Hostname: page-component-84b7d79bbc-5lx2p Total loading time: 0 Render date: 2024-08-04T10:46:40.662Z Has data issue: false hasContentIssue false

Toxicology of pharmaceutical and nutritional longevity compounds

Published online by Cambridge University Press:  22 June 2023

Cigdem Kahraman
Affiliation:
Department of Pharmacognosy, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
Duygu Kaya Bilecenoglu
Affiliation:
Faculty of Health Sciences, Aydın Adnan Menderes University, Aydın, Turkey
Suna Sabuncuoglu
Affiliation:
Department of Pharmaceutical Toxicology, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
Irem Tatli Cankaya*
Affiliation:
Department of Pharmaceutical Botany, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
*
Corresponding author: Irem Tatli Cankaya; Email: itatli@hacettepe.edu.tr; iremcankaya@gmail.com
Rights & Permissions [Opens in a new window]

Abstract

Aging is the most prominent risk factor for many diseases, which is considered to be a complicated biological process. The rate of aging depends on the effectiveness of important mechanisms such as the protection of DNA from free radicals, which protects the structural and functional integrity of cells and tissues. In any organism, not all organs may age at the same rate. Slowing down primary aging and reaching maximum lifespan is the most basic necessity. In this process, it may be possible to slow down or stabilise some diseases by using the compounds for both dietary and pharmacological purposes. Natural compounds with antioxidant and anti-inflammatory effects, mostly plant-based nutraceuticals, are preferred in the treatment of age-related chronic diseases and can also be used for other diseases. An increasing number of long-term studies on synthetic and natural compounds aim to elucidate preclinically and clinically the mechanisms underlying being healthy and prolongation of life. To delay age-related diseases and prolong the lifespan, it is necessary to take these compounds with diet or pharmaceuticals, along with detailed toxicological results. In this review, the most promising and utilised compounds will be highlighted and it will be discussed whether they have toxic effects in short/long-term use, although they are thought to be used safely.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press

Introduction

Aging can be defined as the accumulation of damage over time, a loss in functional ability, adaptation difficulties to the environment and an increase in illness and mortality. The aging of the global population is posing significant issues for both industrialised and developing countries due to changes in the population pyramid (Refs Reference Proshkina1, Reference Mitnitski2). The percentage of persons over the age of 60 is increasing globally, and it is predicted that by 2050, it would nearly double, rising from 12 per cent in 2015 to 22 per cent (Refs Reference Mc Auley3Reference Yang5). Despite the continued predicted increase in the aging population, the geriatric population's life expectancy and survival are improving significantly (Ref. Reference Chen4).

According to several studies, aging is one of the most important risk factors for chronic diseases including cancer, atherosclerosis, cardiovascular disease, stroke, diabetes, renal failure, chronic lung disease, osteoporosis, rheumatoid arthritis, blindness, dementia and neurological pathologies that cause the greatest global morbidity, mortality and use of medical resources. Additionally, aging increases a person's risk for the geriatric syndrome, a reduction in immunity and slower physical recovery (Refs Reference Yang5Reference Li8).

Due to its significant role as a risk factor for morbidity and age-related diseases, the biology of aging has considerable interest among researchers.

Many hypotheses have been developed to expand our comprehension of the aging process and to help us develop methods for extending life. Based on the level at which the aging mechanism is targeted, the theories of aging are divided into categories including evolutionary, systemic, molecular and cellular theories (Ref. Reference Jayanthi, Joshua and Ranganathan9).

In aging research, several natural products and dietary supplements continue to be studied extensively for their antiaging properties or are used as research materials to discover molecular mechanisms of biological aging (Refs Reference Newman and Cragg10, Reference Gómez-Linton11). Using natural resources may be an effective way to delay physiological and pathological aging and, in turn, avoid the development of these disorders, according to recent research (Refs Reference Herranz and Gil12Reference Mechchate14). Natural compounds that are separated or obtained from plants are distinguished by the synthesis of secondary metabolites and are abundant in nature. Numerous chemical families, primarily flavonoids, phenolic acids and terpenoids, are included in secondary metabolites. Other compounds that are part of the primary metabolites have also demonstrated important anti-aging benefits (Refs Reference Herranz and Gil12Reference Mechchate14) (Table 1)

Table 1. Examples of anti-aging compounds from natural sourcesa (Refs Reference Ding15, Reference Fan16)

a Compounds in bold and italic font have been evaluated for their safety and toxicological properties in this review.

Natural products encompass a diverse group of substances made from a variety of sources, including marine organisms, bacteria, fungi and plants. Before the market, the safety of natural products is sometimes under- or unstudied, which raises the possibility of unpleasant responses with the natural products alone as well as with accompanying medications and other natural products. For anticipating and reducing natural product-drug interactions and adverse events, standardised procedures and in vitro testing have not been developed and specialised clinical research may be prohibitively expensive. In addition, some natural products contain ingredients or additives that are unidentified and uncharacterised, which poses a serious risk to users (Ref. Reference Gaston17).

These compounds and their active components work include tissue formation, telomere activation, anti-senescence action, DNA repair and targeted antioxidant activity. In this review, the evidence supporting the anti-aging properties of some of the most promising natural substances in this area will be discussed and evaluated by summarising pharmacological and toxicological properties.

Molecular mechanisms of aging

Significant influences on aging can be found in genetics, the environment and intrinsic variables. Common age-related functional loss and an increase in multimorbidity with age are caused by damage accumulation (Ref. Reference Niedernhofer18). Human aging is a progressive time-related process. It varies among individuals and, biologically, relates to a loss of homoeostasis, an increase in the organism's sensitivity and susceptibility to disease and death, as well as the gradual aging-related deterioration of cells, tissues and organs. Senescence refers to these features of aging and is what causes a person's health to deteriorate. Additionally, they contribute to age-related disease including coronary heart disease and physiological changes like diminished renal function in aging (Refs Reference Ferrucci19, Reference Wu20).

As a result of a genetic programme, the loss of homoeostasis is eventually caused by a combination of factors. Changes in mitochondria, the accumulation of lipofuscins that cause cellular aging, changes in the cellular structure or macromolecular aggregation at the molecular level, telomere loss and shortening at the chromosomal level and amyloid deposition are all monitored parts of the aging process. As a consequence of genetic changes, the phenotype, functionality and behaviour of an organism can be affected (Ref. Reference Mechchate14).

Researchers claim that aging is a physiological deterioration caused by damage to essential molecules in aging populations (Refs Reference Rozhok and DeGregori21, Reference Piotrowski22). The aging process differs greatly from aging in a single organism, tissue or cell. While studying the various genes involved in senescence, which can be categorised into three groups: genes regulating somatic maintenance and repair, genes promoting early survival and genes causing late deleterious mutations, other academics have concentrated on understanding the evolutionary foundation of senescence in the aging mechanism. Due to their increased development and repeatability, these critical genes have been found to affect a species' evolution and survival, and they may also contribute to shortened lifespans. Cellular senescence may have a pleiotropic effect that prevents cancer while also speeding up the aging process of the body (Refs Reference Kirkwood23, Reference Dodig, Čepelak and Pavić24). It is known that aging is associated with hallmarks of aging: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion and altered intercellular communication (Ref. Reference López-Otín25).

The accumulation of genetic damage, which may interfere with cell homoeostasis and lead to genomic instability, is one widely accepted factor in aging. Copy number mutations, somatic mutations and chromosomal aneuploidy all contribute to escalating DNA damage. Age-related abnormalities in the DNA repair process influence the expression of vital genes and transcriptional pathways, which in turn causes cell malfunction (Refs Reference Li8, Reference Tiwari and Wilson26). Numerous aging models have implications for the various processes that can cause transcriptional alterations related to aging. Since mutations in genes like daf-2 or daf-16, which encode the IGF-1 receptor and FOXO, respectively, can lengthen worm lifetime, aging can be controlled (Caenorhabditis elegans). Additionally, there are clear links between lifespan and genetic loci such as APOE, FOXO3, 5q33.3 and ACE (Ref. Reference Stegeman and Weake27). Premature aging disorders have been linked to impaired DNA repair capacity, according to different preclinical investigations (Refs Reference Li8, Reference Tiwari and Wilson26). DNA helicases are crucial components of the response to DNA damage. Since several human RecQ helicases are deficient in conditions linked to cancer and premature aging, the RecQ family of DNA helicases is of great interest (Ref. Reference Brosh and Bohr28).

Telomere length is linked to age-related diseases and DNA damage, and telomere shortening can affect somatic stem cell activity. Human somatic cells have low levels of telomerase expression, which causes telomere shortening and replicative senescence. The typical length of the terminal restriction fragment of chromosomes diminishes with age in fibroblasts and peripheral blood cells (Refs Reference Smith, Pendlebury and Nandakumar29, Reference Katayama30). These results demonstrate that the most important factor of cellular aging is telomere length, but not telomerase activity. Furthermore, rather than the average length of telomeres, the shortest telomere affects cell viability and chromosome stability. In this regard, numerous investigations have shown that telomere lengthens with age in both humans and animals (Refs Reference Smith, Pendlebury and Nandakumar29, Reference Shu31). Telomere length affects mortality risk and replicative senescence, which stops cancer cells from proliferating forever. Telomere shortening continues to be a characteristic and a counting mechanism of senescent cells (Refs Reference Proshkina1, Reference Mechchate14).

Another potential factor in aging is the discovery that the bulk of age-related changes is brought on by free radical atoms or molecules that have an unpaired and reactive electron. These oxygen-derived species are secondary messengers in signalling pathways involved in the regulation of various mechanisms, including changes in gene expression, cell replication, differentiation and apoptotic cell death (Refs Reference Sohal and Weindruch32, Reference Harman33). They can also react with macromolecules to produce free radicals from the molecules that were attacked. Maximal lifespan is impacted by the production of these free radicals in human organs such as the heart, kidney and liver. In this situation, nutritional antioxidants have been shown to lower the risk of cancer, heart disease and dementia. For instance, higher dietary intake of vitamin C, carotenoids and α-tocopherol was associated with reduced risk of cardiovascular disease, cancer and all-cause mortality. Reactive oxygen species (ROS) itself, which are one of the developmental-genetic features of aging, contribute to the somatic accumulation of mutations in mitochondrial DNA. These mutations cause aging and cell death, as well as a gradual reduction of bioenergetic capacity (Refs Reference Mechchate14, Reference Barja34, Reference Liu35).

Epigenetic alterations play also an important role in the aging process. DNA methylation, histone changes and chromatin remodelling are chromatin modifications that are connected to cell aging. Through modifications to the chromatin structure and DNA accessibility to transcription factors, epigenetic processes control gene expression. On the one hand, the epigenetic alterations linked to senescence function as compensatory mechanisms that let cells resist growing stress and avoid irreparable DNA damage. On the other hand, the modifications result in side processes that hurt cells and the body as a whole, speeding up aging and the emergence of age-related disorders (Refs Reference Proshkina1, Reference Li8).

A genetically designed continuum of growth and maturation is thought to be the primary driver of the aging process. The maximum lifespan is very species-specific, as humans have a maximum lifespan 30 times longer than mice. Natural resources present a great potential for searching for longevity features. In this regard, it has been shown that the complete lifespan potential is determined by a set of particular genes that are present in a wide variety of species. These genes cause the synthesis of substances that are involved in the regulation of the species' life through a variety of methods, including the modulation of stress and resistance, the increase in metabolic capacity and the silencing of genes that promote aging (Refs Reference Mechchate14, Reference Singh36).

The disturbance of protein homoeostasis is also linked to the onset of aging and most aging-related illnesses. Almost all tissues of older organisms, particularly those with a low rate of proliferation, include modified and misfolded proteins, as well as protein, aggregates Aging symptoms include protein aggregation, post-translational modification and altered protein turnover. Proteostasis is distorted by nonenzymatic posttranslational changes that accumulate with aging in all cell compartments. The process of nonenzymatic glycosylation (glycation), which results in the production of advanced glycation end products, hazardous intermediate products, and crosslinks between protein molecules, is best understood in the context of aging (Refs Reference Proshkina1, Reference Mechchate14).

Recent investigations have shown that bioactive compounds from different medicinal and nutritional species exhibit remarkable anti-aging effects. According to a comprehensive review by Ding et al., 185 natural compounds and 55 complex/extracts among the approximate 300 longevity compounds were revealed. The selected popular anti-aging natural and pharmaceutical products have been evaluated below regarding their safety and toxicological properties. These molecules showed antiaging properties in at least two aging models (Ref. Reference Ding15). Model organisms such as the nematode Caenorhabditis elegans, the fruit fly Drosophila melanogaster, the mouse Mus musculus, and the yeast Saccharomyces cerevisiae were mostly used for the explanation of the longevity properties of candidates (Ref. Reference Gómez-Linton11). Table 2 shows the average percentage of the mean life-prolonging effects of the compounds of interest in different models.

Table 2. The longevity effects of the selected compounds

Safety and toxicological properties of selected longevity compounds

Caffeine

Caffeine (Fig. 1), a methylxanthine derivative (13,7-trimethylxanthine) is a secondary metabolite classified as a purine alkaloid and synthesised majorly in various plants such as Coffea arabica L. (Arabica coffee), Camellia sinensis (L.) Kuntze (tea), Paullinia cupana Kunth (Guarana) and Cola sp. (Ref. Reference Ashihara and Suzuki54). Psychomotor stimulant properties of caffeine are primarily associated with its reducing properties on adenosine transmission via the blockade of adenosine A2A in the brain (Ref. Reference Fisone, Borgkvist and Usiello55). Other mechanisms of action proposed for caffeine, inhibition of phosphodiesterase and mobilisation of intracellular calcium, require high concentrations of caffeine than likely to be taken in daily consumption (Ref. Reference Cappelletti56).

Figure 1. The chemical structure of caffeine.

Today, caffeine is used as an adjuvant in some analgesics or as a major constituent in some energy beverages (Refs Reference Derry, Derry and Moore57, Reference Alsunni58). The FDA and EFSA reported that habitual consumption of 400 mg of caffeine (approximately 4 or 5 cups of coffee and about 5.7 mg/kg body weight (BW) per day for a 70-kg adult) throughout the day can be considered safe except for pregnant women, however, it should be considered that may differ according to individual sensitivity, lactating, medication use, etc. (Refs 59, 60). Single doses of caffeine up to 200 mg (about 3 mg/kg BW for a 70-kg adult) in healthy adults (or when consumed less than two hours before an intense physical exercise) do not give rise to health concerns. Habitual consumption and a single dose of caffeine for adults may also consider safe for children and adolescents because of similar caffeine clearance (Ref. 60).

It has been suggested that age-related neurodegenerative diseases are reduced by chronic, moderate caffeine consumption as well as habitual consumption was correlated with reduced mortality and positively affected healthspan features. In addition, its lifespan extending effect on the worm model was reported (Ref. Reference Sutphin39). However, National Health and Nutrition Examination Survey on 5826 adults from two genders, investigating the relationships between caffeine and coffee intakes, and telomere length, presented that caffeine consumption caused a tendency to shorten telomeres in US adults. However, telomere lengths were positively affected by coffee consumption (Ref. Reference Tucker61).

After intake of therapeutic amounts of caffeine, hypertension can be noted, however, hypotension is frequently noted in the case of overdoses. Acute toxicologic symptoms occur with 15 mg/l and higher plasma concentrations of caffeine, while 50 mg/l is considered toxic. Generally reaching ≥ 80 mg/l of plasma concentrations, symptoms indicating the lethal phase can be observed. The clinical symptoms of caffeine intoxication from mild to lethal level have included nausea, vomiting, agitation, anxiety, seizures, delusions, hypokalemia, rhabdomyolysis, hypertension, hypotension, tachycardia, bradycardia, myocardial infarction, cardiac arrest, respiratory and renal failure and death. Daily intakes of 1 to 1.5 g of caffeine can result in chronic toxicity of which symptoms are similar to chronic anxiety (Ref. Reference Willson62). Besides 2 g of caffeine ingestion requiring hospitalisation, ≥5 g of caffeine ingestion is estimated to be an overdose in adults (Ref. Reference Banerjee63).

Maximum plasma concentration of caffeine is reached in 15–120 min after ingestion. Caffeine is mainly metabolised to its pharmacologically active but less toxic metabolite paraxanthine (1,7-dimethylxanthine) by cytochrome P450 1A2 (CYP1A2) in the liver. Caffeine is also metabolised to 1-demethylated product theobromine and 7-demethylated product theophylline, which are also pharmacologically active, by CYP1A2 (Ref. Reference Willson62). Thus, possible metabolic interaction with CYP1A2 may change the metabolism of caffeine. For example, the potent inhibitory effect of the selective serotonin reuptake inhibitor fluvoxamine on CYP1A2 reduces the clearance of caffeine during concomitant intake which may lead to caffeine intoxication (Refs Reference Rasmussen, Nielsen and Brøsen64, Reference Jeppesen65). Yet another example, antibacterial quinolone derivatives, enoxacin, pipemidic acid, ciprofloxacin and norfloxacin, competitive and dose-dependent inhibitors of CYP1A2 enzyme, have been reported for their AUC-enhancing and caffeine clearance-reducing effects (Ref. Reference Carrillo and Benitez66). 5-methoxypsoralen (bergapten) and 8-methoxypsoralen (methoxsalen), which are naturally occurring linear furanocoumarins and used to treat psoriasis, inhibit the metabolism of caffeine by CYP1A2 mediated-inhibition (Ref. Reference Carrillo and Benitez66).

Metformin

Metformin (MET, Fig. 2) is a biguanide used as a first-line treatment for type-2 diabetes mellitus (Ref. Reference Gong67). The discovery of MET (dimethylbiguanide) is linked to a plant Galega officinalis L., which was traditionally used in Europe to treat the symptoms of diabetes and was later found to be rich in guanidine with blood sugar-lowering effects. The synthesised guanidine derivative MET was introduced as an antidiabetic agent in the 1990s in the USA (Ref. Reference Aroda and Ratner68). It is a potent antihyperglycemic agent, which counters insulin resistance, reduces hepatic gluconeogenesis and increases glucose uptake, without causing weight gain and overt hypoglycemia (Ref. Reference Wiernsperger and Bailey69). The drug also has beneficial features for diabetes-related polycystic ovary disease, fatty liver disease and cardiovascular complications, alongside being suggested as an adjuvant treatment for cancer or gestational diabetes in the pre-diabetic population (Ref. Reference Viollet70).

Figure 2. The chemical structure of metformin.

The enhancing effects of MET on insulin sensitivity are assigned to its stimulation of the tyrosine kinase activity of the β subunit of the insulin receptor (Ref. Reference Wiernsperger and Bailey69). At the molecular level, MET blocks the mitochondrial respiratory chain, which leads to the activation of AMPK resulting in suppression the production of gluconeogenic enzymes (Ref. Reference Rena, Hardie and Pearson71). MET's potential as an anti-aging candidate was shown in several studies. In the context of telomere length, telomere shortening was reduced by metformin in the mild age-related diabetes (MARD) group. The telomere length of MARD group was significantly shorter than the NON-MARD group, however, was disappeared after MET (Ref. Reference Hu72).

MET circulates in the plasma unbound and is excreted unchanged in the urine. The bioavailability of the MET formulations is about 50–60% (Ref. Reference DeFronzo73). Pharmacokinetic distribution of MET is mediated by organic cation transporters (OCT1, OCT2 and OCT3), multidrug and toxin extruders (MATE1 and MATE2-K) and plasma membrane monoamine transporter (PMAT). Oral absorption, hepatic and renal uptake are mediated by OCTs. Renal excretion is mediated by OTC2 and MATEs (Refs Reference Gong67, Reference Pakkir Maideen, Jumale and Balasubramaniam74, Reference Stage, Brøsen and Christensen75). As MET is not metabolised and excreted unchanged, drug-drug interaction is not expected for MET. However, inhibition of MET transporters, which could be resulting in increased plasma concentration, is associated with drug interactions. (Refs Reference Pakkir Maideen, Jumale and Balasubramaniam74, Reference Stage, Brøsen and Christensen75). Increased plasma concentration and AUC of MET were observed when co-administration with the proton pump inhibitors, lansoprazole, pantoprazole and rabeprazole, as well as a decrease in renal excretion by 13%, and prolonged elimination half-life of MET from 3.9 to 4.5 h accompanied by lansoprazole. Thus, MET and lansoprazole together were suggested as a risk for those with lactic acidosis (Ref. Reference Adak76).

The most frequent side effects of MET are gastrointestinal defects. MET-associated lactic acidosis, suggests being contraindicated in those who have substantial renal dysfunction, and anaemia due to vitamin B12 malabsorption and deficiency has been rarely reported (Ref. Reference Adak76). The overdose administration of this drug results specifically in metabolic acidosis and hyperlactatemia, which are indicated to be the markers of metformin toxicity. Metformin attracts attention with a secondary effect on weight loss, for this reason causing long-term drug abuse. A report serves metformin intoxication symptoms such as cardiogenic shock and hypotension, cardiac dysrhythmia and ultimately cardiac arrest (Ref. Reference Hajsadeghi77). Another study on patients with type 2 diabetes concludes that vitamin B12 deficiency is a potential side effect in long-term and high-dose metformin therapy (Ref. Reference Infante78). Studies categorised metformin overdose toxicity into two groups: MALA (metformin-associated lactic acidosis) and MILA (metformin-induced lactic acidosis). Clinical reports also remark on hypoglycemia accompanying metabolic acidosis but the toxic dose has not been determined yet (Ref. Reference Wang and Hoyte79).

Fucoxanthin

Fucoxanthin (Fig. 3) is a naturally occurring marine carotenoid, which was found in algae such as Undaria pinnatifida, Laminaria japonica, Phaeodactylum tricornutum and Cylindrotheca closterium, and has a unique structure. The molecular structure, which was composed of a conjugated carbonyl group with an unusual allenic bond, a 5,6-monoepoxide and some oxygenic functional groups, is unstable and easily degradable by oxygen, heat, light, etc (Ref. Reference Zhang80). Trans-fucoxanthin, which is responsible for its biological activities, is shown to produce its two minor cis-isomers according to storage conditions (Refs Reference Kawee-ai, Kuntiya and Kim81, Reference Mohibbullah82).

Figure 3. The chemical structure of fucoxanthin.

Fucoxanthin has been shown to have potential health benefits for lifestyle-related diseases by the evidence of hepatoprotective, anti-inflammatory, antioxidant, antidiabetic, antitumoural and anti-obesity activities (Ref. Reference Zhang80) along with anti-aging properties. Especially with its enhanced antioxidant capacity, fucoxanthin inhibits the ROS which increases in some diseases like ischaemic stroke and acute lung injury (Ref. Reference Mao83).

Fucoxanthin is hydrolysed into fucoxanthinol through deacetylation by digestive enzymes, lipase and cholesterol esterase in the gastrointestinal tract before absorption, and fucoxanthinol is later converted into amarouciaxanthin A by short-chain dehydrogenase/reductase in the liver (Ref. Reference Shikov84). The pharmacokinetics of fucoxanthin depends on the species (Refs Reference Kawee-ai, Kuntiya and Kim81, Reference Mohibbullah82). Fucoxanthinol did not show significant adverse effects at 200 mg/kg BW in C.B-17/Icr-SCID mice for 28 days (Ref. Reference Ishikawa85). Several studies on this bioactive compound containing extracts have been performed to determine the short- and long-term toxicity and repeated oral dose toxicity as well. No significant toxicity and abnormalities have been observed in rats and mice after 14, 30 and 90 days of administration (Ref. Reference Abu-Ghannam and Shannon86).

Orally administrated fucoxanthin at the doses of 500, 1000 and 2000 mg/kg did not cause any mutagenicity in mice. In addition, fucoxanthinol, the major metabolite of fucoxanthin after oral administration, was tested with the in vitro Ames test, in which no mutagenicity was detected under the experimental condition (Ref. Reference Beppu87)

Fucoxanthin can be accepted as a safe product. In a 14-day single-dose study (1000 and 2000 mg/kg) and a 30-day repeat-dose study (500 and 1000 mg/kg) in ICR mice, no abnormalities and mortalities were shown. However, increased total cholesterol concentrations were observed by plasma biochemical analyses in all fucoxanthin-treated groups (Ref. Reference Beppu88). In a double-blind placebo-controlled study on Japanese obese subjects, 1 or 3 mg daily fucoxanthin capsule for 4 weeks exhibited no side effects (Ref. Reference Hitoe and Shimoda89). The Panel on Dietetic Products, Nutrition and Allergies of the European Commission gave an opinion about the consumption of the thallus part of Undaria pinnatifida, which has to carry an amount equivalent to 15 mg fucoxanthin per day. However, this food source has not enough evidence on the maintenance or achievement of normal body weight as claimed by the manufacturer (Ref. Reference Bresson90). Although there is a lack of evidence on the safety of fucoxanthin consumption, the Food and Drug Administration announced fucoxanthin extracted from the alga, Phaeodactylum tricornutum, as a new dietary ingredient that can be consumed at a level of 3 mg/day with no time limit or 5 mg/day fucoxanthin for up to 90 days (Ref. Reference Bae91). It is obvious that to present a safety profile for the supplementation levels of fucoxanthin in humans, reported clinical trials remain limited.

Spermidine

Spermidine (Fig. 4) is a natural aliphatic polyamine and is found in all living cells of plants, animals and microorganisms, as well as can also be found in plant- and animal-originated foods (Ref. Reference Munoz-Esparza92). For several organisms, the proportion of polyamines like spermidine is due to either administration via dietary sources or synthesised by the gut microbiota (Ref. Reference Ramos-Molina93). Spermidine is found high in foods such as wheat germ, fermented soybeans (Natto), mushrooms, nuts and some fruits and vegetables. It is absorbed mainly in the duodenum and proximal jejunum parts of the small intestine (Ref. Reference Zou94). The nutritional intake of foods that contain arginine (precursor of polyamines) helps polyamine-producing bacteria produce spermin and spermidine in the microbiota of mammals (Ref. Reference Madeo95). The studies suggest that a polyamine rich diet including spermidine helps humans reach older ages healthy, recover especially after surgery and wound healing because of its antioxidant properties (Refs Reference Munoz-Esparza92, Reference Ramos-Molina93). It has antitumour, cardiovascular protective, neuromodulator, anti-obesity and anti-inflammatory features, and is an autophagy-inducing agent, as well (Refs Reference Munoz-Esparza92, Reference Zou94, Reference Madeo95). Besides, spermidine treatment induced a reduced percentage of the nuclei with short telomeres (Ref. Reference Wirth96).

Figure 4. The chemical structure of spermidine.

An in vivo study, 0.5, 5 or 50 g/kg mice BW daily intake over 28 days, and a 3-month randomised, placebo-controlled, double-blind Phase II (at the dose of 1.2 mg/day) study showed that spermidine rich wheat extract to be safe and well tolerable by mice and older human (Ref. Reference Schwarz97). Til et al. determined the acute oral toxicity of spermidine at 600 mg/kg BW in Wistar rats. A significant increase in plasma activities of alkaline phosphatase, alanine aminotransferase and aspartate aminotransferase at the dose of 10.000 ppm and decrease in plasma protein level in female rats; and a significant decrease in food intake and body weight along with plasma calcium and potassium levels in male rats were observed (Ref. Reference Til98). The homoeostasis of polyamines in the mammalian tissue is regulated by several functions including catabolic pathways. According to the studies, the catabolic reactions may result in some toxic products such as acrolein and other reactive aldehyde species, which are being catalysed by spermidine/spermine N(1)-acetyltransferase and serum polyamine oxidase (Ref. Reference Pegg99). Although spermidine is an abundant polyamine in mammalian cells and has lower toxicity than other polyamines like spermine, experimental studies show that these byproducts may be harmful to patients with cancer and chronic renal failure (Ref. Reference Madeo95). However, the toxic dose for humans as supplementary spermidine has not been determined yet (Refs Reference Madeo100, Reference Soda101).

Successive development of spermidine-related products as natural dietary supplements (SpermidineLIFE® Immunity+, SpermidineLIFE® Original 365+, SpermidineLIFE® Memory+) and launch on the market in 2019 suggests the safety of spermidine in food supplements (Refs Reference Zou102, 103)

Rapamycin

Rapamycin (RAPA, Fig. 5), also known as FK506 or sirolimus, a lipophilic macrolide lactone was initially isolated from Streptomyces hygroscopicus from a soil sample found on Easter Island (Rapa Nui). The antifungal activity of RAPA, particularly against Candida albicans, was first demonstrated, and immunosuppressive and anticancer properties were later proven (Refs Reference Abraham and Wiederrecht104, Reference Seto105). FDA approved rapamycin according to results of the 2 double-blind clinical trials at the fixed doses of 2 and 5 mg/day with co-administration of cyclosporin and corticosteroid prednisone, for use in kidney transplant (Refs 59, Reference Mahalati and Kahan106). It is now a prescription drug named Rapamune® for preventing tissue rejection after kidney transplantation for people ≥13 years old, but not recommended for liver and lung transplant patients (Ref. 107).

Figure 5. The chemical structure of rapamycin.

The immunosuppressive features have been shown to procure at the plasma concentration of 5 to 20 μg/l (Ref. Reference Mahalati and Kahan106), and 5–15 μg/l is recommended for patients at standard risk of rejection (Ref. Reference MacDonald108). Rapa is shown to be at the highest concentration in the red blood cells at the ratio of 94.5%, and the remaining drug is distributed among the plasma (3.1%), lymphocytes (1.01%) and granulocytes (1.0%) (Ref. Reference Mahalati and Kahan106).

Rapamycin acts as an immunosuppressant through the inhibition of the mammalian target of rapamycin (mTOR) by binding to its intracellular receptor FK506-binding protein 12 (FKBP12). mTOR is inhibited via the interaction of the FKBP12-rapamycin complex directly with the relevant domain of mTOR (Ref. Reference Yoo109). mTOR inhibition is also regarded to be important for preventing aging and age-related disease. However, a recent finding demonstrated that mTOR is upregulated acting as a survival pathway in mice with short telomeres, thus the inhibition of this pathway was found to be harmful to telomere-deficient mice (Ref. Reference Ferrara-Romeo110).

RAPA is metabolised by the cytochrome CYP3A4/P-glycoprotein system, which is mainly localised in the liver and intestine that may be responsible for the first pass effect resulting in low drug bioavailability (~14%) in oral administration. (Refs Reference MacDonald108, Reference Sádaba111). 4′-O-demethyl, 7-O-demethyl, several hydroxy, dihydroxy, hydroxydemethyl and didemethyl sirolimus, which do not serve an important contribution significantly to the activity of the parent drug, were characterised as metabolites of RAPA (Ref. Reference Leung112).

Strong CYP3A4/P-glycoprotein inhibitors clinically increase the RAPA concentration. Antifungal azoles; corticosteroids; calcineurin inhibitor cyclosporine; non-dihydropyridine calcium channel blocker diltiazem was shown to inhibit the CYP3A4 in the pharmacokinetic studies, leading the increased RAPA levels in blood (Ref. Reference Glotzbecker113). Cyclosporine is also a P-glycoprotein inhibitor, and in the case of concurrent treatment, RAPA is recommended to be taken 4 hours after cyclosporine administration. In addition, ketoconazole, voriconazole, itraconazole, erythromycin, telithromycin and clarithromycin are strong inhibitors of CYP3A4/P-glycoprotein as well as grapefruit Juice inhibits the CYP3A4-mediated metabolism of RAPA (Ref. 107).

Some antiaging studies about rapamycin on mice revealed unexpected side effects such as insulin resistance, testicular degeneration and ophthalmic problems (Refs Reference Fischer114Reference Wilkinson116). Clinical studies confirm the in vitro and in vivo findings about the adverse effects of rapamycin treatment on peripheral insulin insensitivity, by showing deleterious effects on pancreatic β-cells with the inhibition of mTORC1 and mTORC2 complexes (Ref. Reference Barlow, Nicholson and Herbert117). A cohort study on male C57BL/6J mice testing rapamycin on healthy aging resulted in some side effects on the kidney and reproductive tract compared to the control group. In addition to nephrotoxicity and gonadotoxicity, impaired glucose tolerance and hypercholesterolaemia were observed on rapamycin-treated animals after treatment with 14 mg/kg encapsulated rapamycin in food for 1 year (Ref. Reference Neff115). Ross et al. studied long-term rapamycin exposure on marmoset monkeys (Callithrix jacchus) to clarify the underlying mechanism of hyperglycaemia, also seen in rodent studies. In contrast with the other animal models, hyperglycaemia was not seen in healthy marmosets. It was concluded that one of the theories was the increase in hepatic gluconeogenesis (Ref. Reference Ross118). In a study on 24 middle-aged healthy dogs, non-immunosuppressive doses (0.05 or 0.1 mg/kg orally three times per week) of rapamycin for 10 weeks did not cause significant clinical side effects or abnormal haematological changes. However, corpuscular volume (the volume of the red blood cells) was significantly decreased in RAPA-treated groups (Ref. Reference Urfer119).

Serious allergic reactions such as swelling on the face, eyes or mouth, trouble in breathing, chest pain or tightness, rash or peeling of the skin, feeling dizzy or faint, chest pain or tightness; edema; poor wound healing, increased levels of cholesterol and triglycerides; increased protein in the urine; increased risk for viral infections, lung and breathing problems are indicated as the possible serious side effects of the drug Rapamune® (Ref. Reference Marti and Frey120). The nephrotoxicity in a clinical study appeared on not healthy people but patients with renal health problems. More detailed mechanism studies needed to be discussed to evaluate the side effects of rapamycin in the treatment dose in humans (Ref. Reference Nguyen and Gupta121).

α-Lipoic acid

α-Lipoic acid (α-LA, Fig. 6), also called thioctic acid (CAS RN 1077-28-7), is a caprylic acid-derived fatty acid found in several dietary sources such as red meats, heart, kidney, liver, wheat germ and to a lesser degree, fruits and vegetables. The compound has a dithiol functional group, which reacts with free radicals (Ref. Reference Shay122). The studies reveal that it provides detoxification and promotes some biochemical reactions, especially the renewal of damaged detoxification enzymes that helps to treat chronic diseases associated with oxidative stress. Both in vitro and in vivo studies have shown α-lipoic acid demonstrating very high radical scavenging activity, the main reason for which has been added as an ingredient in several dietary supplements such as multivitamins also used by intravenous injection. Especially its reduced metabolite dihydrolipoic acid (DHLA) showed quite high radical scavenging activity (Ref. Reference Xiong123). PGC-1α (peroxisome proliferator-activated receptor γ coactivator-1α) is a receptor, plays a role in protecting from age-related chronic diseases. Xiong et.al. 2005 indicates that α-LA, as a cofactor, upregulated PGC-1α-dependent-TERT (telomerase reverse transcriptase), therefore modulating age-dependent arteriopathy which helps reducing vascular aging (Ref. Reference Cakatay and Kayali124). Studies also indicate that it is not possible to keep enough levels of α-LA only from natural sources to see its anti-aging effect, so to have significant antioxidant activity it is needed to take α-LA as a supplement in higher doses. Therefore, the need of assessing its daily dose for humans occurred in the light of acute and subchronic toxicity, and potential mutagenic/genotoxic activity. Studies currently have found no adverse effects in the acute toxicity studies when given 175–550 mg/kg BW α-LA by oral gavage for 14-days to female CD Sprague–Dawley rats. At 2000 mg/kg BW, some rats ‘were reported to show signs of reduced well-being, including sedation, apathy, piloerection, hunched posture and/or eye closure. There was no effect of treatment observable on body weight gain or gross pathological examination. As a result of the study, the acute oral LD50 of α-LA was concluded to exceed 2000 mg/kg (BW), the highest dose tested in the study which indicates a very low order of acute toxicity. A 4-week sub-chronic toxicity study on both male and female Wistar rats was performed with the determined doses from low (31.6 mg/kg BW/day) to high (121 mg/kg BW/day). The no-observed-adverse-effect level (NOAEL) is considered to be 61.9 mg/kg BW/day. The results of these studies support the safety of α-LA (Cremer et al. 2006). The experimental safety dosage values were found 400 to 500 mg/kg for dogs, 30 mg/kg for cats and 500 mg/kg for mice (Ref. Reference Xiong123).

Figure 6. The chemical structure of α-lipoic acid.

Shay et al. 2019 compiled a series of clinical trials [ALADIN (I, II and III), SYDNEY (I and II) and ORPIL] using α-LA which also assessed adverse health effects in the participants. Oral doses of 1800 mg LA (600 mg t.i.d.) for 6 months did not elicit significant adverse effects compared to placebo (Ref. Reference Xiong123). However, there are a series of studies that determined increased plasma lipid hydroperoxide levels and oxidative protein damage with the intraperitoneal administration of racemic LA (100 mg/kg BW/day for 2 weeks) in aged rats (Refs Reference Kayali125, Reference Fogacci126). When the equivalent dose is calculated (5 to 10 g per day in humans) it can be seen as too high for human administration. In a systematic review and meta-analysis of randomised placebo-controlled clinical studies, Fogacci et al. 2020 discussed α-LA ‘safe’ as a supplement to improve health outcomes in overall healthy individuals and patients affected by other diseases (Ref. Reference Dos Santos127).

Although its therapeutic potential, drug/herbal interactions and adverse reactions have not been well established in clinical trials with populations at higher risk for diseases of aging (Ref. Reference Emir, Ozturan and Yilmaz128). α-LA has a low risk in prolonged use, however, according to two published case reports, scientists have drawn attention to serious α-LA acute intoxication by suicidal intention with doses of 18 and 6 g respectively (Refs Reference Hadzik129, Reference Parente130). The authorities concluded that α-LA can be considered safe as supplementation without any side effects with a range of daily doses ranging from 200 to 2400 mg/day. However, monitoring is advised when taking high doses of α-LA frequently (Ref. Reference Xiong123). Researchers conclude that for the safety and optimal dose of α-LA more studies are needed.

Since there is not any established contraindication of α-LA in pregnant women or children, it has been advised to consult with their healthcare providers before taking α-LA, considering the lack of evidence (Refs Reference Shay122, Reference Konrad131). Even though α-LA intoxication is extremely rare, close monitoring is necessary for children who are around diabetic patients, as most of the reported α-LA intoxication cases belong to children ingesting α-LA by accident (Ref. Reference Shay122).

Drug interactions: Patients with type 2 diabetes commonly intend to use dietary/herbal supplements to help decrease blood sugar. In the case of regarding a possible risk of herbal-drug interaction, patients who use metformin should avoid using α-LA as a supplement because of the synergistic effect (Ref. Reference Malviya132).

Resveratrol

Resveratrol (35,4′- trans-hydroxystilbene, Fig. 7) is a plant-specific polyphenolic compound produced by numerous plant species generally synthesised in terms of some stress conditions in fruits (especially grapes and several berries), nuts and table wines. The fruits of Vitis vinifera (grape) are dedicated as the most important dietary source of resveratrol (Ref. Reference Handore and Khandelwal133). It also belongs to the phytoalexin group because it has occurred under some stress conditions such as microbial infections, intense radiation and heavy metals (Ref. Reference Salehi134). The importance of resveratrol as a supplement for human health comes from several study results showing antioxidant, anticarcinogenic and antitumour activity (Ref. Reference Plauth135). The radical scavenging mechanism of resveratrol can be explained as acting as a pro-oxidant first, stimulating the free radical generation and then eliminating the ROS. This phenomenon is called ‘hormesis’ (Ref. Reference Kulkarni and Canto136). Besides its high antioxidant capacity studies showed that resveratrol activates proteins such as Sir2/SIRT1 and AMPK, which modulate lifespan in organisms (Ref. Reference Gutlapalli137). Resveratrol could be a potential therapeutic supplement in preventing long-term cardiovascular morbidity and mortality (Ref. Reference Wang138). Studies claim that resveratrol also significantly increased telomerase activity in vitro. Resveratrol has been shown to delay endothelial progenitor cells senescence by increasing the number of the cells, which may be dependent on telomerase activation (Ref. Reference Bresson139). Resveratrol was proven to prolong the lifetime with a range of 10 to 72% in several study models (Ref. Reference Gómez-Linton11). Resveratrol has cis- and trans- isomeric forms, of which numerous studies have focused on the biological activities and safety studies of trans-form (Refs Reference Plauth135, Reference Juan, Vinardell and Planas140, Reference Bhullar and Hubbard141). trans-Resveratrol has been found more stable with high bioactive effects (Ref. Reference Plauth135). According to the studies, resveratrol increases longevity and provides recovery for age-related diseases via scavenging free radicals (Refs Reference Ding15, Reference Wenzel and Somoza142). Together with rapamycin and curcumin, resveratrol is in the preclinical test phase of the ‘Interventions Testing Program of the National Institute on Aging’ to be a candidate for phytomedicinal drug (Ref. Reference Gómez-Linton11).

Figure 7. The chemical structures of cis-resveratrol and trans-resveratrol.

In human and rodent studies, it has been shown that resveratrol is well absorbed, distributed to various organs and mainly metabolised to glucuronide conjugates but has quite poor oral bioavailability and 75% of resveratrol is excreted via faeces and urine (Ref. Reference Anton143). After trans-resveratrol was administered orally to male Sprague-Dawley rats for 28 days at repeating doses of 20 mg/(kg day), body weight and other variables did not differ between rats treated with trans-resveratrol and the control group (Ref. Reference Bhullar and Hubbard141). A placebo-controlled trial performed on overweight and older adults suggest that short-term resveratrol supplementation at doses of 300 and 1000 mg/day does not adversely affect blood chemistries (Ref. Reference Lai144). The European Food Safety Authority (EFSA) reports that, intended the intake level of 150 mg/day for adults is within the safety margin of synthetic trans-resveratrol. It also notes that diarrhoea or other gastrointestinal symptoms were reported at doses of 1 g resveratrol/day or higher. The Panel remarks on a possible interaction with medicines that are mainly metabolised by CYP2C9 since the metabolite trans-resveratrol sulphate could inhibit CYP enzymes in humans (Ref. Reference Juan, Vinardell and Planas140).

Astaxanthin

Astaxanthin (AX, Fig. 8) is a carotenoid pigment found in Haematococcus pluvialis algae, and the marine animals that feed on them: the yeast Pfaffia rhodozyma (Xanthophyllomyces dendrorhous) and the bacterium Paracoccus carotinifaciens (Ref. Reference Goto145).

Figure 8. The chemical structure of astaxanthin.

Human nutritional sources like salmon, fish (like trout), and crustaceans provide the administration of astaxanthin in a normal diet. Astaxanthin has been found antioxidant many times higher than other antioxidant natural compounds known, especially scavenging singlet oxygen and peroxyl radicals (Refs Reference Goto145, Reference Brendler and Williamson146). Astaxanthin (AX)-containing preparations are recently popular as food supplements because of the potential to improve health in cardiovascular, neurodegenerative and skin diseases since the antioxidant activity has been affirmed (Refs Reference Niu147, Reference Buesen148). Because of the high demand for AX supplements, the separation and production of AX from natural sources have become ecologically unsustainable. Therefore, it is synthetically available however, the compounds are not identical. Natural AX has two isomers, 3S,3′S- and 3R,3′R-stereoisomers, in free and esterified form; whereas synthetic AX consists of a mixture of the isomers (3S, 3′S)-astaxanthin, (3R, 3′S)-astaxanthin and (3R and 3′R)-astaxanthin (Ref. Reference Niu147).

The studies provide information about the acute oral toxicity of natural astaxanthin, from H. pluvialis resulting in no adverse effect on mice. In some oral acute toxicity studies, the maximum tolerated dose of astaxanthin esters in ICR male mice was 21.5g/kg⋅BW, whereas the estimated oral LD50 of astaxanthin is greater than 20 g/kg⋅BW, observing no abnormalities in diet, behaviour, body weight or organ weight at mentioned a dose in pregnant mice. The repeat-dose toxicity test ranging from 100 to 500 mg/kg⋅BW astaxanthin showed no abnormalities in clinical observation throughout the pregnancy as well (Ref. Reference Buesen148). In a subchronic toxicity study of synthetic [3S, 3′S]-Astaxanthin in a gelatin/carbohydrate formulation, researchers determined a high dose of 700–920 mg/kg BW/day did not cause any adverse effects on Wistar rats (Ref. Reference Edwards149). Synthetic [3S, 3′S]-astaxanthin is nongenotoxic but rat carcinogenicity has been observed in females, at 200 mg/kg BW/day and above (Ref. Reference Turck150).

Brandler and Williamson 2019 listed eight clinical studies, testing natural AX at the safety of high doses ranging from 8 to 45 mg/day and over 4 to 12 weeks. No serious adverse effects and no changes in liver parameters were observed in any of the clinical studies except a red coloration of the stool as a minor effect was noted at the beginning. According to the review, natural AX has been revealed as a clinically safe supplement at short-term daily doses of up to 100 mg and long-term daily doses averaging between 8 and 12 mg (Ref. Reference Niu147).

According to the European Commission products that have AX-rich oleoresin from H. pluvialis should not exceed 8 mg of AX, the maximum ADI level (Ref. Reference Zia151). Additionally, the commission stipulated that companies add labelling for infants, children and adolescents younger than 14 years to not be consumed. USA authorities evaluate the safety condition of AX supplements into three categories: FDA-affirmed generally recognised as safe (GRAS), self-affirmed GRAS and New Dietary Ingredient Notifications. Regarding the clinical studies on the administration of astaxanthin, authorities all over the world determine the recommended or approved doses under the category ‘novel food ingredient’ ranging between 2 and 24 mg daily (Ref. Reference Niu147). Daily doses of maximum 12 mg is approved in Canada as well as Australia/New Zealand (Ref. Reference Niu147).

Curcumin

Curcumin (1,7-bis(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione, Fig. 9), has been reported to exert several health effects as the main natural polyphenol found in the rhizome of Curcuma longa L. (turmeric). Most of the benefits of curcumin are attributed to its antioxidant and anti-inflammatory effects. It is stated that the high free radical scavenging activity and inhibitory effect on acute and chronic inflammation are due to the electron-donating group on the phenolic rings of curcumin (Ref. Reference Khaw and Hande152). In a study performed on human brain tumour cells, curcumin inhibited telomerase activity in all the telomerase-positive cell lines. Long-term curcumin treatment has also resulted in significant telomere shortening in cancer cell lines, which explains its potent anti-tumorigenic and antiproliferative activity (Ref. Reference Aguilar153). Curcumin is now one of the most consumed food supplements in several countries. It is also accepted as a yellow pigment being used as a food additive (Ref. Reference Soleimani, Sahebkar and Hosseinzadeh154). In some acute toxicity studies with a single dose of 5.000 mg/kg body weight curcumin given orally to Swiss albino mice or albino Wistar rats, no toxic effects or adverse signs were observed for 14 days (Ref. Reference Suresh and Srinivasan155).

Figure 9. The chemical structure of curcumin.

Turmeric and curcumin are approved as nonmutagenic and nongenotoxic (Ref. Reference Suresh and Srinivasan155). Both turmeric and curcumin showed no mutagenic activity in bacteria treated with turmeric preparations containing up to 85% curcumin. The reproductive toxicity study also concluded that the NOAEL is 250–320 mg/kg BW/day (Ref. Reference Soleimani, Sahebkar and Hosseinzadeh154). Curcumin is absorbed in rats quantitatively higher than in humans, with results varying from 3.88–60% of the administered dose (Ref. Reference Hewlings and Kalman156).

According to JECFA (The Joint United Nations and World Health Organization Expert Committee on Food Additives) and EFSA (European Food Safety Authority) reports, the 0–3 mg/kg body weight dose is the allowable daily intake (ADI) of curcumin (Ref. Reference Goel, Jhurani and Aggarwal157). The United States Food and Drug Administration (FDA) has announced curcumin as being GRAS (Ref. Reference Anand158). Phase I clinical trials evaluated the safety profile of curcumin and it has been established that curcumin is safe given at high doses (12 g/day) over 3 months (Ref. Reference Bahramsoltani, Rahimi and Farzaei159). During pregnancy, curcumin was found safe in animals but there is not enough evidence of curcumin preparations for pregnant women (Ref. Reference Suresh and Srinivasan155). According to pharmacokinetic interaction with drug studies, the researchers refer to the possibility of curcumin, inhibiting cytochrome (CYP) isoenzymes and P-glycoprotein so it may interact with pharmacological agents like cardiovascular drugs, antidepressants, anticoagulants, antibiotics, chemotherapeutic agents and antihistamine. More studies are needed to determine the exact potential (Ref. Reference Juturu160).

Curcumin exhibits poor absorption, rapid metabolism and systemic elimination, resulting in poor bioavailability (Ref. Reference Goel, Jhurani and Aggarwal157). Therefore, in recent studies, scientists generate different formulations and complexes by nano-formulation techniques to increase the bioavailability of curcumin and its metabolites. Those supplements should be evaluated for their safety, toxicology and pharmacokinetics (Refs Reference Storka161, Reference Pancholi162, 165).

Conclusion and future perspective

Aging is the most prominent risk factor for many diseases, which is considered to be a complicated biological process. The biology of aging has only lately been thoroughly explored by scientists, who have focused on the evolutionarily conserved mechanisms of aging available to control functional decreases and the onset of disorders associated with aging processes. Regulatory hallmark methodologies that describe how aging is connected to physiological processes and lifestyle choices have provided a path for possible medication development.

Natural products are connected to age-associated chronic diseases, according to the combined findings from in vitro, animal, and clinical investigations over the past few decades. The body contains more plant-based nutraceuticals, such as antioxidants and anti-inflammatory compounds, when it consumes diets high in natural foods. Furthermore, plants and botanicals, medicinally derived substances from nature have been shown to have anti-aging characteristics and are utilised as a springboard for developing effective anti-aging medications. The scientific community now makes considerable use of dietary supplements and small-molecule natural products to find aging mechanisms that have been preserved throughout evolution. Additionally, they have been shown to affect senescence, nutrient-sensing metabolic signalling and mitochondrial function to slow down cellular aging and diseases associated with aging, including metabolic, cardiovascular, neurological and degenerative joint conditions.

Despite significant efforts, the most of our knowledge regarding the possible therapeutic advantages of natural products and nutritional supplements during cellular aging originates from fundamental studies employing native species that have been carefully modified for laboratory use. We cannot be assured, therefore, of the extent to which these natural products and dietary supplements alter aging mechanisms to reflect the effectiveness of aging in outbred populations of various environments. It is necessary to find solutions to the low bioavailability, off-target and steric effects problems related to natural products to produce information that could more easily be applied to human aging.

Additionally, research on the effects of natural products and dietary supplements on the health of the senior population has produced conflicting results in human clinical studies. A small sample size characterises the majority of these trials, and subgroup and sensitivity analyses are typically lacking. To identify the sequence of actual events that are crucial for comprehending the efficacy of natural products and dietary supplements in extending life, carefully controlled, large-scale interventional trials, along with biomedical informatics and multi-omics data, are required. Together, these initiatives will enable us to determine the effectiveness and discover novel genes associated with longevity that are impacted by these bioactive foods and dietary supplements.

Although natural products have benefits against aging with different molecular mechanisms, they should be used with caution due to limited information about their possible toxic properties and unknown dose interval. It is recommended that natural goods be treated with the same ‘hormesis’ theory that is used to describe the health benefits of calorie restriction and exercise. Many natural substances have a hormetic effect, such as low doses are advantageous while high doses are poisonous. Additionally, when natural products are taken in conjunction with pharmacological agents, the precipitant natural products might affect drug disposition and delivery, hence boosting or decreasing the therapeutic impact of the target drugs. Polypharmacy is another very common problem among elderly patients due to their chronic conditions. Older people also frequently use nutritional supplements and natural medications. Although, herbal medicinal products and dietary supplements are typically excluded from traditional definitions of polypharmacy, however, their possible interactions with other medications make them more likely to cause adverse drug interactions. Therefore, healthcare professionals should carefully follow elderly patients and routinely ask questions about the other medications that are not prescribed.

Even though the scientific studies implicate that most natural products are safe, tolerable and non-toxic, it is necessary to perform well-designed, large-scale randomised control trials to evaluate the short- and long-term effects and efficacy of these products. To conclusively show if natural products could extend the human lifespan, however, real-world clinical trials and fundamental research on dietary supplements and natural products are still needed.

References

Proshkina, E et al. (2020) Key molecular mechanisms of aging, biomarkers, and potential interventions. Molecular Biology 54, 777811.CrossRefGoogle ScholarPubMed
Mitnitski, A et al. (2017) Aging, frailty and complex networks. Biogerontology 18, 433446.CrossRefGoogle ScholarPubMed
Mc Auley, MT et al. (2017) Modelling the molecular mechanisms of aging. Bioscience Reports 37, BSR20160177.CrossRefGoogle ScholarPubMed
Chen, Y et al. (2022) Dietary supplements and natural products: an update on their clinical effectiveness and molecular mechanisms of action during accelerated biological aging. Frontiers in Genetics 13, 880421.Google ScholarPubMed
Yang, C et al. (2020) A natural product solution to aging and aging-associated diseases. Pharmacology & Therapeutics 216, 107673.CrossRefGoogle ScholarPubMed
Crimmins, E et al. (2008) Biomarkers related to aging in human populations. Advances in Clinical Chemistry 46, 161216.CrossRefGoogle ScholarPubMed
Prasad, S, Sung, B and Aggarwal, BB (2012) Age-associated chronic diseases require age-old medicine: role of chronic inflammation. Preventive Medicine 54, S29S37.CrossRefGoogle ScholarPubMed
Li, Z et al. (2021) Aging and age-related diseases: from mechanisms to therapeutic strategies. Biogerontology 22, 165187.CrossRefGoogle ScholarPubMed
Jayanthi, P, Joshua, E and Ranganathan, K (2010) Ageing and its implications. Journal of oral and Maxillofacial Pathology: JOMFP 14, 48.CrossRefGoogle ScholarPubMed
Newman, DJ and Cragg, GM (2020) Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. Journal of Natural Products 83, 770803.CrossRefGoogle ScholarPubMed
Gómez-Linton, DR et al. (2019) Some naturally occurring compounds that increase longevity and stress resistance in model organisms of aging. Biogerontology 20, 583603.CrossRefGoogle ScholarPubMed
Herranz, N and Gil, J (2018) Mechanisms and functions of cellular senescence. The Journal of Clinical Investigation 128, 12381246.CrossRefGoogle ScholarPubMed
Di Micco, R et al. (2021) Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nature Reviews Molecular Cell Biology 22, 7595.CrossRefGoogle ScholarPubMed
Mechchate, H et al. (2022) Vegetables and their bioactive compounds as anti-aging drugs. Molecules 27, 2316.CrossRefGoogle ScholarPubMed
Ding, AJ et al. (2017) Current perspective in the discovery of anti-aging agents from natural products. Natural Products Bioprospecting 7, 335404.CrossRefGoogle ScholarPubMed
Fan, X et al. (2022) Flavonoids-natural gifts to promote health and longevity. International Journal of Molecular Sciences 23, 2176.CrossRefGoogle ScholarPubMed
Gaston, TE et al. (2020) “Natural” is not synonymous with “safe”: toxicity of natural products alone and in combination with pharmaceutical agents. Regulatory Toxicology and Pharmacology 113, 104642.CrossRefGoogle Scholar
Niedernhofer, LJ et al. (2018) Nuclear genomic instability and aging. Annual Review of Biochemistry 87, 295322.CrossRefGoogle ScholarPubMed
Ferrucci, L et al. (2020) Measuring biological aging in humans: a quest. Aging Cell 19, e13080.CrossRefGoogle ScholarPubMed
Wu, Y et al. (2017) Association of grip strength with risk of all-cause mortality, cardiovascular diseases, and cancer in community-dwelling populations: a meta-analysis of prospective cohort studies. Journal of the American Medical Directors Association 18, 551.e17551.e35.CrossRefGoogle ScholarPubMed
Rozhok, A and DeGregori, J (2019) A generalized theory of age-dependent carcinogenesis. eLife 8, e39950.CrossRefGoogle ScholarPubMed
Piotrowski, I et al. (2017) Carcinogenesis induced by low-dose radiation. Radiology and Oncology 51, 369377.CrossRefGoogle ScholarPubMed
Kirkwood, TB (1996) Human senescence. Bioessays 18, 10091016.CrossRefGoogle ScholarPubMed
Dodig, S, Čepelak, I and Pavić, I (2019) Hallmarks of senescence and aging. Biochemia medica 29, 483497.CrossRefGoogle ScholarPubMed
López-Otín, C et al. (2013) The hallmarks of aging. Cell 153, 11941217.CrossRefGoogle ScholarPubMed
Tiwari, V and Wilson, DM III (2019) DNA damage and associated DNA repair defects in disease and premature aging. The American Journal of Human Genetics 105, 237257.CrossRefGoogle ScholarPubMed
Stegeman, R and Weake, VM (2017) Transcriptional signatures of aging. Journal of Molecular Biology 429, 24272437.CrossRefGoogle ScholarPubMed
Brosh, RM Jr. and Bohr, VA (2007) Human premature aging, DNA repair and RecQ helicases. Nucleic Acids Research 35, 75277544.CrossRefGoogle ScholarPubMed
Smith, EM, Pendlebury, DF and Nandakumar, J (2020) Structural biology of telomeres and telomerase. Cellular and Molecular Life Sciences 77, 6179.CrossRefGoogle ScholarPubMed
Katayama, M et al. (1991) Deleted mitochondrial DNA in the skeletal muscle of aged individuals. Biochemistry International 25, 4756.Google ScholarPubMed
Shu, Y et al. (2020) Association of dietary selenium intake with telomere length in middle-aged and older adults. Clinical Nutrition 39, 30863091.CrossRefGoogle ScholarPubMed
Sohal, RS and Weindruch, R (1996) Oxidative stress, caloric restriction, and aging. Science (New York, N.Y.) 273, 5963.CrossRefGoogle ScholarPubMed
Harman, D (2002) Aging: a theory based on free radical and radiation chemistry. Science of Aging Knowledge Environment 2002, cp14.CrossRefGoogle Scholar
Barja, G (2004) Free radicals and aging. TRENDS in Neurosciences 27, 595600.CrossRefGoogle ScholarPubMed
Liu, Z et al. (2018) Role of ROS and nutritional antioxidants in human diseases. Frontiers in Physiology 9, 477.CrossRefGoogle ScholarPubMed
Singh, PP et al. (2019) The genetics of aging: a vertebrate perspective. Cell 177, 200220.CrossRefGoogle ScholarPubMed
Wanke, V et al. (2008) Caffeine extends yeast lifespan by targeting TORC1. Molecular Microbiology 69, 277285.CrossRefGoogle ScholarPubMed
Lublin, A et al. (2011) FDA-approved drugs that protect mammalian neurons from glucose toxicity slow aging dependent on Cbp and protect against proteotoxicity. PLOS ONE 6, e27762.CrossRefGoogle ScholarPubMed
Sutphin, GL et al. (2012) Caffeine extends life span, improves healthspan, and delays age-associated pathology in Caenorhabditis elegans. Longevity & Healthspan 1, 9.CrossRefGoogle ScholarPubMed
Martin-Montalvo, A et al. (2013) Metformin improves healthspan and lifespan in mice. Nature Communications 4, 21922192.CrossRefGoogle ScholarPubMed
Onken, B and Driscoll, M (2010) Metformin induces a dietary restriction-like state and the oxidative stress response to extend C. elegans healthspan via AMPK, LKB1, and SKN-1. PLOS ONE 5, e8758.CrossRefGoogle ScholarPubMed
Lashmanova, E et al. (2015) Fucoxanthin increases lifespan of Drosophila melanogaster and Caenorhabditis elegans. Pharmacological Research 100, 228241.CrossRefGoogle ScholarPubMed
Eisenberg, T et al. (2009) Induction of autophagy by spermidine promotes longevity. Nature Cell Biology 11, 13051314.CrossRefGoogle ScholarPubMed
Harrison, DE et al. (2009) Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 460, 392395.CrossRefGoogle ScholarPubMed
Miller, RA et al. (2014) Rapamycin-mediated lifespan increase in mice is dose and sex dependent and metabolically distinct from dietary restriction. Aging Cell 13, 468477.CrossRefGoogle ScholarPubMed
Bjedov, I et al. (2010) Mechanisms of life span extension by rapamycin in the fruit fly Drosophila melanogaster. Cell Metabolism 11, 3546.CrossRefGoogle ScholarPubMed
Bauer, JH et al. (2004) An accelerated assay for the identification of lifespan-extending interventions in Drosophila melanogaster. Proceedings of the National Academy of Sciences of the United States of America 101, 1298012985.CrossRefGoogle ScholarPubMed
Wang, C et al. (2013) The effect of resveratrol on lifespan depends on both gender and dietary nutrient composition in Drosophila melanogaster. Age (Dordrecht, Netherlands) 35, 6981.CrossRefGoogle ScholarPubMed
Morselli, E et al. (2010) Caloric restriction and resveratrol promote longevity through the Sirtuin-1-dependent induction of autophagy. Cell Death & Disease 1, e10e10.CrossRefGoogle ScholarPubMed
Wu, W et al. (2014) Astaxanthin alleviates brain aging in rats by attenuating oxidative stress and increasing BDNF levels. Food & Function 5, 158166.CrossRefGoogle ScholarPubMed
Yazaki, K et al. (2011) Supplemental cellular protection by a carotenoid extends lifespan via Ins/IGF-1 signaling in Caenorhabditis elegans. Oxidative Medicine and Cellular Longevity 2011, 596240.CrossRefGoogle ScholarPubMed
Shen, LR et al. (2013) Curcumin-supplemented diets increase superoxide dismutase activity and mean lifespan in Drosophila. Age (Dordr) 35, 11331142.CrossRefGoogle ScholarPubMed
Cuanalo-Contreras, K et al. (2017) Delaying aging in Caenorhabditis elegans with protein aggregation inhibitors. Biochemical and Biophysical Research Communications 482, 6267.CrossRefGoogle ScholarPubMed
Ashihara, H and Suzuki, T (2004) Distribution and biosynthesis of caffeine in plants. Frontiers in Bioscience (Landmark Ed) 9, 18641876.CrossRefGoogle ScholarPubMed
Fisone, G, Borgkvist, A and Usiello, A (2004) Caffeine as a psychomotor stimulant: mechanism of action. Cellular and Molecular Life Sciences 61, 857872.CrossRefGoogle ScholarPubMed
Cappelletti, S et al. (2015) Caffeine: cognitive and physical performance enhancer or psychoactive drug? Current Neuropharmacology 13, 7188.CrossRefGoogle ScholarPubMed
Derry, CJ, Derry, S and Moore, RA (2012) Caffeine as an analgesic adjuvant for acute pain in adults. Cochrane Database of Systematic Reviews. CD009281.CrossRefGoogle Scholar
Alsunni, AA (2015) Energy drink consumption: beneficial and adverse health effects. International Journal of Health Sciences (Qassim) 9, 468474.Google ScholarPubMed
EFSA (2015) Scientific opinion on the safety of caffeine. EFSA Journal 13, 4102.Google Scholar
Tucker, LA (2017) Caffeine consumption and telomere length in men and women of the National Health and Nutrition Examination Survey (NHANES). Nutrition & Metabolism 14, 10.CrossRefGoogle ScholarPubMed
Willson, C (2018) The clinical toxicology of caffeine: a review and case study. Toxicology Reports 5, 11401152.CrossRefGoogle ScholarPubMed
Banerjee, P et al. (2014) Fatal caffeine intoxication: a series of eight cases from 1999 to 2009. Journal of Forensic Sciences 59, 865868.CrossRefGoogle ScholarPubMed
Rasmussen, BB, Nielsen, TL and Brøsen, K (1998) Fluvoxamine is a potent inhibitor of the metabolism of caffeine in vitro. Pharmacology & Toxicology 83, 240245.CrossRefGoogle Scholar
Jeppesen, U et al. (1996) A fluvoxamine-caffeine interaction study. Pharmacogenetics 6, 213222.CrossRefGoogle ScholarPubMed
Carrillo, JA and Benitez, J (2000) Clinically significant pharmacokinetic interactions between dietary caffeine and medications. Clinical Pharmacokinetics 39, 127153.CrossRefGoogle ScholarPubMed
Gong, L et al. (2012) Metformin pathways: pharmacokinetics and pharmacodynamics. Pharmacogenetics and Genomics 22, 820827.CrossRefGoogle ScholarPubMed
Aroda, VR and Ratner, RE (2018) Metformin and type 2 diabetes prevention. Diabetes Spectrum 31, 336342.CrossRefGoogle ScholarPubMed
Wiernsperger, N.F. and Bailey, C.J. (1999) The antihyperglycaemic effect of metformin: therapeutic and cellular mechanisms. Drugs 58(suppl. 1), 3139, discussion 75–82.CrossRefGoogle ScholarPubMed
Viollet, B et al. (2011) Cellular and molecular mechanisms of metformin: an overview. Clinical Science 122, 253270.CrossRefGoogle Scholar
Rena, G, Hardie, DG and Pearson, ER (2017) The mechanisms of action of metformin. Diabetologia 60, 15771585.CrossRefGoogle ScholarPubMed
Hu, D et al. (2021) Metformin: a potential candidate for targeting aging mechanisms. Aging and Disease 12, 480493.CrossRefGoogle ScholarPubMed
DeFronzo, R et al. (2016) Metformin-associated lactic acidosis: current perspectives on causes and risk. Metabolism: Clinical and Experimental 65, 2029.CrossRefGoogle ScholarPubMed
Pakkir Maideen, NM, Jumale, A and Balasubramaniam, R (2017) Drug interactions of metformin involving drug transporter proteins. Advanced Pharmacetical Bulletin 7, 501505.CrossRefGoogle ScholarPubMed
Stage, TB, Brøsen, K and Christensen, MMH (2015) A comprehensive review of drug–drug interactions with metformin. Clinical Pharmacokinetics 54, 811824.CrossRefGoogle ScholarPubMed
Adak, T et al. (2018) A reappraisal on metformin. Regulatory Toxicology and Pharmacology 92, 324332.CrossRefGoogle ScholarPubMed
Hajsadeghi, S et al. (2022) A concealed history behind the disaster: extremely rare presentations of metformin toxicity in a patient with body dysmorphic disorder. Toxicology Reports 9, 848851.CrossRefGoogle Scholar
Infante, M et al. (2021) Long-term metformin therapy and vitamin B12 deficiency: an association to bear in mind. World Journal Diabetes 12, 916931.CrossRefGoogle ScholarPubMed
Wang, GS and Hoyte, C (2019) Review of biguanide (metformin) toxicity. Journal of Intensive Care Medicine 34, 863876.CrossRefGoogle ScholarPubMed
Zhang, H et al. (2015) Fucoxanthin: a promising medicinal and nutritional ingredient. Evidence-Based Complementary and Alternative Medicine 2015, 723515.CrossRefGoogle ScholarPubMed
Kawee-ai, A, Kuntiya, A and Kim, SM (2013) Anticholinesterase and antioxidant activities of fucoxanthin purified from the microalga Phaeodactylum tricornutum. Natural Product Communications 8, 13811386.CrossRefGoogle ScholarPubMed
Mohibbullah, M et al. (2022) A systematic review on marine algae-derived fucoxanthin: an update of pharmacological insights. Marine Drugs 20, 279.CrossRefGoogle ScholarPubMed
Mao, H et al. (2022) Fucoxanthin attenuates oxidative damage by activating the sirt1/nrf2/ho-1 signaling pathway to protect the kidney from ischemia-reperfusion injury. Oxidative Medicine and Cellular Longevity 2022, 7444430.CrossRefGoogle ScholarPubMed
Shikov, AN et al. (2020) Pharmacokinetics of marine-derived drugs. Marine Drugs 18, 557.CrossRefGoogle ScholarPubMed
Ishikawa, C et al. (2008) Anti-adult T-cell leukemia effects of brown algae fucoxanthin and its deacetylated product, fucoxanthinol. International Journal of Cancer 123, 27022712.CrossRefGoogle ScholarPubMed
Abu-Ghannam, N and Shannon, E (2017) Seaweed carotenoid, fucoxanthin, as functional food. Microbial Functional Foods and Nutraceuticals, John Wiley & Sons Ltd., pp. 39–64.CrossRefGoogle Scholar
Beppu, F et al. (2009) In vitro and in vivo evaluation of mutagenicity of fucoxanthin (FX) and its metabolite fucoxanthinol (FXOH). Journal of Toxicological Sciences 34, 693698.CrossRefGoogle ScholarPubMed
Beppu, F et al. (2009) Single and repeated oral dose toxicity study of fucoxanthin (FX), a marine carotenoid, in mice. The Journal of Toxicological Science 34, 501510.CrossRefGoogle ScholarPubMed
Hitoe, S and Shimoda, H (2017) Seaweed fucoxanthin supplementation improves obesity parameters in mild obese Japanese subjects. Functional Foods in Health and Disease 7, 246262.CrossRefGoogle Scholar
Bresson, JL et al. (2009) Scientific opinion on the substantiation of health claims related to Undaria pinnatifida (Harvey) Suringar and maintenance or achievement of a normal body weight (ID 2345) pursuant to Article 13(1) of Regulation (EC) No 1924/2006. EFSA Journal 7, 1302.Google Scholar
Bae, M et al. (2020) Health benefits of fucoxanthin in the prevention of chronic diseases. Biochimica et Biophysica Acta (BBA) – Molecular and Cell Biology of Lipids 1865, 158618.CrossRefGoogle ScholarPubMed
Munoz-Esparza, NC et al. (2019) Polyamines in food. Frontiers in Nutritions 6, 108.CrossRefGoogle ScholarPubMed
Ramos-Molina, B et al. (2019) Dietary and gut microbiota polyamines in obesity- and age-related diseases. Frontiers in Nutritions 6, 24.CrossRefGoogle ScholarPubMed
Zou, D et al. (2022) A comprehensive review of spermidine: safety, health effects, absorption and metabolism, food materials evaluation, physical and chemical processing, and bioprocessing. Comprehensive Reviews in Food Science Food Safety 21, 28202842.CrossRefGoogle ScholarPubMed
Madeo, F et al. (2018) Spermidine in health and disease. Science (New York, N.Y.) 359, eaan2788.CrossRefGoogle ScholarPubMed
Wirth, A et al. (2021) Novel aspects of age-protection by spermidine supplementation are associated with preserved telomere length. Geroscience 43, 673690.CrossRefGoogle ScholarPubMed
Schwarz, C et al. (2018) Safety and tolerability of spermidine supplementation in mice and older adults with subjective cognitive decline. Aging (Albany NY) 10, 1933.CrossRefGoogle ScholarPubMed
Til, HP et al. (1997) Acute and subacute toxicity of tyramine, spermidine, spermine, putrescine and cadaverine in rats. Food and Chemical Toxicology 35, 337348.CrossRefGoogle ScholarPubMed
Pegg, AE (2013) Toxicity of polyamines and their metabolic products. Chemical Research in Toxicology 26, 17821800.CrossRefGoogle ScholarPubMed
Madeo, F et al. (2019) Spermidine: a physiological autophagy inducer acting as an anti-aging vitamin in humans? Autophagy 15, 165168.CrossRefGoogle ScholarPubMed
Soda, K et al. (2009) Long-term oral polyamine intake increases blood polyamine concentrations. Journal of Nutritional Science and Vitaminology 55, 361366.CrossRefGoogle ScholarPubMed
Zou, D et al. (2022) A comprehensive review of spermidine: safety, health effects, absorption and metabolism, food materials evaluation, physical and chemical processing, and bioprocessing. Comprehensive Reviews in Food Science and Food Safety 21, 28202842.CrossRefGoogle ScholarPubMed
SpermidineLIFE. Available at https://www.spermidinelife.com/en/ (Accessed on July 2022).Google Scholar
Abraham, RT and Wiederrecht, GJ (1996) Immunopharmacology of rapamycin. Annual Review of Immunology 14, 483510.CrossRefGoogle ScholarPubMed
Seto, B (2012) Rapamycin and mTOR: a serendipitous discovery and implications for breast cancer. Clinical and Translational Medicine 1, 29.CrossRefGoogle ScholarPubMed
Mahalati, K and Kahan, BD (2001) Clinical pharmacokinetics of sirolimus. Clinical Pharmacokinetics 40, 573585.CrossRefGoogle ScholarPubMed
MacDonald, A et al. (2000) Clinical pharmacokinetics and therapeutic drug monitoring of sirolimus. Clinical Therapeutics 22, B101BB21.CrossRefGoogle ScholarPubMed
Yoo, YJ et al. (2017) An overview of rapamycin: from discovery to future perspectives. Journal of Industrial Microbiology and Biotechnology 44, 537553.CrossRefGoogle ScholarPubMed
Ferrara-Romeo, I et al. (2020) The mTOR pathway is necessary for survival of mice with short telomeres. Nature Communications 11, 1168.CrossRefGoogle ScholarPubMed
Sádaba, B et al. (2004) Clinical relevance of sirolimus drug interactions in transplant patients. Transplantation Proceedings 36, 32263228.CrossRefGoogle ScholarPubMed
Leung, LY et al. (2006) Pharmacokinetics and metabolic disposition of sirolimus in healthy male volunteers after a single oral dose. Therapeutic Drug Monitoring 28, 5161.CrossRefGoogle ScholarPubMed
Glotzbecker, B et al. (2012) Important drug interactions in hematopoietic stem cell transplantation: what every physician should know. Biology of Blood and Marrow Transplantation 18, 9891006.CrossRefGoogle ScholarPubMed
Fischer, KE et al. (2015) Health effects of long-term rapamycin treatment: the impact on mouse health of enteric rapamycin treatment from four months of age throughout life. PLoS One 10, e0126644.CrossRefGoogle ScholarPubMed
Neff, F et al. (2013) Rapamycin extends murine lifespan but has limited effects on aging. The Journal of Clinical Investigation 123, 32723291.CrossRefGoogle ScholarPubMed
Wilkinson, JE et al. (2012) Rapamycin slows aging in mice. Aging Cell 11, 675682.CrossRefGoogle ScholarPubMed
Barlow, AD, Nicholson, ML and Herbert, TP (2013) Evidence for rapamycin toxicity in pancreatic beta-cells and a review of the underlying molecular mechanisms. Diabetes 62, 26742682.CrossRefGoogle Scholar
Ross, C et al. (2015) Metabolic consequences of long-term rapamycin exposure on common marmoset monkeys (Callithrix jacchus). Aging (Albany NY) 7, 964973.CrossRefGoogle ScholarPubMed
Urfer, SR et al. (2017) A randomized controlled trial to establish effects of short-term rapamycin treatment in 24 middle-aged companion dogs. Geroscience 39, 117127.CrossRefGoogle ScholarPubMed
Marti, HP and Frey, FJ (2005) Nephrotoxicity of rapamycin: an emerging problem in clinical medicine. Nephrology Dialysis Transplantation 20, 1315.CrossRefGoogle ScholarPubMed
Nguyen, H and Gupta, V (2022) Alpha-Lipoic Acid, StatPearls (Internet), StatPearls Publishing, Treasure Island (FL).Google Scholar
Shay, KP et al. (2009) Alpha-lipoic acid as a dietary supplement: molecular mechanisms and therapeutic potential. Biochimica et Biophysica Acta 1790, 11491160.CrossRefGoogle ScholarPubMed
Xiong, S et al. (2015) PGC-1α modulates telomere function and DNA damage in protecting against aging-related chronic diseases. Cell Reports 12, 13911399.CrossRefGoogle ScholarPubMed
Cakatay, U and Kayali, R (2005) Plasma protein oxidation in aging rats after alpha-lipoic acid administration. Biogerontology 6, 8793.Google ScholarPubMed
Kayali, R et al. (2006) Effect of alpha-lipoic acid supplementation on markers of protein oxidation in post-mitotic tissues of ageing rat. Cell Biochemistry & Function 24, 7985.CrossRefGoogle ScholarPubMed
Fogacci, F et al. (2020) Safety evaluation of alpha-lipoic acid supplementation: a systematic review and meta-analysis of randomized placebo-controlled clinical studies. Antioxidants (Basel) 9, 1011.Google ScholarPubMed
Dos Santos, SM et al. (2019) Mitochondrial dysfunction and alpha-lipoic acid: beneficial or harmful in Alzheimer's Disease? Oxidative Medicine and Cellular Longevity 2019, 8409329.CrossRefGoogle ScholarPubMed
Emir, DF, Ozturan, IU and Yilmaz, S (2018) Alpha lipoic acid intoxication: an adult. The American Journal of Emergency Medicine 36, 1125.e31125.e5.CrossRefGoogle ScholarPubMed
Hadzik, B et al. (2014) Fatal non-accidental alpha-lipoic acid intoxication in an adolescent girl. Klinische Pädiatrie 226, 292294.Google Scholar
Parente, E et al. (2017) Safety of oral alpha-lipoic acid treatment in pregnant women: a retrospective observational study. European Review for Medical and Pharmacological Sciences 21, 42194227.Google ScholarPubMed
Konrad, T et al. (1999) alpha-Lipoic acid treatment decreases serum lactate and pyruvate concentrations and improves glucose effectiveness in lean and obese patients with type 2 diabetes. Diabetes Care 22, 280287.CrossRefGoogle ScholarPubMed
Malviya, V et al. (2022) A brief review on resveratrol. Asian Journal of Research in Pharmaceutical Sciences 12, 157162.CrossRefGoogle Scholar
Handore, AV and Khandelwal, SR (2017) Resveratrol - The nutraceutical, whose real time has come: a systematic review. International Journal of Advanced Biotechnology and Research 8, 1516–U220.Google Scholar
Salehi, B et al. (2018) Resveratrol: a double-edged sword in health benefits. Biomedicines 6, 91.CrossRefGoogle ScholarPubMed
Plauth, A et al. (2016) Hormetic shifting of redox environment by pro-oxidative resveratrol protects cells against stress. Free Radical Biology and Medicine 99, 608622.CrossRefGoogle ScholarPubMed
Kulkarni, SS and Canto, C (2015) The molecular targets of resveratrol. Biochimica Et Biophysica Acta-Molecular Basis of Disease 1852, 11141123.CrossRefGoogle ScholarPubMed
Gutlapalli, SD et al. (2020) The effects of resveratrol on telomeres and post myocardial infarction remodeling. Cureus 12, e11482.Google ScholarPubMed
Wang, XB et al. (2011) Resveratrol-induced augmentation of telomerase activity delays senescence of endothelial progenitor cells. Chinese Medical Journal 124, 43104315.Google ScholarPubMed
Bresson, JL et al. (2016) Safety of synthetic trans-resveratrol as a novel food pursuant to Regulation (EC) No 258/97. EFSA Journal 14, 4368.Google Scholar
Juan, ME, Vinardell, MP and Planas, JM (2002) The daily oral administration, of high doses of trans-resveratrol to rats for 28 days is not harmful. Journal of Nutrition 132, 257260.CrossRefGoogle Scholar
Bhullar, KS and Hubbard, BP (2015) Lifespan and healthspan extension by resveratrol. Biochimica Et Biophysica Acta-Molecular Basis of Disease 1852, 12091218.CrossRefGoogle ScholarPubMed
Wenzel, E and Somoza, V (2005) Metabolism and bioavailability of trans-resveratrol. Molecular Nutrition & Food Research 49, 472481.CrossRefGoogle ScholarPubMed
Anton, SD et al. (2014) Safety and metabolic outcomes of resveratrol supplementation in older adults: results of a twelve-week, placebo-controlled pilot study. Experimental Gerontology 57, 181187.CrossRefGoogle ScholarPubMed
Lai, J-P et al. (2004) Separation and determination of astaxanthin from microalgal and yeast samples by molecularly imprinted microspheres. Journal of Chromatography B 804, 2530.CrossRefGoogle ScholarPubMed
Goto, S et al. (2001) Efficient radical trapping at the surface and inside the phospholipid membrane is responsible for highly potent antiperoxidative activity of the carotenoid astaxanthin. Biochimica et Biophysica Acta (BBA) – Biomembranes 1512, 251258.CrossRefGoogle ScholarPubMed
Brendler, T and Williamson, EM (2019) Astaxanthin: how much is too much? A safety review. Phytotherapy Research 33, 30903111.CrossRefGoogle Scholar
Niu, T et al. (2020) Safety assessment of astaxanthin from Haematococcus pluvialis: acute toxicity, genotoxicity, distribution and repeat-dose toxicity studies in gestation mice. Regulatory Toxicology and Pharmacology 115, 104695.CrossRefGoogle ScholarPubMed
Buesen, R et al. (2015) Safety assessment of [3S, 3’S]-astaxanthin-aubchronic toxicity study in rats. Food and Chemical Toxicology 81, 129136.CrossRefGoogle ScholarPubMed
Edwards, JA et al. (2016) Review of genotoxicity and rat carcinogenicity investigations with astaxanthin. Regulatory Toxicology and Pharmacology 75, 519.CrossRefGoogle ScholarPubMed
Turck, D et al. (2020) Safety of astaxanthin for its use as a novel food in food supplements. EFSA Journal 18, e05993.Google ScholarPubMed
Zia, A et al. (2021) The role of curcumin in aging and senescence: molecular mechanisms. Biomedicine & Pharmacotherapy 134, 111119.CrossRefGoogle ScholarPubMed
Khaw, AK and Hande, MP (2007) Inhibition of telomerase activity and cell growth by curcumin in human brain tumour cells. Cancer Research 67(9_Supplement), 12181218.Google Scholar
Aguilar, F et al. (2010) Scientific opinion on the re-evaluation of curcumin (E 100) as a food additive EFSA panel on food Additives and Nutrient Sources added to food (ANS). EFSA Journal 8, 1679.Google Scholar
Soleimani, V, Sahebkar, A and Hosseinzadeh, H (2018) Turmeric (Curcuma longa) and its major constituent (curcumin) as nontoxic and safe substances: review. Phytotherapy Research 32, 985995.CrossRefGoogle ScholarPubMed
Suresh, D and Srinivasan, K (2010) Tissue distribution & elimination of capsaicin, piperine & curcumin following oral intake in rats. Indian Journal of Medical Research 131, 682691.Google ScholarPubMed
Hewlings, SJ and Kalman, DS (2017) Curcumin: a review of its effects on human health. Foods (Basel, Switzerland) 6, 92.Google ScholarPubMed
Goel, A, Jhurani, S and Aggarwal, BB (2008) Multi-targeted therapy by curcumin: how spicy is it? Molecular Nutrition & Food Research 52, 10101030.CrossRefGoogle Scholar
Anand, P et al. (2007) Bioavailability of curcumin: problems and promises. Molecular Pharmaceutics 4, 807818.CrossRefGoogle ScholarPubMed
Bahramsoltani, R, Rahimi, R and Farzaei, MH (2017) Pharmacokinetic interactions of curcuminoids with conventional drugs: a review. Journal of Ethnopharmacology 209, 112.CrossRefGoogle ScholarPubMed
Juturu, V et al. (2015) Antioxidant properties and safety evaluation of curcumin (CurcuWINTM). Faseb Journal 29, 294.5.CrossRefGoogle Scholar
Storka, A et al. (2015) Safety, tolerability and pharmacokinetics of liposomal curcumin in healthy humans. International Journal of Clinical Pharmacology and Therapeutics 53, 5465.CrossRefGoogle ScholarPubMed
Pancholi, V et al. (2021) Safety assessment of a highly bioavailable curcumin-galactomannoside complex (CurQfen) in healthy volunteers, with a special reference to the recent hepatotoxic reports of curcumin supplements: a 90-days prospective study. Toxicology Reports 8, 12551264.CrossRefGoogle Scholar
Figure 0

Table 1. Examples of anti-aging compounds from natural sourcesa (Refs 15, 16)

Figure 1

Table 2. The longevity effects of the selected compounds

Figure 2

Figure 1. The chemical structure of caffeine.

Figure 3

Figure 2. The chemical structure of metformin.

Figure 4

Figure 3. The chemical structure of fucoxanthin.

Figure 5

Figure 4. The chemical structure of spermidine.

Figure 6

Figure 5. The chemical structure of rapamycin.

Figure 7

Figure 6. The chemical structure of α-lipoic acid.

Figure 8

Figure 7. The chemical structures of cis-resveratrol and trans-resveratrol.

Figure 9

Figure 8. The chemical structure of astaxanthin.

Figure 10

Figure 9. The chemical structure of curcumin.