41598-017-03635-7
  105. 105. Gertz M, Nguyen G, Fischer F, Suenkel B, Schlicker C, Franzel B, Tomaschewski J, Aladini F, Becker C, Wolters D, Steegborn C. A molecular mechanism for direct sirtuin activation by resveratrol. PLoS One. 2012;7:e49761. DOI: 10.1371/journal.pone.0049761
  106. 106. Hubbard B, Sinclair D. Small molecule SIRT 1 activators for the treatment of aging and age-related diseases. Trends in Pharmacological Sciences. 2014;35:146-154. DOI: 10.1016/j.tips.2013.12.004
  107. 107. Minor R, Baur J, Gomes A, Ward T, Csizar A, Mercken E, Abdelmohsen K, Shin Canto C, et al. SRT1720 improves survival and health span of obese mice. Scientific Reports. 2011;1:70. DOI: 10.1038/srep00070
  108. 108. Mitchell S, Martin-Monsalvo A, Mercken E, Palacios H, Ward T, Abulwerdi G, Minor R, Vlasuk G, et al. The SIRT1 activator SRT1720 extends lifespan and improves health of mice fed a standard diet. Cell Reports. 2014;6:836-843. DOI: 10.1016/j.cel rep. 2014.01.031
  109. 109. Mercken E, Mitchell J, Martin-Monsalvo A, Minor R, Almeida M, Gomes A, Scheibe-Knudsen M, Palacios H, Licata J, Zhang Y. SRT2104 extends survival of male mice on a standard diet and preserves bone and muscle mass. Aging Cell. 2014;13:787-796. DOI: 10.1111/acel.12220
  110. 110. Zainabadi K, Liu C, Caldwell A, Guarente L. SIRT1 is a positive regulator of in vivo bone mass and a therapeutic target for osteoporosis. PLoS One. 2017;12:e0185236. DOI: 10.1371/journal.pone.0185236
  111. 111. Artsi H, Cohen-Kfir E, Gurt I, Shahar R, Bajayo A, Kalish N, Bellido T, Gabet Y, Dresner-Pollak R. The sirtuin 1 activator SRT3025 down-regulates sclerostin and rescues ovariectomy-induced bone loss and biomechanical deterioration in female mice. Endocrinology. 2014;155:3508-3515. DOI: 10.1210/en.2014-1334
  112. 112. Zhou X, Xu J, Ni Y, Chen X, Zhang H, Zhang X, Liu W, Luo L, Fu Y. SIRT1 activator (SRT1720) improves the follicle reserve and prolongs the ovarian lifespan of diet-induced obesity in female mice via activating SIRT1 and suppressing mTOR signaling. Journal of Ovarian Research. 2014;7:97. DOI: 10.1186/s13048-014-0097.z
  113. 113. Chauhan D, Bandi M, Singh A, Ray A, Raje N, Richardson P, Anderson K. Preclinical evaluation of a novel SIRT1 modulator SRT1720 in multiple myeloma cells. British Journal of Haematology. 2011;155:588-598. DOI: 10.1111/J.1365-2141.2011.08888.x
  114. 114. Carafa V, Rotili D, Forgione M, Cuomo F, Serretiello E, Hailu G, Jarho E, Lahtela-Kakkonen M, Mai A, Altucci L. Sirtuin functions and modulation: From chemistry to the clinic. Clinical Epigenetics. 2016;8:61. DOI: 10.1186/s13148-016-0224-3
  115. 115. Hoffmann E, Wald J, Lavu S, Roberts J, Beaumont C, Haddad J, Elliott P, Westphal C, Jacobson E. Pharmacokinetics and tolerability of SRT2104, a first-class small molecule activator of SIRT1, after single and repeat oral administration in man. British Journal of Clinical Pharmacology. 2012;75:186-196. DOI: 10.1111/j.1365-2125.2012.04340.x
  116. 116. Libri V, Brown A, Gambarota G, Hada J, Shields G, Dawes H, Pinato P, Hoffman E, Elliot P, Vlasuk G, et al. A pilot randomized, placebo controlled, double blind phase I trial of the novel SIRT1 activator SRT2104 in elderly volunteers. PLoS One. 2012;7:e51395. DOI: 10.1037/journal.pone.0051395
  117. 117. Krueger J, Suares-Farinas M, Cueto I, Khacherian A, Matheson R, Parish L, Leonardi C, Shortino D, Gupta A, Haddad J, Vlasuk G, Jacobson E. A randomized, placebo-controlled study of SRT2104, a SIRT1 activator, in patients with moderate to severe psoriasis. PLoS One. 2015;10:e0142081. DOI: 10.1371/journal.pone.0142081
  118. 118. Noh R, Venkatasubramanian S, Daga S, Langrish J, Mills N, Lang N, Hoffmann E, Waterhouse B, Newby D, Frier B. Cardiometabolic effects of a novel SIRT1 activator, SRT2104, in people with type 2 diabetes mellitus. Open Heart. 2017;4:e000647. DOI: 10.1136/0penhrt-2017-000647
  119. 119. Sands B, Joshi S, Haddad J, Freudenberg J, Oommen D, Hoffmann E, McCallum S, Jacobson E. Assessing colonic exposure, safety, and clinical activity of SRT2104, a novel oral SIRT1 activator, in patients with mild to moderate ulcerative colitis. Inflammatory Bowel Diseases. 2016;22:607-614. DOI: 10.1097/MIB. 0000000000000597
  120. 120. Renisalo M, Karlund A, Kokela A, Kaarn I, Ranta K, Karjalaineen R. Polyphenol stilbenes: Molecular mechanisms of defence against oxidative stress and aging-related diseases. Oxidative Medicine and Cellular Longevity. 2015;2015:340520. DOI: 10.1155/2015/340520
  121. 121. Madreiter-Sokolowsi C, Sokolowski A, Graier W. Dosis facit sanitatem—Concentration-dependent effects of resveratrol on mitochondria. Nutrients. 2017;9:1117. DOI: 10.3390/nu9101117
  122. 122. Militaru C, Donoiu I, Craciun A, Score I, Bulearca A, Scorei R. Oral resveratrol and calcium fructoborate supplementation in subjects with stable angina pectoris: Effect on lipid profiles, inflammation markers and quality of life. Nutrition. 2013;29:178-183. DOI: 10.1016/J.nut.2012.07.006
  123. 123. Zhang C, Yuan W, Fang J, Wang W, He P, Lei J, Wang C. Efficacy of resveratrol supplementation against non alcoholic fatty liver disease: A meta-analysis of placebo-controlled clinical studies. PLoS One. 2016;11:e0161792. DOI: 10.1371/journal.pone.0161792
  124. 124. Moussa C, Hebron H, Huang X, Ahn J, Rissman R, et al. Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer’s disease. Journal of Neuroinflammation. 2017;14:1. DOI: 10.1186/s12974-016-0779-0

Written By

Alessandra Stacchiotti, Gaia Favero and Rita Rezzani

Submitted: 14 February 2018 Reviewed: 21 May 2018 Published: 05 November 2018