Chapter 22

Calorie Restriction Mimetics From Functional Foods

Impact on Promoting a Healthy Life Span

Wai Yan Sun and Yu Wang,    The University of Hong Kong, Hong Kong, China

Abstract

Aging is a complex process of functional decline at the molecular, cellular, and organ system levels. Even as human life expectancy is being prolonged, age-related diseases continue to be major obstacles for healthy aging and longevity. Evidence from different biological models ranging from yeasts, worms, flies, rodents, and primates to humans suggests a number of conserved aging mechanisms offer pathways to maintain the “youthful” states. Calorie restriction remains the most robust nongenetic approach for extending health and life span. It slows primary aging and effectively protects secondary aging in both animals and humans. Calorie restriction mimetics (CRMs) targeting the molecular aging pathways have emerged as a promising strategy for combating age-related morbidities. This chapter introduces a number of CRMs derived from functional foods and discusses their mechanisms of action in mimicking calorie restriction.

Keywords

Calorie restriction; intermediary metabolites; aging; autophagy; protein acetylation; SIRT-1

Introduction: Calorie Restriction

The life expectancy of humans has dramatically increased over the past few centuries, which is imposing negative impacts on age-associated diseases and the medical cost of health care. Aging is a multifactorial biological process, progressively degenerative in nature and resulting in loss of physical or mental functions (or both) (Dabhade and Kotwal, 2013; Shokolenko et al., 2014). Primary aging refers to the currently inevitable process of the functional deterioration of organs as well as slowing movement, fading vision, progressive hearing loss, and increased susceptibility to infection. In contrast, secondary aging is preventable and results largely from disease processes, harmful environmental factors, and poor lifestyle decisions such as smoking and a lack of physical activity (Anstey et al., 1993). Numerous signaling pathways that are commonly or specifically involved in age-related diseases—including type 2 diabetes mellitus, cardiovascular disease, and Alzheimer’s disease (AD)—have been identified and characterized (Masri, 2015). As a result, the protections against secondary aging lead to a significant increase in the average life span (Fontana et al., 2010). However, our knowledge of the underlying causes for primary aging remain primitive. Accordingly, the maximal longevity of humans remains largely unchanged.

Calorie intake influences the rate of aging and the onset of age-related diseases in animals and humans. Calorie restriction is a dietary regimen to limit the consumption of food without causing malnutrition, which effectively slows the process of primary aging and protects secondary aging in rodents (McCay et al. 1935; Weindruch and Sohal 1997) as well as in nonhuman primates (Lane et al., 1996; Colman et al., 2009). The effects are robust as evident in mice with long-term 55% to 65% calorie restriction leading to a 65% extension of maximal life span when compared to the control group with ad libitum feeding (Weindruch, 1996). Two long-term calorie restriction studies using rhesus monkeys by the Wisconsin National Primate Research Center (WNPRC) and the National Institute on Aging (NIA) produced controversial results. The former was initiated in 1989, and its results were published in 2014 with the conclusion that a long-term approximately 30% restricted diet significantly improved age-related and all-cause survival in rhesus monkeys (Colman et al., 2014). The NIA initiated its study in 1987, and the interim report in 2012 showed that calorie restriction did not increase the median life span of rhesus monkeys (Mattison et al., 2012). Notably, WNPRC study performed 30% dietary restriction that was individualized for each monkey, whereas the monkeys of NIA study were fed with 22% to 24% reduced calories based on the standardized chart of food intake by the National Academy of Science. In addition, all monkeys in the NIA study were supplemented with minerals and vitamins above the adequate amount in order to avoid any compromising factors. However, only the calorie-restricted monkeys were supplemented in WNPRC study. The control monkeys at NIA weighed less than those at the WNPRC at all time points, suggesting an inadequate food intake (Tenenbaum, 2014).

Long-term calorie restriction reduces body weight and has beneficial effects on the health span of nonhuman primates by reducing cancer development, neurodegeneration, and metabolic disorders (Mattison et al., 2012; Libert and Guarente, 2013). Intermittent calorie restriction also shows similar benefits in improving the health span in humans, but consistent weight loss is unlikely to be seen (Heilbronn et al., 2005). In mice, calorie restriction not only reduces body weight and fat mass but also decreases insulin-like growth factor 1 (IGF-1), leptin, fasting blood glucose, and insulin levels (Mai et al., 2003; Tatar et al., 2003; Wang et al., 2013). Overall, calorie restriction enhances metabolic and anti-inflammatory profiles. Effects such as lowered blood glucose and insulin occur quickly but also diminish rapidly when calorie restriction stops (Anderson and Herman, 1972; Mitchell et al., 2015). Thus, to be beneficial, intervention must be sustained. Nevertheless, calorie restriction lowers the susceptibility of a wide spectrum of age-associated diseases. For instance, mice on a calorie-restricted diet showed attenuation of age-related neuronal loss seen in Parkinson’s disease (Duan and Mattson, 1999) and in AD (Zhu et al., 1999); improvement for the brain’s plasticity in learning (Mattson, 2000); prevention of age-associated declines in psychomotor and spatial memory tasks (Ingram et al., 1987); and reductions in autoimmune disease, kidney damage, and diabetes (Fernandes and Good, 1984). Taken together, calorie restriction protects secondary aging in rodents and nonhuman primates.

Long-term population studies of calorie restriction are hard to conduct for primary aging in humans (Holloszy and Fontana, 2007). Physiological and metabolic variables are determined as the biomarkers for secondary aging. The Biosphere 2 study initiated in 1991 with eight healthy humans (four males and four females) sealed inside a 3.15-acre enclosure began with a limited food supply. After six months, the occupants’ body weights, blood pressure, serum cholesterol, fasting glucose, and leukocyte counts were significantly reduced without any changes in physical or mental health status (Walford et al., 1992). The Comprehensive Assessment of Long-Term Effects of Reducing Intake of Energy was the first random trial in free-living nonobese men and women investigating the effects of calorie restriction (Stewart et al., 2013; Romashkan et al., 2016). During the two-year study period, individuals on an 11% calorie-restricted diet exhibited 10% weight loss, whereas the body weight of the control counterparts remained unchanged (Ravussin et al., 2015). Calorie restriction significantly reduced the levels of triiodothyronine and tumor necrosis factor alpha (a marker of inflammation) (Ravussin et al., 2015). Memory and learning performance is enhanced, and insulin sensitivity is improved in healthy elderly persons subject to calorie restriction (Witte et al., 2009). Furthermore, diabetic elderly patients on calorie restriction for 12 weeks show improved blood pressure, glucose and lipid profiles, as well as liver functions, all of which contribute to low cardiovascular risks (Choi et al., 2013).

In the 1950s, Okinawan elders were consuming a calorie-restricted diet that was approximately 1800 cal/day and approximately 20% lower than the national average of Japan (Heilbronn and Ravussin, 2003). Public health care and the quality of the diet on Okinawa were sufficiently good to prevent nutritional deficiencies and infectious diseases. Thus, life expectancy was 81.2 years, significantly higher than those of European countries (e.g., 78.3 years in Italy and 78.1 years in Greece) and the United States (76.8 years) (Sohal and Weindruch, 1996). Because of poverty, however, elderly Okinawans growth was significantly stunted. In fact, long-term calorie restriction may not be a desirable dietary method for improving health span as well as longevity in humans. On the one hand, personal motivation to undergo long-term calorie restriction may be lacking. Thus, a 30% to 60% calorie restriction is not achievable by most human subjects, especially the elderly. On the other hand, the side effects of long-term calorie reduction remain unclear, but include reduced bone mineral densities, functional disability, infertility, and slow recovery from injuries (Ingram and Roth, 2011; Romashkan et al., 2016). Moreover, calorie restriction can provoke unpleasant common side effects such as reduced body temperature and libido (Ingram and Roth, 2011).

Recent decades have seen the introduction of calorie restriction mimetics (CRMs), which are also known as energy restriction mimetics, in which a class of natural or synthetic pharmacological substances are provided as food supplements to mimic the beneficial anti-aging effects of calorie restriction (Chen and Guarente, 2007; Lee and Min, 2013). The genes and pathways involved in the actions of calorie restriction are the targets for discovering and developing CRMs (Ingram et al., 2006). An effective CRM activates the same physiological and metabolic pathways as calorie restriction, triggers stress-response mechanisms for protection against cellular damage, and extends health and life span without the requirement of dietary or energy restriction. In the following sections, the mechanisms of action for CRMs derived from functional foods will be discussed with a focus on their roles in mimicking the metabolic or signaling pathways of calorie restriction.

CRMS in Mediating Metabolic Pathways of Calorie Restriction

A precise understanding of the mechanisms underlying the actions of calorie restriction is needed for identifying and generating CRMs. Starvation of cells or dietary restriction in mammals leads to a rapid reduction of intracellular acetyl coenzyme A (acetyl-CoA), which is coupled to the status of protein acetylation and deacetylation (Bao and Sack, 2010). Decreased acetyl-CoA levels occur before the reduction of adenosine triphosphate (ATP), oxidation of nicotinamide adenine dinucleotide phosphate (NADH), and the depletion of amino acids. The coordinated alterations of these metabolites trigger a number of key cellular defense mechanisms via the activation of nutrient sensors, including adenosine monophosphate (AMP)-activated protein kinase (AMPK) and silent mating type information regulation 2 homologue 1 (SIRT-1), as well as inactivation of mammalian target of rapamycin (Cetrullo et al., 2015).

Targeting Glycolysis and the Citric Acid Cycle to Limit Energy Supply

In cells, glycolysis is a multistep process that utilizes energy (ATP) to break down glucose into pyruvate, which is then decarboxylated into acetyl-CoA (Akram, 2014). Glycolytic inhibition has been suggested as a strategy for developing CRMs (Roth et al., 1999) based on the rationale that reducing energy supply enhances the metabolic responses in cells, a process mimicking calorie restriction (Ingram and Roth. 2011). There are several key enzymes for targeting the glycolytic pathway. In particular, phosphoglucose isomerase, which converts glucose-6-phosphate to fructose-6-phosphate, is the target of 2-deoxy-D-glucose (2DG) (Ingram and Roth, 2011). Low doses of 2DG enhance oxidative stress protection and neuronal resistance by upregulating heat shock protein 70 (HSP-70) and glucose-regulated protein 78, resulting in neuroprotection (Lee et al., 1999). Glycolytic inhibition delays secondary aging and prevents neurodegenerative disorders.

Glyceraldehyde-3-phosphate dehydrogenase is another target of glycolytic inhibition in vitro in which administration of iodoacetate that inhibits this enzyme can upregulate the expression of HSP-70 and HSP-90 in neurons against excitotoxic and oxidative injury (Guo et al., 2001). Other glycolytic inhibitors—including glucosamine, mannoheptulose, and 3-bromopyruvate—are also candidate CRMs. The phosphorylated form of glucosamine, glucosamine-6-phosphate, is an inhibitor of hexokinase, which is involved in the first step of glycolysis (Marshall et al., 2005). Glucosamine activates autophagy, which is an important cellular mechanism for anti-aging and discussed in later (Carames et al., 2013). In antitumor studies, mannoheptulose and 3-bromopyruvate exhibit inhibitory effects on the glycolytic enzyme hexokinase activity, where this early part of the glycolytic reactions is utilized by cancer cells to generate their own energy (Pedersen, 2007; Wang et al., 2016). However, the underlying mechanisms for CRM are not fully known in vivo. When dogs were fed mannoheptulose, their fasting insulin levels were reduced in a dose-dependent manner, but the serum glucose levels did not differ (McKnight et al., 2014; Ingram and Roth, 2015). These agents can be candidates for CRM, but further studies are required to investigate their full mechanisms of action.

The citric acid cycle (also known as the Krebs cycle or tricarboxylic acid cycle) consists of a series of reactions conducted within the mitochondria of eukaryotic cells or in the cytoplasm of prokaryotic cells (Kornberg, 2000; Korla and Mitra, 2014). It is a central driver of cellular respiration and takes acetyl-CoA as the starting material. Oxaloacetate is the end product of one citric acid cycle. Supplementation with oxaloacetate has been shown to increase longevity in Caenorhabditis elegans via an increase in the ratio of nicotinamide adenine dinucleotide (NAD) to NADH and activation of the AMPK pathway (Williams et al., 2009). The conversion of oxaloacetate to malate promotes the conversion of NADH to NAD. Unlike 2DG, oxaloacetate supports glycolysis and cellular respiration (Wilkins et al., 2016). In mice, administration of oxaloacetate increases brain bioenergetic function by activating hippocampal neurogenesis and reducing neuroinflammation via the Akt signaling pathway (Wilkins et al., 2014). A recent study has revealed the safety profile of oxaloacetate in humans, but the efficacy in AD or other age-associated diseases has yet to be elucidated (Swerdlow et al., 2016).

Acetyl-CoA Depletion to Induce Cellular Stress Responses

Formation of intracellular acetyl-CoA occurs in both mitochondrial and nucleocytosolic compartments. In mammals, the mitochondrial acetyl-CoA is produced from pyruvate, fatty acid, and branched-chain amino acids via glycolysis, lipolysis, and oxidative decarboxylation (Madeo et al., 2014). Acetyl-CoA in the mitochondrial matrix is condensed with oxaloacetate into citrate, which can be exported into the cytoplasm, where it freely diffuses in and out of nucleus. The extramitochondrial acetyl-CoA is produced from citrate by ATP citrate lyase or from acetate by acetyl-CoA synthase (Choudhary et al., 2014; Marino et al., 2014; Schroeder et al., 2014). Excessive nutrient supply enhances the transportation of acetyl-CoA from mitochondria to cytosol for the synthesis of fatty acids and sterols (Wellen et al., 2009; Cai et al., 2011; Shi and Tu, 2015). Short-term removal of nutrients in human cell culture and overnight fasting of rodents rapidly reduces acetyl-CoA levels (Marino et al., 2014). Notably, when cells commit to proliferate, the intracellular levels of acetyl-CoA are substantially elevated to promote histone acetylation so the gene expression for growth can be switched on (Cai et al., 2011). This mechanism requires energy consumption and is pro-aging. Conversely, reduction of acetyl-CoA levels is observed when arresting cell growth to reserve energy, which favors anti-aging.

A number of natural compounds act as CRMs to reduce acetyl-CoA or inhibit protein acetylation. For example, hydroxycitrate (Hackenschmidt et al., 1972) and epigallocatechin-3-gallate (EGCG), one of the major active compounds in green tea (Santana et al., 2015), stimulate the activity of acetyl-CoA carboxylase, a biotin-dependent enzyme that irreversibly catalyzes the carboxylation of acetyl-CoA to form malonyl-CoA. Anacardic acid (6-pentadecyl-salicylic acid from the nutshell of the cashew, Anacardium occidentale), garcinol (from the fruit of the kokum tree, Garcinia indica), and spermidine (a polyamine found at particular high concentrations in durian fruit, fermented soybeans, and wheat germs) reduce the overall lysine acetylation of cellular protein via inhibition of acetyltransferases, in turn inducing autophagy (Pietrocola et al., 2015).

Nucleocytosolic acetyl-CoA production is a metabolic repressor of autophagy, a self-cannibalistic pathway to eliminate damaged organelles and toxic protein aggregates, thus mobilizing energy reserves when external resources are limited (Levine and Kroemer, 2008). During this process, the unique organelle known as the autophagosome delivers the cellular components to a lysosome for degradation. A number of autophagy-related proteins (Atg) are involved in the formation of autophagosome (Ravikumar et al., 2010). Notably, the biological process of autophagy decreases with age (Cavallini et al., 2007). Deficiency or attenuation of autophagy causes premature aging and leads to various age-dependent diseases (Levine and Kroemer, 2008; Hartleben et al., 2010; Cui et al., 2013). Conversely, activation of autophagy by overexpression of Atg can increase the life span of mice (Pyo et al., 2013).

Calorie restriction is one physiological trigger for autophagy. Depletion of nucleocytosolic acetyl-CoA in yeast and knockdown of acetyl-CoA synthase in Drosophila melanogaster attenuates the global acetylation of proteins, activating autophagy to promote longevity (Eisenberg et al., 2014; Schroeder et al., 2014). Starvation-induced autophagy is associated with the reduced acetylation of Atg, tubulin, and microtubule-associated proteins (Lee and Finkel, 2009; Geeraert et al., 2010). Inhibition of endogenous E1A-binding protein p300 (EP300), an acetyl transferase and transcription coactivator, causes induction of forkhead box protein O promotor activity (Senf et al., 2011) and reduces acetylation of Atg5, Atg7, Atg8, and Atg12 (Lee and Finkel, 2009) A variety of agents that inhibit EP300 are used in traditional medicine, including curcumin (from the South Asian spice turmeric, Curcuma longa, one of the principal ingredients of curry powder), EGCG, garcinol, and spermidine (Pietrocola et al., 2015).

Spermidine belongs to the family of polyamines that can be found naturally in oranges, soybeans, rice bran, green pepper, broccoli, mushrooms, and green tea (Morselli et al., 2011). At the cellular level, polyamines interact with negatively charged molecules such as DNA, RNA, and lipids and regulate cell growth, proliferation, and death as well as lipid metabolism (Minois et al., 2011). Spermidine increases the life span in yeast Saccharomyces cerevisiae, nematode worm C. elegans, and fruit flies D. melanogaster (Eisenberg et al., 2009; Madeo et al., 2010; Minois et al., 2012). Autophagy has been identified as the main mechanism of action underlying the antiaging effects of spermidine (Minois, 2014). Administration of spermidine at low doses induces full autophagic responses and extends life span in yeast (Morselli et al., 2011). Spermidine-fed flies show enhanced autophagy and are protected from age-induced memory impairment (Gupta et al., 2013). In addition, spermidine inhibits acetyltransferases and affects global protein acetylation status by triggering deacetylation in the cytosol or acetylation in the nucleus (or both) (Eisenberg et al., 2009; Minois et al., 2012). The intracellular concentrations of polyamines decline with age in rodents and humans (Nishimura et al., 2006; Eisenberg et al., 2009). Supplementation with spermidine reduces age-associated pathology and mortality in aged mice (Soda et al., 2009; LaRocca et al., 2013). However, the evidence for spermidine to increase primate and human life spans has yet to be revealed. Recently, the endogenous levels of spermidine have been shown to be positively correlated with left ventricular ejection fraction and heart rate in humans (Meana et al., 2016). Whether supplementation of spermidine has beneficial effects for preventing age-associated diseases, especially cardiovascular diseases, needs further investigation.

Increasing NAD Levels to Reverse Aging

Calorie restriction increases mitochondrial NAD in order to promote survival (Yang et al., 2007). The salvage pathway for NAD biosynthesis begins with either nicotinamide (NAM) or nicotinic acid (NA), collectively known as niacin or vitamin B3 (Rongvaux et al., 2003). NAM is converted to nicotinamide mononucleotide (NMN) by nicotinamide phosphoribosyltransferase (NAMPT). The production of NAD from NMN and ATP is catalyzed by a family of NMN adenylyltransferases (NMNATs). In lower eukaryotes—including S. cerevisiae, D. melanogaster, and C. elegans—no NAMPT activity has been found. NAM is converted to NA, which then enters the parallel salvage pathway found in all eukaryotic species. Alternatively, nicotinamide riboside (NR) forms a precursor for NAD synthesis, connecting to the NAM salvage pathway through NMN. While the NAM salvage biosynthesis pathway is considered the main pathway for maintaining NAD levels, the latter can also be de novo synthesized from L-tryptophan (L-Trp). Note that the three major precursors for NAD biosynthesis (NAM, NA, and L-Trp) are all approved drugs or food supplements. NA has been a treatment option for lowering circulating lipid levels for many years (MacKay et al., 2012).

The intermediary metabolites previously mentioned regulate life span in different ways. Supraphysiological concentrations of NAD and NA extend life span in a fashion that depends on SIR-2.1, the nematodal ortholog of SIRT-1 (Hashimoto et al., 2010). By contrast, high concentration of NAM decreases nematodal life span, whereas low physiological doses of NAM or its metabolite, N1-methylnicotinamide (MNA), extend life span independently of SIR-2.1 (Schmeisser et al., 2013). Supplementation with NAM reduces chronic infection in humans (Bogan and Brenner, 2008), improves cognition in mice (Green et al., 2008; Gong et al., 2013), and increases longevity in yeast (Schmeisser et al., 2013). However, excessive NAM can cause immune intolerance and cancers (Bogan and Brenner, 2008). Nicotinamide N-methyltransferase (NNMT) is the mammalian enzyme that methylates NAM using S-adenosylmethionine as a methyl donor to generate MNA. In contrast to the discovery in lower organisms, Kraus et al. (2014) reported that knocking down NNMT protects against diet-induced obesity. Moreover, increased Nnmt expression is found in obese compared to nonobese human subjects, and the urinary MNA is elevated in humans and animals with obesity and type 2 diabetes (Salek et al., 2007).

Poly(ADP-ribose) polymerases (PARPs) catalyze the polymerization of ADP–ribose units of the target proteins with donor NAD molecules, which causes the formation of linear or branched polymers (Kim et al., 2005). A strong positive correlation between longevity and the polymer synthesis capacity of PARPs has been shown in mammalian cells (Beneke et al., 2010). Mice with PARP1 genetically knocked out exhibit faster aging than their wild-type counterparts (Piskunova et al., 2008). Thus, inhibition of PARPs has been proposed to increase NAD availability to elicit the anti-aging activity (El Ramy et al., 2009). Overall, maintaining high levels of NAD can increase the life span of the lower organisms by preventing primary aging, and it promotes the health spans of higher organisms.

Targeting SIRT-1 for CRM Discovery and Development

Sirtuins are a family of NAD-dependent protein deacetylases that exert diversified cellular functions by interacting with a wide range of signaling molecules, transcription factors, histones, and enzymes (Wang et al., 2012). Sir2 (silent information regulator 2), the first gene discovered in this family, was originally shown to extend life span in yeast and promote transcriptional silencing through deacetylation of the epsilon-amino groups of lysine in the amino-terminal domains of histones (Gottlieb and Esposito, 1989; Aparicio et al., 1991). Budding yeast cultured under low glucose conditions (i.e., calorie restriction) exhibits a significant extension of proliferative life span, which is Sir2-dependent (Kaeberlein et al., 1999). Depletion or mutation of Sir2 significantly reduces life span in yeast, worms, and flies (Longo and Kennedy, 2006; Chen and Guarente, 2007).

Over the last decade, sirtuins have attracted major attention due to their potential in expanding life span and protecting against age-associated disorders (Baur et al., 2012). Of all mammalian sirtuins, SIRT-1 is the ortholog most highly related to Sir2 (Law et al., 2009; Wang et al., 2011; Wang et al., 2012). Both proteins possess NAD-dependent deacetylase and ADP-ribosyltransferase activities and are upregulated by calorie restriction (Hassa et al., 2006; Kanfi et al., 2008). Genetic variations of SIRT-1 are associated with accelerated aging in human populations (Kuningas et al., 2007). Mice lacking SIRT-1 fail to show an increased activity and extended life span in response to calorie restriction (Chen et al., 2005; Boily et al., 2008). Small molecular activators of SIRT-1 replicate signaling pathways triggered by calorie restriction and extend life span in mice (Smith et al., 2009; Mitchell et al., 2014). In vitro overexpression of SIRT-3 shows protective benefit of calorie restriction by increasing NADPH levels against oxidative stress-induced cell death (Someya et al., 2010). Similarly, overexpression of SIRT-6 in male mice increases the maximal life span with reduced levels of IGF-1 in serum (Kanfi et al., 2012). Recent studies have shown that a genetic variant within the SIRT-1 gene promoter is not associated with longevity in elderly Italians (Albani et al., 2015), but single nucleotide polymorphisms of SIRT-3 are involved in the longevity of female elderly within the same population (Albani et al., 2014).

The anti-aging activity of SIRT-1 has been largely attributed to its role in regulating energy metabolism. The requirement of NAD as a cosubstrate implies that SIRT-1 acts as a sensor of cellular energy and reduction–oxidation status (Lin et al., 2004). SIRT-1 not only mediates the metabolic changes associated with calorie restriction but also protects against metabolic damage caused by chronic exposure to a high-fat diet (Pfluger et al., 2008). It is a principal regulator of energy homeostasis and systemic insulin sensitivity (Chang and Guarente, 2014). Mice overexpressing SIRT-1 are leaner and exhibit improved energy homeostasis (Bordone et al., 2007). Genetic variations of SIRT-1 in humans are associated with body mass index and adiposity, especially visceral obesity (Peeters et al., 2008; Shimoyama et al., 2011). Although the molecular basis underlying the antiaging functions of SIRT-1 remains to be elucidated, activators of this longevity element represent a promising class of drugs for the treatment of aging-related diseases (Imai and Guarente, 2014).

Tissue-Specific Activation of SIRT-1 Mediates the Beneficial Effects of Calorie Restriction

Aging is associated with a significant change of body fat composition that is characterized by a gradual decrease in fat free mass (especially skeletal muscle) and a significant increase in fat mass (especially in the abdominal cavity). These changes, referred to as sarcopenic obesity, contributes to impaired physical and psychological functions and the development of many aging-related ailments (Batsis et al., 2013). In fact, adiposity, particularly around the abdomen, is positively linked to reduced life expectancy (Bluher, 2008). While various factors such as decreased energy expenditure and alterations of hormone productions influence body fat distribution in aged individuals, as an endocrine and metabolic organ, adipose tissue per se actively participates in the aging process (Tchkonia et al., 2013). For example, accumulation of visceral fat (central obesity) leads to metabolic syndrome and shortens life span, in part by increasing the inflammatory factors and decreasing adiponectin, an insulin-sensitizing and cardiovascular protective hormone (Hanauer, 2005). In addition, fat redistribution is often associated with the dysregulation of fatty acid storage and release. The overflow of fatty acids in the form of diacylglycerols, ceramides, and long-chain acyl-coenzymes A exert adverse effects on systemic energy homeostasis and insulin sensitivity (Jensen, 2008; Jung and Choi, 2014). Since abnormal adipose tissue distribution and function are key components of a vicious cycle accelerating the aging process, adipose tissue engineering may represent an effective anti-aging approach. Indeed, many longevity modulators including calorie restriction and physical excise elicit their beneficial effects via regulating fat distribution and metabolism (Jung and Choi, 2014). Long-term calorie restriction decreases adiposity, elevates gene expression involved in energy metabolism, and reduces the expression of more than 50 proinflammatory genes in white adipose tissues (Higami et al., 2006).

Calorie restriction reduces body weight and fat mass, especially visceral fat, which is an indicator of metabolic disorders in mammals (Li et al., 2003; Ye and Keller, 2010). Calorie restriction-induced reduction of visceral fat leads to an increase in life span and reduced renal diseases in rodents (Muzumdar et al., 2008). Calorie restriction study in overweight men and women has shown that visceral fat is largely reduced in conjugation with other benefits of lowering metabolic rate, core body temperature, and insulin resistance as well as the prevention of cardiovascular diseases (Larson-Meyer et al., 2006; Lefevre et al., 2009). Specific depletion or expansion of the visceral fat depot using genetic or surgical tools in rodents have direct effects on aging-related disease risks (Huffman and Barzilai, 2009). Mice with genetically increased life spans (Ames dwarf, Snell dwarf, insulin growth hormone receptor, pregnancy-associated plasma protein A knockout mice) have substantially reduced visceral fat (Junnila et al., 2013; Menon et al., 2014). Elevated insulin sensitivity, reduced oxidative stress, augmented adiponectin production, significantly improved whole body glucose homeostasis, and lipid metabolism contribute to the increased life span in these mice.

SIRT-1 is a key mediator coordinating the physiological responses to calorie restriction. The expression and activity of SIRT-1 is dynamically regulated in major metabolic organs. Calorie restriction-induced SIRT-1 expression occurs predominantly in adipose tissues but not liver or muscle tissues (Chen et al., 2008). In fact, SIRT-1 exerts its effects on metabolism and insulin sensitivity in a tissue-specific manner (Wang et al., 2012). SIRT-1 in liver may not be the major contributor to systemic glucose homeostasis and insulin sensitivity (Rodgers et al., 2005; Erion et al., 2009; Purushotham et al., 2009) but appears to maintain hepatic lipid homeostasis and control the bile acid metabolism (Garcia-Rodriguez et al., 2014; Li et al., 2014). In skeletal muscle, SIRT-1 expression is decreased under insulin-resistant conditions (Sun et al., 2007). It regulates the differentiation and metabolism of muscle cells (Lee and Min, 2013). However, specific overexpression or knockout of SIRT-1 in skeletal muscle does not affect whole-body energy expenditure in mice on either a standard diet or energy-restricted diets (White et al., 2013).

The role of adipose SIRT-1 as an important regulator of energy metabolism and systemic insulin sensitivity has been suggested by both clinical and animal studies (Picard and Guarente, 2005; Wang et al., 2012). In humans, SIRT-1 expression is decreased in both obese and type 2 diabetic patients (Pedersen et al., 2008; Song et al., 2013). Extensive weight loss significantly increases SIRT-1 expression in adipose tissues (Moschen et al., 2013). In mice, both expression and activity of SIRT-1 in adipose tissues progressively decrease during aging but are markedly elevated by calorie restriction (Xu et al., 2013; Xu et al., 2015). SIRT-1 acts as a nutrient-dependent modulator to inhibit adipose tissue inflammation (Gillum et al., 2011; Chalkiadaki and Guarente, 2012; Kotas et al., 2013). In 3T3-L1 adipocytes, SIRT-1 promotes fat mobilization by interacting with corepressors NcoR and SMART (Picard et al., 2004). SIRT-1–dependent deacetylation of peroxisome proliferator activator receptor gamma (PPARγ) facilitates the brown remodeling of white adipose tissues (Qiang et al., 2012). Knockdown of SIRT-1 inhibits insulin-stimulated glucose uptake and GLUT4 translocation, whereas treatment of adipocytes with specific SIRT-1 activators leads to an increase in insulin-stimulated glucose uptake (Yoshizaki et al., 2009). SIRT-1 also modulates the expression and secretion of adiponectin (Qiao and Shao, 2006; Qiang et al., 2007). In mice, activation of SIRT-1 selectively in adipose tissue protects mice against metabolic deterioration during aging and facilitates energy homeostasis; these effects resemble those elicited by calorie restriction (Wang et al., 2012; Xu et al., 2013; Wang, 2014; Xu et al., 2015).

Intermediary Metabolites as Modulators of SIRT-1 Function

The expression and activity of SIRT-1 decline during aging and contribute to the development of age-related complications (Yuan et al., 2016). Thus, a forced SIRT-1 activation or induction represents a strategy for identifying and developing CRMs. SIRT-1 is dynamically regulated by intermediary metabolites. Its cofactor, the pyridine nucleotide NAD, metabolically acts as a hydride acceptor to form NADH (donor for oxidoreductases) and as an enzyme substrate. However, NAD usually presents at a much higher level than its Km for SIRT-1 activation, precluding this metabolite from being a rate-limiting factor for SIRT-1 functions in vivo (Sauve and Youn, 2012). On the other hand, NAD metabolites may be more dynamically involved in regulating SIRT-1 (Lee et al., 2008).

SIRT-1 activity can be increased by supplementation of NA or NAM for NAD biosynthesis (Yoshino et al., 2011) or by inhibiting the enzyme CD38 or PARP to reduce NAD consumption (Aksoy et al., 2006; de Picciotto et al., 2016). Supplementation with purine nucleotide metabolites of NAD, such as NR, enhances oxidative metabolism and protects against high-fat-diet–induced metabolic abnormalities in SIRT-1–dependent manners (Canto et al., 2012). In addition, modulating the activity of enzymes involved in NAD metabolic pathways such as NAMPT (Song et al., 2014), NNMT (Kraus et al., 2014), and PARP (Bai et al., 2011) exerts marked effects on energy metabolism in mice. Recent evidence has further expanded the scope of intermediary metabolites for SIRT-1 regulation, including various acyl-CoA donors (Feldman et al., 2012) and biotin and biotin-5ʹ-AMP (Xu et al., 2013; Wang, 2014).

The initial breakthrough of identification of Sir2 as a deacetylase with weak ADP–ribosyltransferase activity came along with the identification of the Salmonella typhimurium CobB protein as a Sir2 homolog (Tsang and Escalante-Semerena, 1998). CobB compensates for the lack of CobT mutants during vitamin B12 biosynthesis and possesses nicotinate mononucleotide (NaMN)–dependent phosphoribosyltransferase activity. Thus, CobB catalyzes the release of NA from NaMN, whereas Sir2 and SIRT-1 remove NAM from NAD. This evidence suggests that the biosynthesis or homeostasis of B vitamins [including vitamin B3 (NA/NAM) and B7 (biotin)] regulate the enzymatic activities of SIRT-1. Indeed, SIRT-1 can be potently inhibited by biotin and its metabolite biotinyl-5ʹ-AMP (Xu et al., 2013). Chronic biotin supplementation abolishes SIRT-1-mediated health benefits (Wang, 2014). In mice, biotin storage in adipose tissues increases with age, which is accompanied by a progressively decreased SIRT-1 activity (Xu et al., 2013). Calorie restriction activates SIRT-1 partly by preventing the accumulation of biotin in adipose tissue (Xu et al., 2013, 2015). In summary, the dynamic changes of endogenous intermediary metabolites that regulate SIRT-1 activity are potential pathways for rational design and development of CRMs.

SIRT-1–Activating Compounds Derived From Functional Foods

Various functional foods are able to boost SIRT-1’s activity or expression (Park et al., 2016). It is implicated in mitochondrial homeostasis, autophagy regulation, protein deacetylation, and longevity induced by a number of CRMs derived from functional foods (Lee et al., 2008; Mouchiroud et al., 2013; Ou et al., 2014). Resveratrol, known as 3,4ʹ,5-trihydroxystilbene, is a natural polyphenolic compound primarily found in plants (Fremont, 2000). It presents in either trans- or cis- isomers, where the trans-resveratrol has been widely studied and found enriched in grapes and grape products, berries, peanuts, and some herbal plants (Wenzel and Somoza, 2005; Giovinazzo et al., 2012). Many health benefits of trans-resveratrol have been reported, ranging from being cardio- and neuroprotective, antioxidant, antiinflammatory, antiatherosclerotic to anti-tumorigenic (Sale et al., 2004; Das and Maulik, 2006; Kwon et al., 2011; Smoliga et al., 2011).

Resveratrol is an activator of SIRT-1 and is able to induce AMPK, both of which are involved in its antiaging functions (Diaz-Ruiz et al., 2015). Resveratrol has been reported to increase DNA stability and extension of life span in yeast (Howitz et al., 2003). However, the effects on life span failed to be observed in higher organisms such as D. melanogaster (Bass et al., 2007) and rodents (Pearson et al., 2008; Miller et al., 2011). Nevertheless, resveratrol exerts beneficial effects on secondary aging—that is, the prevention of age-associated diseases and functional loss. Resveratrol-treated mice had lower total cholesterol levels in plasma and reduced incidence of atherosclerotic lesion formation when compared to control counterparts (Do et al., 2008). Treatment with resveratrol in the aged mice activated SIRT-1 expression, where the subsequent apoptotic pathway involving forkhead box protein O3 was inhibited (Lin et al., 2014). It increases glucose uptake in response to insulin stimulation in skeletal muscle, adipocytes, and hepatocytes via activation of SIRT-1 and AMPK (Sun et al., 2007; Breen et al., 2008). Resveratrol can penetrate the blood–brain barrier to reach the central nervous system. In the animal model of Alzheimer’s disease, resveratrol-treated mice show a delayed onset as well as a slow progression of impaired memory (Kim et al., 2007; Venigalla et al., 2016) due partly at least to SIRT-1-mediated inhibition of nuclear factor kappa B (NFκB) signaling (Chen et al., 2005). Resveratrol rescued SIRT-1 activity suppressed by betaamyloid peptide 25–35 in a dose-dependent manner, which led to significant attenuation of apoptosis in mouse neurons (Feng et al., 2013; Witte et al., 2014; Zhang et al., 2014; Turner et al., 2015).

Fisetin (3, 7, 3′, 4′-tetrahydroxyflavone) is a flavonol that can be found in vegetables and fruits such as strawberries and apples (Kim et al., 2015). It increases the expression of adiponectin in a dose-dependent manner to protect obesity-related malfunctions and vascular abnormalities (Jin et al., 2014). By suppressing PPARs, it regulates cell metabolism (Kim et al., 2015). Mechanistically, fisetin not only promotes SIRT-1 activity to mediate the deacetylation of PPARγ and FOXO1 but also enhances the association of SIRT-1 with the promoter of PPARγ, which results in attenuation of adipogenesis (Kim et al., 2015). Oligonol contains catechin-type oligomers that are found in lychee fruit and grape seed (Fujii et al., 2007). Mice treated with oligonol exhibit reduced serum lipid concentration, improved kidney function, and attenuated cognitive impairment (Noh et al., 2010; Sakurai et al., 2013). By upregulation of SIRT-1 expression, oligonol attenuates mitochondrial superoxide formation and activates autophagy (Park et al., 2016).

Melatonin (known as N-acetyl 5-methoxy tryptamine) is an indole hormone that is expressed by various plants and animals (Reiter et al., 2013). Plants synthesize melatonin from the essential amino acid L-trytophan, but melatonin in animals is mainly secreted from the pineal gland but is also present in skin, the immune system, the reproductive system, and the gastrointestinal tract (Slominski et al., 2002; Acuna-Castroviejo et al., 2014; Ramis et al., 2015). However, the absorption of melatonin is low in humans because the plasma levels of melatonin after the ingestion of melatonin-rich food are far less than the amount of melatonin ingested (Kennaway, 2015). Melatonin regulates circadian cycle (Hardeland et al., 2012) and elicits antioxidant (Galano et al., 2013) and anti-inflammatory activities (Agil et al., 2013). The circadian rhythm of melatonin is disturbed and deteriorated by aging (Waller et al., 2016). The loss of melatonin rhythm in the elderly not only causes sleep disorders but also leads to desynchronization of gene expression that affects general well-being (Jung-Hynes et al., 2010). Melatonin increases the expression of SIRT-1 protein and decreases the levels of acetylated p53 and NFκB in mice prone to accelerated senescence (Gutierrez-Cuesta et al., 2008). However, melatonin exhibits anti-tumor activity by downregulating SIRT-1 and upregulating acetylated p53 in human osteosarcoma cells (Cheng et al., 2013). Whether or not melatonin-mediated SIRT-1 expressions are specific to certain cell types warrants further study.

Concluding Remarks and Perspectives

The average life expectancy has been creeping upward in most developed countries for many years. While the ultimate goal of science and technology is to extend the life span beyond its current natural limits, the contemporary challenge of anti-aging program is to eliminate or reduce aging-associated medical burdens from the world’s leading causes of death, including cancer and cardiovascular, neurodegenerative, musculoskeletal, and metabolic diseases. To focus on molecules that mediate the beneficial effects of caloric restriction represents one of the most straightforward approaches for developing anti-aging remedies. Accordingly, a bunch of CRMs emerge for either depletion of the energy supply or targeting specific signaling pathways such as autophagy. Functional foods represent the most promising source of CRMs. In particular, those that activate the expression or function of SIRT-1 hold great potential to delay the onset of aging and age-related diseases. In this respect, actions specific to adipose tissue by SIRT-1 are directly related to the anti-aging effects of caloric restriction. Adipose tissue is the body’s largest endocrine organ that secretes inflammatory and immune mediators. During aging, the most prominent changes in our body are the loss of functional lean (fat-free) mass and the accumulation of fat, especially in the abdominal region (Jackson et al., 2012). From the ages of 40 to 79, visceral fat in men increased by 42.9% and by 65.3% in women, whereas the fat-free mass decreased by 6.6% to 23.3% in both genders (Yamada et al., 2014). This phenomenon, referred as sarcopenic obesity, is assuming a prominent role in aging-related diseases (Prado et al., 2012). The important role of adipose tissues in human aging provides a more convenient but focused strategy for discovering SIRT-1–activating compounds in functional foods.

References

1. Acuna-Castroviejo D, Escames G, Venegas C, et al. Extrapineal melatonin: sources, regulation, and potential functions. Cell Mol Life Sci. 2014;71(16):2997–3025.

2. Agil A, Reiter RJ, Jimenez-Aranda A, et al. Melatonin ameliorates low-grade inflammation and oxidative stress in young Zucker diabetic fatty rats. J Pineal Res. 2013;54(4):381–388.

3. Akram M. Citric acid cycle and role of its intermediates in metabolism. Cell Biochem Biophys. 2014;68(3):475–478.

4. Aksoy P, Escande C, White TA, et al. Regulation of SIRT 1 mediated NAD dependent deacetylation: a novel role for the multifunctional enzyme CD38. Biochem Biophys Res Commun. 2006;349(1):353–359.

5. Albani D, Ateri E, Mazzuco S, et al. Modulation of human longevity by SIRT3 single nucleotide polymorphisms in the prospective study “Treviso Longeva (TRELONG).”. Age (Dordr). 2014;36(1):469–478.

6. Albani D, Mazzuco S, Chierchia A, et al. The SIRT1 promoter polymorphic site rs12778366 increases IL-6 related human mortality in the prospective study “Treviso Longeva (TRELONG).”. Int J Mol Epidemiol Genet. 2015;6(1):20–26.

7. Anderson JW, Herman RH. Effect of fasting, caloric restriction, and refeeding on glucose tolerance of normal men. Am J Clin Nutr. 1972;25(1):41–52.

8. Anstey K, Stankov L, Lord S. Primary aging, secondary aging, and intelligence. Psychol Aging. 1993;8(4):562–570.

9. Aparicio OM, Billington BL, Gottschling DE. Modifiers of position effect are shared between telomeric and silent mating-type loci in S cerevisiae. Cell. 1991;66(6):1279–1287.

10. Bai P, Canto C, Oudart H, et al. PARP-1 inhibition increases mitochondrial metabolism through SIRT1 activation. Cell Metab. 2011;13(4):461–468.

11. Bao J, Sack MN. Protein deacetylation by sirtuins: delineating a post-translational regulatory program responsive to nutrient and redox stressors. Cell Mol Life Sci. 2010;67(18):3073–3087.

12. Bass TM, Weinkove D, Houthoofd K, Gems D, Partridge L. Effects of resveratrol on lifespan in Drosophila melanogaster and Caenorhabditis elegans. Mech Ageing Dev. 2007;128(10):546–552.

13. Batsis JA, Barre LK, Mackenzie TA, Pratt SI, Lopez-Jimenez F, Bartels SJ. Variation in the prevalence of sarcopenia and sarcopenic obesity in older adults associated with different research definitions: dual-energy X-ray absorptiometry data from the National Health and Nutrition Examination Survey 1999–2004. J Am Geriatr Soc. 2013;61(6):974–980.

14. Baur JA, Ungvari Z, Minor RK, Le Couteur DG, de Cabo R. Are sirtuins viable targets for improving healthspan and lifespan? Nat Rev Drug Discov. 2012;11(6):443–461.

15. Beneke S, Scherr AL, Ponath V, Popp O, Burkle A. Enzyme characteristics of recombinant poly(ADP-ribose) polymerases-1 of rat and human origin mirror the correlation between cellular poly(ADP-ribosyl)ation capacity and species-specific life span. Mech Ageing Dev. 2010;131(5):366–369.

16. Bluher M. Fat tissue and long life. Obes Facts. 2008;1(4):176–182.

17. Bogan KL, Brenner C. Nicotinic acid, nicotinamide, and nicotinamide riboside: a molecular evaluation of NAD+precursor vitamins in human nutrition. Annu Rev Nutr. 2008;28:115–130.

18. Boily G, Seifert EL, Bevilacqua L, et al. SirT1 regulates energy metabolism and response to caloric restriction in mice. PLoS One. 2008;3(3):e1759.

19. Bordone L, Cohen D, Robinson A, et al. SIRT1 transgenic mice show phenotypes resembling calorie restriction. Aging Cell. 2007;6(6):759–767.

20. Breen DM, Sanli T, Giacca A, Tsiani E. Stimulation of muscle cell glucose uptake by resveratrol through sirtuins and AMPK. Biochem Biophys Res Commun. 2008;374(1):117–122.

21. Cai L, Sutter BM, Li B, Tu BP. Acetyl-CoA induces cell growth and proliferation by promoting the acetylation of histones at growth genes. Mol Cell. 2011;42(4):426–437.

22. Canto C, Houtkooper RH, Pirinen E, et al. The NAD(+) precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity. Cell Metab. 2012;15(6):838–847.

23. Carames B, Kiosses WB, Akasaki Y, et al. Glucosamine activates autophagy in vitro and in vivo. Arthritis Rheum. 2013;65(7):1843–1852.

24. Cavallini G, Donati A, Taddei M, Bergamini E. Evidence for selective mitochondrial autophagy and failure in aging. Autophagy. 2007;3(1):26–27.

25. Cetrullo S, D’Adamo S, Tantini B, Borzi RM, Flamigni F. mTOR, AMPK, and Sirt1: key players in metabolic stress management. Crit Rev Eukaryot Gene Expr. 2015;25(1):59–75.

26. Chalkiadaki A, Guarente L. High-fat diet triggers inflammation-induced cleavage of SIRT1 in adipose tissue to promote metabolic dysfunction. Cell Metab. 2012;16(2):180–188.

27. Chang HC, Guarente L. SIRT1 and other sirtuins in metabolism. Trends Endocrinol Metab. 2014;25(3):138–145.

28. Chen D, Guarente L. SIR2: a potential target for calorie restriction mimetics. Trends Mol Med. 2007;13(2):64–71.

29. Chen D, Bruno J, Easlon E, et al. Tissue-specific regulation of SIRT1 by calorie restriction. Genes Dev. 2008;22(13):1753–1757.

30. Chen J, Zhou Y, Mueller-Steiner S, et al. SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-kappaB signaling. J Biol Chem. 2005;280(48):40364–40374.

31. Cheng Y, Cai L, Jiang P, et al. SIRT1 inhibition by melatonin exerts antitumor activity in human osteosarcoma cells. Eur J Pharmacol. 2013;715(1-3):219–229.

32. Choi KM, Han KA, Ahn HJ, et al. The effects of caloric restriction on fetuin-A and cardiovascular risk factors in rats and humans: a randomized controlled trial. Clin Endocrinol (Oxf). 2013;79(3):356–363.

33. Choudhary C, Weinert BT, Nishida Y, Verdin E, Mann M. The growing landscape of lysine acetylation links metabolism and cell signalling. Nat Rev Mol Cell Biol. 2014;15(8):536–550.

34. Colman RJ, Anderson RM, Johnson SC, et al. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science. 2009;325(5937):201–204.

35. Colman RJ, Beasley TM, Kemnitz JW, Johnson SC, Weindruch R, Anderson RM. Caloric restriction reduces age-related and all-cause mortality in rhesus monkeys. Nat Commun. 2014;5:3557.

36. Cui J, Shi S, Sun X, et al. Mitochondrial autophagy involving renal injury and aging is modulated by caloric intake in aged rat kidneys. PLoS One. 2013;8(7):e69720.

37. Dabhade P, Kotwal S. Tackling the aging process with bio-molecules: a possible role for caloric restriction, food-derived nutrients, vitamins, amino acids, peptides, and minerals. J Nutr Gerontol Geriatr. 2013;32(1):24–40.

38. Das DK, Maulik N. Resveratrol in cardioprotection: a therapeutic promise of alternative medicine. Mol Interval. 2006;6(1):36–47.

39. de Picciotto NE, Gano LB, Johnson LC, et al. Nicotinamide mononucleotide supplementation reverses vascular dysfunction and oxidative stress with aging in mice. Aging Cell. 2016.

40. Diaz-Ruiz C, Rodriguez-Perez AI, Beiroa D, Rodriguez-Pallares J, Labandeira-Garcia JL. Reciprocal regulation between sirtuin-1 and angiotensin-II in the substantia nigra: implications for aging and neurodegeneration. Oncotarget. 2015;6(29):26675–26689.

41. Do GM, Kwon EY, Kim HJ, et al. Long-term effects of resveratrol supplementation on suppression of atherogenic lesion formation and cholesterol synthesis in apo E-deficient mice. Biochem Biophys Res Commun. 2008;374(1):55–59.

42. Duan W, Mattson MP. Dietary restriction and 2-deoxyglucose administration improve behavioral outcome and reduce degeneration of dopaminergic neurons in models of Parkinson’s disease. J Neurosci Res. 1999;57(2):195–206.

43. Eisenberg T, Knauer H, Schauer A, et al. Induction of autophagy by spermidine promotes longevity. Nat Cell Biol. 2009;11(11):1305–1314.

44. Eisenberg T, Schroeder S, Andryushkova A, et al. Nucleocytosolic depletion of the energy metabolite acetyl-coenzyme a stimulates autophagy and prolongs lifespan. Cell Metab. 2014;19(3):431–444.

45. El Ramy R, Magroun N, Messadecq N, et al. Functional interplay between Parp-1 and SirT1 in genome integrity and chromatin-based processes. Cell Mol Life Sci. 2009;66(19):3219–3234.

46. Erion DM, Yonemitsu S, Nie Y, et al. SirT1 knockdown in liver decreases basal hepatic glucose production and increases hepatic insulin responsiveness in diabetic rats. Proc Natl Acad Sci USA. 2009;106(27):11288–11293.

47. Feldman JL, Dittenhafer-Reed KE, Denu JM. Sirtuin catalysis and regulation. J Biol Chem. 2012;287(51):42419–42427.

48. Feng X, Liang N, Zhu D, et al. Resveratrol inhibits beta-amyloid-induced neuronal apoptosis through regulation of SIRT1-ROCK1 signaling pathway. PLoS One. 2013;8(3):e59888.

49. Fernandes G, Good RA. Inhibition by restricted-calorie diet of lymphoproliferative disease and renal damage in MRL/lpr mice. Proc Natl Acad Sci USA. 1984;81(19):6144–6148.

50. Fontana L, Partridge L, Longo VD. Extending healthy life span-from yeast to humans. Science. 2010;328(5976):321–326.

51. Fremont L. Biological effects of resveratrol. Life Sci. 2000;66(8):663–673.

52. Fujii H, Nakagawa T, Nishioka H, et al. Preparation, characterization, and antioxidative effects of oligomeric proanthocyanidin-L-cysteine complexes. J Agric Food Chem. 2007;55(4):1525–1531.

53. Galano A, Tan DX, Reiter RJ. On the free radical scavenging activities of melatonin’s metabolites, AFMK and AMK. J Pineal Res. 2013;54(3):245–257.

54. Garcia-Rodriguez JL, Barbier-Torres L, Fernandez-Alvarez S, et al. SIRT1 controls liver regeneration by regulating bile acid metabolism through farnesoid X receptor and mammalian target of rapamycin signaling. Hepatology. 2014;59(5):1972–1983.

55. Geeraert C, Ratier A, Pfisterer SG, et al. Starvation-induced hyperacetylation of tubulin is required for the stimulation of autophagy by nutrient deprivation. J Biol Chem. 2010;285(31):24184–24194.

56. Gillum MP, Kotas ME, Erion DM, et al. SirT1 regulates adipose tissue inflammation. Diabetes. 2011;60(12):3235–3245.

57. Giovinazzo G, Ingrosso I, Paradiso A, De Gara L, Santino A. Resveratrol biosynthesis: plant metabolic engineering for nutritional improvement of food. Plant Foods Hum Nutr. 2012;67(3):191–199.

58. Gong B, Pan Y, Vempati P, et al. Nicotinamide riboside restores cognition through an upregulation of proliferator-activated receptor-gamma coactivator 1alpha regulated beta-secretase 1 degradation and mitochondrial gene expression in Alzheimer’s mouse models. Neurobiol Aging. 2013;34(6):1581–1588.

59. Gottlieb S, Esposito RE. A new role for a yeast transcriptional silencer gene, SIR2, in regulation of recombination in ribosomal DNA. Cell. 1989;56(5):771–776.

60. Green KN, Steffan JS, Martinez-Coria H, et al. Nicotinamide restores cognition in Alzheimer’s disease transgenic mice via a mechanism involving sirtuin inhibition and selective reduction of Thr231-phosphotau. J Neurosci. 2008;28(45):11500–11510.

61. Guo Z, Lee J, Lane M, Mattson M. Iodoacetate protects hippocampal neurons against excitotoxic and oxidative injury: involvement of heat-shock proteins and Bcl-2. J Neurochem. 2001;79(2):361–370.

62. Gupta VK, Scheunemann L, Eisenberg T, et al. Restoring polyamines protects from age-induced memory impairment in an autophagy-dependent manner. Nat Neurosci. 2013;16(10):1453–1460.

63. Gutierrez-Cuesta J, Tajes M, Jimenez A, Coto-Montes A, Camins A, Pallas M. Evaluation of potential pro-survival pathways regulated by melatonin in a murine senescence model. J Pineal Res. 2008;45(4):497–505.

64. Hackenschmidt J, Barth C, Decker K. Stimulation of acetyl-CoA carboxylase by (-)-Hydroxycitrate. FEBS Lett. 1972;27(1):131–133.

65. Hanauer SB. Obesity and visceral fat: a growing inflammatory disease. Nat Clin Pract Gastroenterol Hepatol. 2005;2(6):245.

66. Hardeland R, Madrid JA, Tan DX, Reiter RJ. Melatonin, the circadian multioscillator system and health: the need for detailed analyses of peripheral melatonin signaling. J Pineal Res. 2012;52(2):139–166.

67. Hartleben B, Godel M, Meyer-Schwesinger C, et al. Autophagy influences glomerular disease susceptibility and maintains podocyte homeostasis in aging mice. J Clin Investig. 2010;120(4):1084–1096.

68. Hashimoto T, Horikawa M, Nomura T, Sakamoto K. Nicotinamide adenine dinucleotide extends the lifespan of Caenorhabditis elegans mediated by sir-2.1 and daf-16. Biogerontology. 2010;11(1):31–43.

69. Hassa PO, Haenni SS, Elser M, Hottiger MO. Nuclear ADP-ribosylation reactions in mammalian cells: where are we today and where are we going? Microbiol Mol Biol Rev. 2006;70(3):789–829.

70. Heilbronn LK, Ravussin E. Calorie restriction and aging: review of the literature and implications for studies in humans. Am J Clin Nutr. 2003;78(3):361–369.

71. Heilbronn LK, Smith SR, Martin CK, Anton SD, Ravussin E. Alternate-day fasting in nonobese subjects: effects on body weight, body composition, and energy metabolism. Am J Clin Nutr. 2005;81(1):69–73.

72. Higami Y, Barger JL, Page GP, et al. Energy restriction lowers the expression of genes linked to inflammation, the cytoskeleton, the extracellular matrix, and angiogenesis in mouse adipose tissue. J Nutr. 2006;136(2):343–352.

73. Holloszy JO, Fontana L. Caloric restriction in humans. Exp Gerontol. 2007;42(8):709–712.

74. Howitz KT, Bitterman KJ, Cohen HY, et al. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature. 2003;425(6954):191–196.

75. Huffman DM, Barzilai N. Role of visceral adipose tissue in aging. Biochim Biophys Acta. 2009;1790(10):1117–1123.

76. Imai S, Guarente L. NAD+and sirtuins in aging and disease. Trends Cell Biol. 2014;24(8):464–471.

77. Ingram DK, Roth GS. Glycolytic inhibition as a strategy for developing calorie restriction mimetics. Exp Gerontol. 2011;46(2-3):148–154.

78. Ingram DK, Roth GS. Calorie restriction mimetics: can you have your cake and eat it, too? Ageing Res Rev. 2015;20:46–62.

79. Ingram DK, Weindruch R, Spangler EL, Freeman JR, Walford RL. Dietary restriction benefits learning and motor performance of aged mice. J Gerontol. 1987;42(1):78–81.

80. Ingram DK, Zhu M, Mamczarz J, et al. Calorie restriction mimetics: an emerging research field. Aging Cell. 2006;5(2):97–108.

81. Jackson MF, Luong D, Vang DD, et al. The aging myostatin null phenotype: reduced adiposity, cardiac hypertrophy, enhanced cardiac stress response, and sexual dimorphism. J Endocrinol. 2012;213(3):263–275.

82. Jensen MD. Role of body fat distribution and the metabolic complications of obesity. J Clin Endocrinol Metab. 2008;93(11 Suppl 1):S57–S63.

83. Jin T, Kim OY, Shin MJ, et al. Fisetin up-regulates the expression of adiponectin in 3T3-L1 adipocytes via the activation of silent mating type information regulation 2 homologue 1 (SIRT1)-deacetylase and peroxisome proliferator-activated receptors (PPARs). J Agric Food Chem. 2014;62(43):10468–10474.

84. Jung-Hynes B, Reiter RJ, Ahmad N. Sirtuins, melatonin and circadian rhythms: building a bridge between aging and cancer. J Pineal Res. 2010;48(1):9–19.

85. Jung UJ, Choi MS. Obesity and its metabolic complications: the role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease. Int J Mol Sci. 2014;15(4):6184–6223.

86. Junnila RK, List EO, Berryman DE, Murrey JW, Kopchick JJ. The GH/IGF-1 axis in ageing and longevity. Nat Rev Endocrinol. 2013;9(6):366–376.

87. 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(19):2570–2580.

88. Kanfi Y, Peshti V, Gozlan YM, Rathaus M, Gil R, Cohen HY. Regulation of SIRT1 protein levels by nutrient availability. FEBS Lett. 2008;582(16):2417–2423.

89. Kanfi Y, Naiman S, Amir G, et al. The sirtuin SIRT6 regulates lifespan in male mice. Nature. 2012;483(7388):218–221.

90. Kennaway DJ. Are the proposed benefits of melatonin-rich foods too hard to swallow? Crit Rev Food Sci Nutr. 2015.

91. Kim D, Nguyen MD, Dobbin MM, et al. SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer’s disease and amyotrophic lateral sclerosis. EMBO J. 2007;26(13):3169–3179.

92. Kim MY, Zhang T, Kraus WL. Poly(ADP-ribosyl)ation by PARP-1: ‘PAR-laying’ NAD+into a nuclear signal. Genes Dev. 2005;19(17):1951–1967.

93. Kim SC, Kim YH, Son SW, Moon EY, Pyo S, Um SH. Fisetin induces Sirt1 expression while inhibiting early adipogenesis in 3T3-L1 cells. Biochem Biophys Res Commun. 2015;467(4):638–644.

94. Korla K, Mitra CK. Modelling the Krebs cycle and oxidative phosphorylation. J Biomol Struct Dyn. 2014;32(2):242–256.

95. Kornberg H. Krebs and his trinity of cycles. Nat Rev Mol Cell Biol. 2000;1(3):225–228.

96. Kotas ME, Gorecki MC, Gillum MP. Sirtuin-1 is a nutrient-dependent modulator of inflammation. Adipocyte. 2013;2(2):113–118.

97. Kraus D, Yang Q, Kong D, et al. Nicotinamide N-methyltransferase knockdown protects against diet-induced obesity. Nature. 2014;508(7495):258–262.

98. Kuningas M, Putters M, Westendorp RG, Slagboom PE, van Heemst D. SIRT1 gene, age-related diseases, and mortality: the Leiden 85-plus study. J Gerontol A Biol Sci Med Sci. 2007;62(9):960–965.

99. Kwon KJ, Kim JN, Kim MK, et al. Melatonin synergistically increases resveratrol-induced heme oxygenase-1 expression through the inhibition of ubiquitin-dependent proteasome pathway: a possible role in neuroprotection. J Pineal Res. 2011;50(2):110–123.

100. Lane MA, Baer DJ, Rumpler WV, et al. Calorie restriction lowers body temperature in rhesus monkeys, consistent with a postulated anti-aging mechanism in rodents. Proc Natl Acad Sci USA. 1996;93(9):4159–4164.

101. LaRocca TJ, Gioscia-Ryan RA, Hearon Jr CM, Seals DR. The autophagy enhancer spermidine reverses arterial aging. Mech Ageing Dev. 2013;134(7-8):314–320.

102. Larson-Meyer DE, Heilbronn LK, Redman LM, et al. Effect of calorie restriction with or without exercise on insulin sensitivity, beta-cell function, fat cell size, and ectopic lipid in overweight subjects. Diabetes Care. 2006;29(6):1337–1344.

103. Law IK, Liu L, Xu A, et al. Identification and characterization of proteins interacting with SIRT1 and SIRT3: implications in the anti-aging and metabolic effects of sirtuins. Proteomics. 2009;9(9):2444–2456.

104. Lee IH, Cao L, Mostoslavsky R, et al. A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Proc Natl Acad Sci USA. 2008;105(9):3374–3379.

105. Lee IH, Finkel T. Regulation of autophagy by the p300 acetyltransferase. J Biol Chem. 2009;284(10):6322–6328.

106. Lee J, Bruce-Keller AJ, Kruman Y, Chan SL, Mattson MP. 2-Deoxy-D-glucose protects hippocampal neurons against excitotoxic and oxidative injury: evidence for the involvement of stress proteins. J Neurosci Res. 1999;57(1):48–61.

107. Lee SH, Min KJ. Caloric restriction and its mimetics. BMB Rep. 2013;46(4):181–187.

108. Lefevre M, Redman LM, Heilbronn LK, et al. Caloric restriction alone and with exercise improves CVD risk in healthy non-obese individuals. Atherosclerosis. 2009;203(1):206–213.

109. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008;132(1):27–42.

110. Li Y, Bujo H, Takahashi K, et al. Visceral fat: higher responsiveness of fat mass and gene expression to calorie restriction than subcutaneous fat. Exp Biol Med (Maywood). 2003;228(10):1118–1123.

111. Li Y, Wong K, Giles A, et al. Hepatic SIRT1 attenuates hepatic steatosis and controls energy balance in mice by inducing fibroblast growth factor 21. Gastroenterology. 2014;146(2):539–549 e537.

112. Libert S, Guarente L. Metabolic and neuropsychiatric effects of calorie restriction and sirtuins. Annu Rev Physiol. 2013;75:669–684.

113. Lin CH, Lin CC, Ting WJ, et al. Resveratrol enhanced FOXO3 phosphorylation via synergetic activation of SIRT1 and PI3K/Akt signaling to improve the effects of exercise in elderly rat hearts. Age (Dordr). 2014;36(5):9705.

114. Lin SJ, Ford E, Haigis M, Liszt G, Guarente L. Calorie restriction extends yeast life span by lowering the level of NADH. Genes Dev. 2004;18(1):12–16.

115. Longo VD, Kennedy BK. Sirtuins in aging and age-related disease. Cell. 2006;126(2):257–268.

116. MacKay D, Hathcock J, Guarneri E. Niacin: chemical forms, bioavailability, and health effects. Nutr Rev. 2012;70(6):357–366.

117. Madeo F, Eisenberg T, Buttner S, Ruckenstuhl C, Kroemer G. Spermidine: a novel autophagy inducer and longevity elixir. Autophagy. 2010;6(1):160–162.

118. Madeo F, Pietrocola F, Eisenberg T, Kroemer G. Caloric restriction mimetics: towards a molecular definition. Nat Rev Drug Discov. 2014;13(10):727–740.

119. Mai V, Colbert LH, Berrigan D, et al. Calorie restriction and diet composition modulate spontaneous intestinal tumorigenesis in Apc(Min) mice through different mechanisms. Cancer Res. 2003;63(8):1752–1755.

120. Marino G, Pietrocola F, Eisenberg T, et al. Regulation of autophagy by cytosolic acetyl-coenzyme A. Mol Cell. 2014;53(5):710–725.

121. Marshall S, Yamasaki K, Okuyama R. Glucosamine induces rapid desensitization of glucose transport in isolated adipocytes by increasing GlcN-6-P levels. Biochem Biophys Res Commun. 2005;329(3):1155–1161.

122. Masri S. Sirtuin-dependent clock control: new advances in metabolism, aging and cancer. Curr Opin Clin Nutr Metab Care. 2015;18(6):521–527.

123. Mattison JA, Roth GS, Beasley TM, et al. Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. Nature. 2012;489(7415):318–321.

124. Mattson MP. Neuroprotective signaling and the aging brain: take away my food and let me run. Brain Res. 2000;886(1-2):47–53.

125. McCay CM, Crowell MF, Maynard LA. The effect of retarded growth upon the length of the life span and upon the ultimate body size. J Nutr. 1935;10:63–70.

126. McKnight LL, Flickinger EA, France J, Davenport GM, Shoveller AK. Mannoheptulose has differential effects on fasting and postprandial energy expenditure and respiratory quotient in adult Beagle dogs fed diets of different macronutrient contents. J Nutr Sci. 2014;3:e17.

127. Meana C, Rubin JM, Bordallo C, Suarez L, Bordallo J, Sanchez M. Correlation between endogenous polyamines in human cardiac tissues and clinical parameters in patients with heart failure. J Cell Mol Med. 2016;20(2):302–312.

128. Menon V, Zhi X, Hossain T, et al. The contribution of visceral fat to improved insulin signaling in Ames dwarf mice. Aging Cell. 2014;13(3):497–506.

129. Miller RA, Harrison DE, Astle CM, et al. Rapamycin, but not resveratrol or simvastatin, extends life span of genetically heterogeneous mice. J Gerontol A Biol Sci Med Sci. 2011;66(2):191–201.

130. Minois N. Molecular basis of the ‘anti-aging’ effect of spermidine and other natural polyamines - a mini-review. Gerontology. 2014;60(4):319–326.

131. Minois N, Carmona-Gutierrez D, Bauer MA, et al. Spermidine promotes stress resistance in Drosophila melanogaster through autophagy-dependent and -independent pathways. Cell Death Dis. 2012;3:e401.

132. Minois N, Carmona-Gutierrez D, Madeo F. Polyamines in aging and disease. Aging (Albany NY). 2011;3(8):716–732.

133. Mitchell SE, Tang Z, Kerbois C, et al. The effects of graded levels of calorie restriction: I impact of short term calorie and protein restriction on body composition in the C57BL/6 mouse. Oncotarget. 2015;6(18):15902–15930.

134. Mitchell SJ, Martin-Montalvo A, Mercken EM, et al. The SIRT1 activator SRT1720 extends lifespan and improves health of mice fed a standard diet. Cell Rep. 2014;6(5):836–843.

135. Morselli E, Marino G, Bennetzen MV, et al. Spermidine and resveratrol induce autophagy by distinct pathways converging on the acetylproteome. J Cell Biol. 2011;192(4):615–629.

136. Moschen AR, Wieser V, Gerner RR, et al. Adipose tissue and liver expression of SIRT1, 3, and 6 increase after extensive weight loss in morbid obesity. J Hepatol. 2013;59(6):1315–1322.

137. Mouchiroud L, Houtkooper RH, Moullan N, et al. The NAD(+)/Sirtuin pathway modulates longevity through activation of mitochondrial UPR and FOXO signaling. Cell. 2013;154(2):430–441.

138. Muzumdar R, Allison DB, Huffman DM, et al. Visceral adipose tissue modulates mammalian longevity. Aging Cell. 2008;7(3):438–440.

139. Nishimura K, Shiina R, Kashiwagi K, Igarashi K. Decrease in polyamines with aging and their ingestion from food and drink. J Biochem. 2006;139(1):81–90.

140. Noh JS, Kim HY, Park CH, Fujii H, Yokozawa T. Hypolipidaemic and antioxidative effects of oligonol, a low-molecular-weight polyphenol derived from lychee fruit, on renal damage in type 2 diabetic mice. Br J Nutr. 2010;104(8):1120–1128.

141. Ou X, Lee MR, Huang X, Messina-Graham S, Broxmeyer HE. SIRT1 positively regulates autophagy and mitochondria function in embryonic stem cells under oxidative stress. Stem Cells. 2014;32(5):1183–1194.

142. Park SK, Seong RK, Kim JA, et al. Oligonol promotes anti-aging pathways via modulation of SIRT1-AMPK-Autophagy Pathway. Nutr Res Pract. 2016;10(1):3–10.

143. Pearson KJ, Baur JA, Lewis KN, et al. Resveratrol delays age-related deterioration and mimics transcriptional aspects of dietary restriction without extending life span. Cell Metab. 2008;8(2):157–168.

144. Peeters AV, Beckers S, Verrijken A, et al. Association of SIRT1 gene variation with visceral obesity. Hum Genet. 2008;124(4):431–436.

145. Pedersen PL. Warburg, me and Hexokinase 2: Multiple discoveries of key molecular events underlying one of cancers’ most common phenotypes, the “Warburg Effect,” i.e., elevated glycolysis in the presence of oxygen. J Bioenergy Biomembr. 2007;39(3):211–222.

146. Pedersen SB, Olholm J, Paulsen SK, Bennetzen MF, Richelsen B. Low Sirt1 expression, which is upregulated by fasting, in human adipose tissue from obese women. Int J Obes (Lond). 2008;32(8):1250–1255.

147. Pfluger PT, Herranz D, Velasco-Miguel S, Serrano M, Tschop MH. Sirt1 protects against high-fat diet-induced metabolic damage. Proc Natl Acad Sci USA. 2008;105(28):9793–9798.

148. Picard F, Guarente L. Molecular links between aging and adipose tissue. Int J Obes (Lond). 2005;29(Suppl 1):S36–S39.

149. Picard F, Kurtev M, Chung N, et al. Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-gamma. Nature. 2004;429(6993):771–776.

150. Pietrocola F, Lachkar S, Enot DP, et al. Spermidine induces autophagy by inhibiting the acetyltransferase EP300. Cell Death Differ. 2015;22(3):509–516.

151. Piskunova TS, Yurova MN, Ovsyannikov AI, et al. Deficiency in poly(ADP-ribose) polymerase-1 (PARP-1) accelerates aging and spontaneous carcinogenesis in mice. Curr Gerontol Geriatr Res. 2008;754190.

152. Prado CM, Wells JC, Smith SR, Stephan BC, Siervo M. Sarcopenic obesity: A Critical appraisal of the current evidence. Clin Nutr. 2012;31(5):583–601.

153. Purushotham A, Schug TT, Xu Q, Surapureddi S, Guo X, Li X. Hepatocyte-specific deletion of SIRT1 alters fatty acid metabolism and results in hepatic steatosis and inflammation. Cell Metab. 2009;9(4):327–338.

154. Pyo JO, Yoo SM, Ahn HH, et al. Overexpression of Atg5 in mice activates autophagy and extends lifespan. Nat Commun. 2013;4:2300.

155. Qiang L, Wang H, Farmer SR. Adiponectin secretion is regulated by SIRT1 and the endoplasmic reticulum oxidoreductase Ero1-L alpha. Mol Cell Biol. 2007;27(13):4698–4707.

156. Qiang L, Wang L, Kon N, et al. Brown remodeling of white adipose tissue by SirT1-dependent deacetylation of Ppargamma. Cell. 2012;150(3):620–632.

157. Qiao L, Shao J. SIRT1 regulates adiponectin gene expression through Foxo1-C/enhancer-binding protein alpha transcriptional complex. J Biol Chem. 2006;281(52):39915–39924.

158. Ramis MR, Esteban S, Miralles A, Tan DX, Reiter RJ. Caloric restriction, resveratrol and melatonin: Role of SIRT1 and implications for aging and related-diseases. Mech Ageing Dev. 2015;146-148:28–41.

159. Ravikumar B, Sarkar S, Davies JE, et al. Regulation of mammalian autophagy in physiology and pathophysiology. Physiol Rev. 2010;90(4):1383–1435.

160. Ravussin E, Redman LM, Rochon J, et al. A 2-year randomized controlled trial of human caloric restriction: feasibility and effects on predictors of health span and longevity. J Gerontol A Biol Sci Med Sci. 2015;70(9):1097–1104.

161. Reiter RJ, Tan DX, Rosales-Corral S, Manchester LC. The universal nature, unequal distribution and antioxidant functions of melatonin and its derivatives. Mini Rev Med Chem. 2013;13(3):373–384.

162. Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P. Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature. 2005;434(7029):113–118.

163. Romashkan SV, Das SK, Villareal DT, et al. Safety of two-year caloric restriction in non-obese healthy individuals. Oncotarget 2016.

164. Rongvaux A, Andris F, Van Gool F, Leo O. Reconstructing eukaryotic NAD metabolism. Bioessays. 2003;25(7):683–690.

165. Roth GS, Ingram DK, Lane MA. Calorie restriction in primates: will it work and how will we know? J Am Geriatr Soc. 1999;47(7):896–903.

166. Sakurai T, Kitadate K, Nishioka H, et al. Oligomerised lychee fruit-derived polyphenol attenuates cognitive impairment in senescence-accelerated mice and endoplasmic reticulum stress in neuronal cells. Br J Nutr. 2013;110(9):1549–1558.

167. Sale S, Verschoyle RD, Boocock D, et al. Pharmacokinetics in mice and growth-inhibitory properties of the putative cancer chemopreventive agent resveratrol and the synthetic analogue trans 3,4,5,4’-tetramethoxystilbene. Br J Cancer. 2004;90(3):736–744.

168. Salek RM, Maguire ML, Bentley E, et al. A metabolomic comparison of urinary changes in type 2 diabetes in mouse, rat, and human. Physiol Genomics. 2007;29(2):99–108.

169. Santana A, Santamarina A, Souza G, et al. Decaffeinated green tea extract rich in epigallocatechin-3-gallate improves insulin resistance and metabolic profiles in normolipidic diet--but not high-fat diet-fed mice. J Nutr Biochem. 2015;26(9):893–902.

170. Sauve AA, Youn DY. Sirtuins: NAD(+)-dependent deacetylase mechanism and regulation. Curr Opin Chem Biol. 2012;16(5-6):535–543.

171. Schmeisser K, Mansfeld J, Kuhlow D, et al. Role of sirtuins in lifespan regulation is linked to methylation of nicotinamide. Nat Chem Biol. 2013;9(11):693–700.

172. Schroeder S, Pendl T, Zimmermann A, et al. Acetyl-coenzyme A: a metabolic master regulator of autophagy and longevity. Autophagy. 2014;10(7):1335–1337.

173. Senf SM, Sandesara PB, Reed SA, Judge AR. p300 Acetyltransferase activity differentially regulates the localization and activity of the FOXO homologues in skeletal muscle. Am J Physiol Cell Physiol. 2011;300(6):C1490–C1501.

174. Shi L, Tu BP. Acetyl-CoA and the regulation of metabolism: mechanisms and consequences. Curr Opin Cell Biol. 2015;33:125–131.

175. Shimoyama Y, Suzuki K, Hamajima N, Niwa T. Sirtuin 1 gene polymorphisms are associated with body fat and blood pressure in Japanese. Transl Res. 2011;157(6):339–347.

176. Shokolenko IN, Wilson GL, Alexeyev MF. Aging: A mitochondrial DNA perspective, critical analysis and an update. World J Exp Med. 2014;4(4):46–57.

177. Slominski A, Pisarchik A, Semak I, et al. Serotoninergic and melatoninergic systems are fully expressed in human skin. FASEB J. 2002;16(8):896–898.

178. Smith JJ, Kenney RD, Gagne DJ, et al. Small molecule activators of SIRT1 replicate signaling pathways triggered by calorie restriction in vivo. BMC Syst Biol. 2009;3:31.

179. Smoliga JM, Baur JA, Hausenblas HA. Resveratrol and health--a comprehensive review of human clinical trials. Mol Nutr Food Res. 2011;55(8):1129–1141.

180. Soda K, Dobashi Y, Kano Y, Tsujinaka S, Konishi F. Polyamine-rich food decreases age-associated pathology and mortality in aged mice. Exp Gerontol. 2009;44(11):727–732.

181. Sohal RS, Weindruch R. Oxidative stress, caloric restriction, and aging. Science. 1996;273(5271):59–63.

182. Someya S, Yu W, Hallows WC, et al. Sirt3 mediates reduction of oxidative damage and prevention of age-related hearing loss under caloric restriction. Cell. 2010;143(5):802–812.

183. Song YS, Lee SK, Jang YJ, et al. Association between low SIRT1 expression in visceral and subcutaneous adipose tissues and metabolic abnormalities in women with obesity and type 2 diabetes. Diabetes Res Clin Pract. 2013;101(3):341–348.

184. Song J, Ke SF, Zhou CC, et al. Nicotinamide phosphoribosyltransferase is required for the calorie restriction-mediated improvements in oxidative stress, mitochondrial biogenesis, and metabolic adaptation. J Gerontol A Biol Sci Med Sci. 2014;69(1):44–57.

185. Stewart TM, Bhapkar M, Das S, et al. Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy Phase 2 (CALERIE Phase 2) screening and recruitment: methods and results. Contemp Clin Trials. 2013;34(1):10–20.

186. Sun C, Zhang F, Ge X, et al. SIRT1 improves insulin sensitivity under insulin-resistant conditions by repressing PTP1B. Cell Metab. 2007;6(4):307–319.

187. Swerdlow RH, Bothwell R, Hutfles L, Burns JM, Reed GA. Tolerability and pharmacokinetics of oxaloacetate 100 mg capsules in Alzheimer’s subjects. BBA Clin. 2016;5:120–123.

188. Tatar M, Bartke A, Antebi A. The endocrine regulation of aging by insulin-like signals. Science. 2003;299(5611):1346–1351.

189. Tchkonia T, Thomou T, Zhu Y, et al. Mechanisms and metabolic implications of regional differences among fat depots. Cell Metab. 2013;17(5):644–656.

190. Tenenbaum, D. 2014. “Monkey caloric restriction study shows big benefit; contradicts earlier study “ Retrieved 20 April 2016, 2016, from <http://news.wisc.edu/monkey-caloric-restriction-study-shows-big-benefit-contradicts-earlier-study/>.

191. Tsang AW, Escalante-Semerena JC. CobB, a new member of the SIR2 family of eucaryotic regulatory proteins, is required to compensate for the lack of nicotinate mononucleotide:5,6-dimethylbenzimidazole phosphoribosyltransferase activity in cobT mutants during cobalamin biosynthesis in Salmonella typhimurium LT2. J Biol Chem. 1998;273(48):31788–31794.

192. Turner RS, Thomas RG, Craft S, et al. A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology. 2015;85(16):1383–1391.

193. Venigalla M, Sonego S, Gyengesi E, Sharman MJ, Munch G. Novel promising therapeutics against chronic neuroinflammation and neurodegeneration in Alzheimer’s disease. Neurochem Int. 2016;95:63–74.

194. Walford RL, Harris SB, Gunion MW. The calorically restricted low-fat nutrient-dense diet in Biosphere 2 significantly lowers blood glucose, total leukocyte count, cholesterol, and blood pressure in humans. Proc Natl Acad Sci USA. 1992;89(23):11533–11537.

195. Waller KL, Mortensen EL, Avlund K, et al. Melatonin and cortisol profiles in late midlife and their association with age-related changes in cognition. Nat Sci Sleep. 2016;8:47–53.

196. Wang Y. Molecular Links between Caloric Restriction and Sir2/SIRT1 Activation. Diabetes Metab J. 2014;38(5):321–329.

197. Wang J, Vanegas SM, Du X, et al. Caloric restriction favorably impacts metabolic and immune/inflammatory profiles in obese mice but curcumin/piperine consumption adds no further benefit. Nutr Metab (Lond). 2013;10(1):29.

198. Wang P, Xu TY, Guan YF, et al. Nicotinamide phosphoribosyltransferase protects against ischemic stroke through SIRT1-dependent adenosine monophosphate-activated kinase pathway. Ann Neurol. 2011;69(2):360–374.

199. Wang TA, Zhang XD, Guo XY, Xian SL, Lu YF. 3-Bromopyruvate and sodium citrate target glycolysis, suppress survivin, and induce mitochondrial-mediated apoptosis in gastric cancer cells and inhibit gastric orthotopic transplantation tumor growth. Oncol Rep. 2016;35(3):1287–1296.

200. Wang Y, Xu C, Liang Y, Vanhoutte PM. SIRT1 in metabolic syndrome: where to target matters. Pharmacol Ther. 2012;136(3):305–318.

201. Weindruch R. The retardation of aging by caloric restriction: studies in rodents and primates. Toxicol Pathol. 1996;24(6):742–745.

202. Weindruch R, Sohal RS. Seminars in medicine of the Beth Israel Deaconess Medical Center Caloric intake and aging. N Engl J Med. 1997;337(14):986–994.

203. Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross JR, Thompson CB. ATP-citrate lyase links cellular metabolism to histone acetylation. Science. 2009;324(5930):1076–1080.

204. Wenzel E, Somoza V. Metabolism and bioavailability of trans-resveratrol. Mol Nutr Food Res. 2005;49(5):472–481.

205. White AT, McCurdy CE, Philp A, Hamilton DL, Johnson CD, Schenk S. Skeletal muscle-specific overexpression of SIRT1 does not enhance whole-body energy expenditure or insulin sensitivity in young mice. Diabetologia. 2013;56(7):1629–1637.

206. Wilkins HM, Harris JL, Carl SM, et al. Oxaloacetate activates brain mitochondrial biogenesis, enhances the insulin pathway, reduces inflammation and stimulates neurogenesis. Hum Mol Genet. 2014;23(24):6528–6541.

207. Wilkins HM, Koppel S, Carl SM, et al. Oxaloacetate enhances neuronal cell bioenergetic fluxes and infrastructure. J Neurochem. 2016;137(1):76–87.

208. Williams DS, Cash A, Hamadani L, Diemer T. Oxaloacetate supplementation increases lifespan in Caenorhabditis elegans through an AMPK/FOXO-dependent pathway. Aging Cell. 2009;8(6):765–768.

209. Witte AV, Fobker M, Gellner R, Knecht S, Floel A. Caloric restriction improves memory in elderly humans. Proc Natl Acad Sci USA. 2009;106(4):1255–1260.

210. Witte AV, Kerti L, Margulies DS, Floel A. Effects of resveratrol on memory performance, hippocampal functional connectivity, and glucose metabolism in healthy older adults. J Neurosci. 2014;34(23):7862–7870.

211. Xu C, Bai B, Fan P, et al. Selective overexpression of human SIRT1 in adipose tissue enhances energy homeostasis and prevents the deterioration of insulin sensitivity with ageing in mice. Am J Transl Res. 2013;5(4):412–426.

212. Xu C, Cai Y, Fan P, et al. Calorie restriction prevents metabolic aging caused by abnormal SIRT1 function in adipose tissues. Diabetes. 2015;64(5):1576–1590.

213. Yamada M, Moriguch Y, Mitani T, Aoyama T, Arai H. Age-dependent changes in skeletal muscle mass and visceral fat area in Japanese adults from 40 to 79 years-of-age. Geriatr Gerontol Int. 2014;14(Suppl 1):8–14.

214. Yang H, Yang T, Baur JA, et al. Nutrient-sensitive mitochondrial NAD+levels dictate cell survival. Cell. 2007;130(6):1095–1107.

215. Ye J, Keller JN. Regulation of energy metabolism by inflammation: a feedback response in obesity and calorie restriction. Aging (Albany NY). 2010;2(6):361–368.

216. Yoshino J, Mills KF, Yoon MJ, Imai S. Nicotinamide mononucleotide, a key NAD(+) intermediate, treats the pathophysiology of diet- and age-induced diabetes in mice. Cell Metab. 2011;14(4):528–536.

217. Yoshizaki T, Milne JC, Imamura T, et al. SIRT1 exerts anti-inflammatory effects and improves insulin sensitivity in adipocytes. Mol Cell Biol. 2009;29(5):1363–1374.

218. Yuan Y, Cruzat VF, Newsholme P, Cheng J, Chen Y, Lu Y. Regulation of SIRT1 in aging: Roles in mitochondrial function and biogenesis. Mech Ageing Dev. 2016;155:10–21.

219. Zhang J, Feng X, Wu J, et al. Neuroprotective effects of resveratrol on damages of mouse cortical neurons induced by beta-amyloid through activation of SIRT1/Akt1 pathway. Biofactors. 2014;40(2):258–267.

220. Zhu H, Guo Q, Mattson MP. Dietary restriction protects hippocampal neurons against the death-promoting action of a presenilin-1 mutation. Brain Res. 1999;842(1):224–229.

..................Content has been hidden....................

You can't read the all page of ebook, please click here login for view all page.
Reset
3.238.79.169