8 Dec 2020

Oxidative ageing: from proximate to ultimate causes

Oxidative stress seems really important in age-related decline and disease—but what causes it? Here I’ve tried to express a broadening perspective, by exploring its core, context and ultimate causes; and largely anchored in human studies where possible.

We all die—what matters is how. While human life expectancy has increased, non-communicable diseases are now the major cause of disability and death globally (WHO and OWID). These are mostly age-related diseases (e.g. CVD, cancer, COPD, dementia, etc.), which develop slowly over time, and coexist as multimorbidity (e.g. most people >65 in US/UK 1,2); resulting in functional decline/frailty and socioeconomic burden (i.e. productivity, sick care). This situation is growing globally, as populations are ageing, and diseases occur earlier—so we may live longer but sicker 1. Moreover, this invisible epidemic underlies susceptibility to (communicable) infectious diseases, such as COVID-19 3, elevating chronic disease to acute threat.

Rather than treat each age-related malady as it arises, the other approach is to consider ageing itself 4. Ageing (i.e. growing old) involves a gradual systemic decline of tissue function and stability, with increasing susceptibility to disease and eventually death. However, variation between individuals and species highlights disconnect between biological and chronological ageing, and opportunities to identify modulators of healthspan. In particular, the accelerating effect of (natural) radiation is evident; and similarly, since the 1950s ageing was suggested to involve the deleterious side effects of free radicals (i.e. Harman’s seminal free radical theory) 5,6, with subsequent incorporation into other theories (e.g. antagonistic pleiotropy 7, mitohormesis 8 and redox 9).

The accrued observational evidence supports that human ageing is accompanied by increasing oxidative stress, alongside decreasing antioxidant/adaptive responses 9–12 (esp. with frailty and disease 9,12,13, but less so in centenarians 14); while higher intakes and blood levels of various antioxidants are (dose-dependently) associated with reduced total and cause-specific mortality 15,16. On the other hand, many antioxidant trials (i.e. mainly vit-A/C/E, β-carotene and/or selenium; primary and secondary prevention; published ~1990–10) and meta-analyses thereof show general failure to lower mortality, and even potential harm 17 (e.g. previous post). This may be due to overly crude interventions based on early paradigms 8,9. As such, this article explores a broader perspective.

Redox theory

Free radicals and antioxidants really represent oxidation and reduction (redox) reactions, respectively, within a more complete concept of redox biology, which recognises all such reactions and their place in redox networks connected to metabolism 18,19. For instance, antioxidants (e.g. vit-C and E) have specific chemistry and cell distribution (e.g. cytoplasm and membranes, respectively), but function together within regenerative networks which draw electrons from intermediary metabolism, and ultimately food (Fig. 1).

Fundamentally, redox reactions couple the biology of life, to the chemistry of our planet and physics of the universe. From a bio-thermodynamic perspective, organisms are open systems, where internal energy/order derives from the external environment. This biochemistry requires manipulation of chemical bonds and electron transfer in thermodynamically favourable redox reactions (i.e. a redox tower). In particular, rising atmospheric oxygen coincided with the evolution of aerobic life, which exploits the electronegativity of oxygen as a terminal electron acceptor in energy metabolism, thereby maximising energy yield; while also serving in biosynthesis and production of reactive oxygen species (ROS—incl. free radicals) 20. This may have been accompanied by concurrent evolution of adaptive antioxidant responses (e.g. Nrf2 21), and elaboration of redox sensing/signalling with increasing metazoan complexity (e.g. NOX 20 and redox proteome 22,23).

Redox reactions are used in a myriad of metabolic and signalling pathways. The initial oxidation of nutrients generates a reductive cache (as NAD(P)H) which fuels electron flow through intermediary metabolism; including the incidental and intentional generation of ROS (e.g. via the ETC and NOX, respectively) and counterpoise antioxidant systems, which together regulate basic cell biology. For instance, the abundant thiol-antioxidants assume specific redox potentials (Eh in mV) in specific subcellular compartments (e.g. cytoplasm, mitochondria, nucleus, etc.), staging redox environments where transient/reversible oxidations can modulate macromolecule structure and function (e.g. metabolic/signalling activity) 22,23, and sustained oxidations mediate destruction/death, as weaponised in immunity (e.g. oxidative burst 24 and cytotoxicity 25). Ultimately, these redox networks follow thermodynamic hierarchy: bioenergetic systems operate at near-equilibrium with high-flux, generating energy for maintenance of thiol-antioxidants at non-equilibrium, where subcellular redox potentials maintain spatiotemporal organisation and structure 18.

Overall, redox biology represents a fundamental interface between environment and genome, which couples energy transfer to organisation/adaptation 18,19,23 (i.e. Gibbs free energy to order). On the other hand, ageing and disease typically involve a general oxidation of redox status (e.g. thiols and macromolecules) 9—so what gives and with what consequence?

Glutathione

Glutathione (GSH) is the major small molecule antioxidant (i.e. thiol tripeptide—307Da) synthesised by our cells; serving as a redox buffer and regulator of numerous processes (e.g. signalling/cysteine proteome 26, energy/mitochondria 27,28, growth–death 28,29, etc.). The reduced to oxidised glutathione ratio (GSH:GSSG) represents a functional marker of redox status, which varies between cell compartments and conditions. In general, ageing is accompanied by decreasing GSH and/or increasing GSSG 10,30,31, and thereby oxidation of GSH:GSSG. More specifically, in a US study on healthy human plasma, cysteine oxidation was linear with age, whereas GSH oxidation started at ~45yrs old, with considerable variation (i.e. Eh of GSH:GSSG vs. age, R2 = 0.32) 9,32.

Proximally, GSH decline has been associated with a lower biosynthetic rate 33,34, enzyme expression (e.g. GCL 10,31 and GSS 35), enzyme activity (e.g. Km of GCL 30,36,37 and GR 38) and precursor levels (i.e. glycine and cysteine) 33; while precursor supplementation has restored GSH and organ function in old animals 39–43 and humans 33–35,44,45 (reviews 46–48).

For instance, in 2011, a small study on healthy older adults (n=8, age 60–75), with stable-isotope tracing, revealed low precursor levels (i.e. RBC glycine –55%, cysteine –24%), while just 2 weeks of glycine-cysteine supplementation (~0.1g of each/kg/day) almost completely normalised RBC GSH synthesis rates, GSH:GSSG ratio and plasma oxidative markers (i.e. d-ROMS, F2-isoprostanes and lipid peroxides) to values of younger controls (n=8, age 30–40; + subgrouping for BMI and glycaemia) 33; and subsequently also mitochondrial fat oxidation and insulin sensitivity 35. The same protocol in older people with HIV similarly improved GSH and oxidative markers, as well as fed/fasting mitochondrial fuel oxidation, insulin sensitivity, blood lipids, body composition, muscle strength 34 and inflammation 44. And subsequent trials report sustained responses at 12 and 24 weeks, which declined after treatment withdrawal 49,50.

Among other research, in 1997, a relatively large placebo-controlled trial on a cohort of mostly older people with chronic diseases (n=262), with the GSH-precursor NAC for 6 months (2x 600mg/day), lowered flu symptoms and restored cell-mediated immunity 45. Accordingly, studies on lymphocytes from older people have linked low GSH and oxidative stress to reversible impairment of cytokine secretion and proliferation 51–53, while treatment of old animals with GSH-precursors has restored type-1 immunity 43 and resistance to infection 42. In humans, plasma aminothiol oxidation has also been associated with cognitive decline, cardiovascular dysfunction and mortality (see refs in 54), while in animal models GSH-precursors have restored synaptic plasticity (to that of the adult) 40, brain markers (e.g. inflammation and neurodegeneration) 41 and cardiac function/markers (e.g. inflammation and mitochondria) 39. Therefore GSH redox may regulate systemic organ function during ageing, including ‘inflamm-ageing’.

Loss of GSH redox may involve broader changes to sulfur metabolism 30. In the ageing mouse liver, there were increased transsulfuration metabolites, including homocysteine (154%), which was shown to lower the affinity of GCL for its substrates, potentially restraining GSH synthesis 37. The lower methionine further suggests slow homocysteine remethylation by MS (B12/folate-dependant) or BHMT (betaine-dependant) pathways. Similarly, a human autopsy study showed the ageing brain is accompanied by elevated homocysteine and lower B12, methionine and SAM/SAH ratios 55. In healthy adults, correlations were also reported between redox markers and NAD(H), with homocysteine, folate and B12, in blood and cerebrospinal fluid 56,57. Stepping further out, several other amino acids decline during ageing, including serine, a precursor of glycine and cysteine, and source of 1-carbon units (via SHMT) for folate-dependent methylation 58. In human skin cells, age-related mitochondrial respiration defects were linked to epigenetic suppression of glycine synthesis (i.e. GCAT and SHMT2 genes) 59. Further, in mid-age people with fatty liver (i.e. NAFLD), low serine/glycine was linked to low GSH, which improved with serine supplementation 60.

Regeneration of GSH (and other antioxidants) depends on electrons from NADPH (Fig. 1), which mainly come from cytosolic pentose phosphate (PPP) and mitochondria-driven pathways (e.g. 1-carbon, Krebs cycle and NNT) 61,62. With ageing, NADPH may decline in cells 63, but increase in human plasma within a dysregulated NAD metabolome 64. Notably, in a small trial on older adults (n=12, mean age 72), the NAD-precursor nicotinamide riboside acutely (post 2hrs) improved redox status (e.g. RBC NAD(P)H, GSH and F2-isoprostanes) and exercise performance 38. NAD may decline with age in relation to increased consumption and decreased synthesis 65,66; the former being associated with oxidative DNA damage and PARP activity in humans 64,67,68, and senescence/inflammation-driven CD38 activity in animals 69. Regardless, NAD use (i.e. via PARP, CD38 or SIRT) specifically removes the ADP-ribose portion, leaving NAM, while resynthesis requires ATP and ribose (as PRPP) 61. Upstream, glucose metabolism via the PPP generates ribose (as R5P), which also fuels de novo purine synthesis and salvage of AMP (i.e. precursor to ADP/ATP). Notably, purine synthesis requires various cofactors, including glycine and folate 70. Moreover, in ageing humans, low plasma NAD was accompanied by increased NAM and methyl-NAM 64; while NAM methylation consumes SAM and may be promoted by ATP deficit and inflammation 71,72. In summary, all these studies suggest potential links from GSH/redox to broader changes in 1-carbon 62 and NAD metabolism 63.

Nrf2 networks

Various antioxidant enzymes also decline during ageing 10, while their manipulation may affect healthspan 73. However, of central importance may be the transcription factor Nrf2 10. Nrf2 serves as a master regulator of redox homeostasis and cytoprotection, by controlling the expression of hundreds of genes, including antioxidants/GSH. Nrf2 has constitutive activity and is heavily induced by oxidative/stress conditions. However, basal and/or inducible Nrf2 activity declines during ageing and related disease 10,11 (e.g. in humans 74–80); and in relation to altered GCLC promoter binding and low GSH 81,82. Note, in the studies above, neither glycine-cysteine or GSH supplementation restored Nrf2 expression 35,42. Moreover, in animals and humans, high-dose antioxidant supplements (e.g. vit-C/E or NAC) can even inhibit exercise-induced Nrf2 signalling and mitohormesis 8,83,84. This may represent a significant shortfall of many antioxidant approaches, since Nrf2 controls so much of relevance to ageing; including many antioxidant/detox systems, mitochondria and intermediary metabolism (e.g. NADPH, 1-carbon, etc.), iron metabolism, AGE metabolism (i.e. glyoxalase-1), autophagy, DNA repair, etc. 85–88.

Accordingly, the Nrf2 pathway has been linked to ageing phenotypes. For instance, Nrf2 signalling is suppressed in human progeria (a genetic disease of premature ageing), where experimental evidence suggests it may drive the condition 89. More generally, in a cohort of community-dwelling adults (n=350, age ≥65), lower Nrf2 expression in blood was associated with frailty (independent of comorbidities) 13. In preclinical studies, Nrf2 deficiency replicates several major transcriptomic changes occurring in the human brain during ageing and Alzheimer’s 90; and regulates many other age-related changes, including sarcopenia 91, vascular function 77, immunity 43, neuroinflammation 92, neurogenesis 93, lens defence 74, collagen breakdown 94, carcinogenesis 95, etc.; as well as many cellular hallmarks of ageing 11,96. Consequently, in animal models, Nrf2 supports stress-resistance, healthspan and lifespan 10,11,90,95,97; as does overexpression of target genes GCL 30 and G6PD 98 (produces NADPH in PPP). Also noteworthy, NADPH-dependant mitochondrial reductase TXNRD2 (and GR) has been associated with cross-species lifespan (i.e. birds, rodents and primates) 99.

Why does Nrf2 activity decline with age? Nrf2 is normally sequestered in the cytoplasm by Keap1, while being regulated at multiple levels by many other proteins and modifications 10. Age-related changes include decreased Nrf2 expression, with corresponding changes to various positive/negative regulators (e.g. Keap1 75,80, Bach1 and c-Myc 78,82,100) and epigenetics 10,11. For instance, in human lens cells, age and metabolic stresses were reported to lower DNA methylation of the Keap1 promoter to suppress Nrf2 activity 80,101. More broadly, Nrf2 functions within integrated metabolite-sensing networks which also decline, including sirtuins and AMPK 102 (Fig. 2). For instance, NAD-dependent sirtuins stimulate Nrf2 (e.g. SIRT1 103–105, 2 106 and 6 107), reciprocally 108–110 (via p53 110); while Nrf2/GSH may also support NAD levels 111–113 (e.g. ↑ NQO1 112,113, ribose/purines 85; ↓ PARP 64,67, CD38 114,115 and SASP 69,116,117). And the energy (AMP:ATP) sensor AMPK induces NAD synthesis (via NAMPT), SIRT1 102 and Nrf2 118,119 (via Bach1 119), also potentially reciprocally 87 (e.g. via Trx1 120).

Besides NAD 65,66, these integrated pathways might decline in relation to various other signals and metabolites. For instance, they are induced by 1-carbon metabolites (e.g. serine 58 and folate 121,122), and several others linked to methionine metabolism (which may decline during ageing), including the methyl donor SAM 55,123, the polyamine spermidine 124,125, the circadian hormone melatonin 126,127 and the gaseous mediator H2S 128–131. Meanwhile, as above, human ageing can involve decreased methionine (i.e. precursor to SAM; and thereby polyamines, melatonin, DNA methylation, etc.) and increased homocysteine (i.e. precursor to methionine and cysteine/H2S 132); in relation to oxidative stress 133 and B12 deficiency 55, and ultimately impaired MS activity 134. Whereas Nrf2 activity may reciprocally oppose all this by supporting serine 85,135 and homocysteine metabolism (i.e. ↑ B12/MS 136,137 and CBS/CSE 138).

The gut microbiome

The gut microbiome shifts during ageing and can be used to predict chronological age 139. This may be of consequence to systemic metabolism and redox. For instance, the prevalence of small intestinal bacterial overgrowth (SIBO) may increase with age 139–141, which could increase nutrient competition with host, resulting in malabsorption (e.g. B12 140). Similarly, the gut microbiome also regulates systemic GSH metabolism, by limiting serine/glycine availability, which might be of importance in age-related metabolic disease 60,142. On the other hand, the colonic microbiome is emerging as a source of host folate 143,144 (also an auxotrophic nutrient for beneficial butyrate-producing bacteria 145­), the NAD-precursor NAM 146 and polyamines 124.

The gut microbiome also profoundly regulates immunity; and since 1907, Metchnikoff already suggested gut barrier disruption may drive chronic inflammation and senescence 147. Accordingly, recent studies in older humans have shown that microbial markers (i.e. LPS, LBP and sCD14) are elevated in plasma 148,149 and correlate inflammation, glucose control (i.e. HbA1c) and poor physical function 148–150; whereas centenarians had lower LPS than even young adults 151. In animal models, the gut microbiota drives inflamm-ageing 147, in association with gut dysbiosis (e.g. Akkermansia; TM7 and proteobacteria 152), gut inflammation and microbial translocation 147,152. Microbial components can also trigger production of antimicrobial ROS/RNS, which may burden systemic redox homeostasis. Accordingly, blood LPS correlated NOX2 and oxidative stress in elderly people with neurodegenerative diseases 153.

Age-related oxidative stress and organ dysfunction was also recently linked to the meta-organismal TMAO pathway 154–158. Here, dietary TMA-precursors (e.g. carnitine and choline) fuel microbial production of TMA, which is converted by liver FMO3 to TMAO, which circulates blood and is excreted by the kidneys. In animals and humans, blood TMAO increases with age 154–158, in relation to oxidative stress and vascular dysfunction 155,157,158; and neuroinflammation, cognitive decline 156 and dementia 159. In animals this occurs alongside gut dysbiosis, including overgrowth of proteobacteria and Desulfovibrio (a sulfur-reducing and TMA-producing genus), and increased FMO3 expression 158, while suppression of microbial TMA formation normalises vascular dysfunction 155,157. Ultimately, TMAO may suppress SIRT1/3 activity to promote oxidative stress, inflammation 160 and senescence 154.

On the other hand, there are likely reciprocal interactions. For instance, gastric acid secretion declines with age, while gastritis increases, which may promote maldigestion and B12 deficiency 161. Of potential relevance, acid-secreting parietal cells of the stomach are extremely densely populated with mitochondria, and their function is sensitive to redox and energy status 162,163. Gut oxidative stress may also slow intestinal motility to promote SIBO 164,165, and directly affect microbiome balance 166 and diversity 167. Note, SOD1-deficient mice also have reduced gut Akkermansia and NAM metabolism 146. Also, age-related gut microbiota change was dependent on inflammatory signalling and reversible with anti-TNF therapy 147.

The environment

Currently, there seem only minor associations between genetic variation and ageing, although a recent meta-analysis of genomic studies implicated those relating to CVD and heme metabolism; and thereby iron-based oxidative stress 168. Many environmental factors are associated with age-related disease and mortality. Accordingly, the reciprocal metabolic and microbial networks discussed above suggest vicious cycles which may be receptive to many environmental cues (e.g. diet, toxins, microbes, exercise, sleep, psych/stress, etc.). Our exposure to these factors throughout life constitute the exposome, which may accumulate as memories/scars in multiple bio-systems/structures to erode quality and induce hallmarks of ageing (e.g. redox 9, information 169 and extracellular matrix (ECM) theories 170); some of which may be readily reversible, others less so. Indeed, cells are equipped with mechanisms to repair and recycle damaged components, while altering the extracellular environment (e.g. ECM 170, blood plasma 171 or Cys redox 172) appears sufficient to rejuvenate old cells/tissues—so what about the external environment?

Diet is the natural source of metabolites for microbiome, metabolism and redox biology 19, with potential to influence age-related changes. Accordingly, in a cohort of Irish elderly (n=178, mean age 78), health status was related to gut microbiota, which was related to diet, where the most microbial and dietary diversity related to a low fat/high fibre pattern 173. Similarly, in a larger trial, adherence to a Mediterranean-style diet (i.e. NU-AGE diet) was associated with favourable modulation of the gut microbiome in relation to other health markers 174; and in another large study, higher Med-diet scores (and lower BMI) associated with less oxidised thiol redox in mid-age people 175. Such plant-rich diets might favourably modulate pathways discussed above (e.g. ↑ carbs/fibre, antiox, folate, polyamines; ↓ Met:Gly, carnitine, heme-iron, etc.). On the other hand, better nutritional status was associated with SIRT1 expression in older adults, independent of Med-diet scores 176. Also, high protein intake, and even more so plasma urea, was associated with markedly lower NAD levels in older adults (independent of age, gender, eGFR and calories), implicating amino acid catabolism in NAD decline 68; while in mice high protein intake upregulated liver mitochondrial protein content (incl. NAD transporter) and oxidative stress 177.

Whole plant foods (e.g. fruit, veg and grains) are dose-dependently associated with healthspan and longevity 178,179, and represent the foundation of many nutritional guidelines. Furthermore, dietary antioxidant capacity is also dose-dependently related to lower total, CVD and cancer mortality 15,16. Whole plant foods are especially rich in many antioxidants 180 and related phytochemicals 181 which can promote Nrf2 activity in cells (e.g. polyphenols, glucosinolates, etc.) 182; e.g. as redox cofactors 183, ROS-stimulants 184,185 or via other pathways (e.g. epigenetics 186, AMPK/SIRT1 105,109, etc.). While non-nutrient phytochemicals typically have limited absorption and rapid metabolism/clearance, brief Nrf2 stimulation has lasting effects on protein expression, which accumulates with repeat stimulation 187. Moreover, limited polyphenol absorption leads to accumulation in the gut, where microbial metabolism generates smaller secondary metabolites, which are more bioavailable and persist in circulation 188,189.

Modulation of the Nrf2 pathway can occur within hours after a single meal in young healthy humans: decreasing after a processed high-fat meal (at 5hrs 75%), but stimulated by addition of polyphenols (at 3hrs 250%) 190. Importantly, some longer trials with polyphenol-rich foods/extracts have improved redox status in older people (e.g. mean age 50–60 191–193 or 60–76 194–197; w/disease 192–194,197 and AMPK/SIRT/Nrf2 191,194,197), alongside various organ markers/functions (Table), suggesting the body remains responsive during ageing and disease. Similarly, in ageing animals, Nrf2/redox and organ function is improved by phenols (e.g. resveratrol 109, green tea catechins 198, cocoa epicatechin 199, curcumin 200, amla 201 and sesamol 202) and glucosinolates (e.g. sulforaphane 43,74,92); as well as by probiotics 203,204 and butyrate 205. Perhaps noteworthy, unlike GSH-precursors 39, polyphenols also improved the ECM (e.g. arteries 200, muscle 206 and skin 207).

Another important component of a Mediterranean diet is fish, which itself generally has positive associations with healthspan, in relation to omega-3s 208–210; and while supplement trials seem less consistent, this may involve several confounding factors obscuring benefit 211,212. Further, a meta-analysis of RCTs suggests omega-3s can also improve general blood redox markers 213; and a recent trial with EPA/DHA (2700mg/day) showed robust induction of Nrf2 in PBMCs in T2D 214. Accordingly, while polyunsaturated fatty acids are highly susceptible to oxidation, especially omega-3s, they generate specific products (e.g. J3-isoprostanes 215, 4-HHE 216 and oxygenase-derived resolvin D1 217,218) which can induce Nrf2 antioxidant responses. In mice, fish oil increased DHA/4-HHE and HO-1 in multiple organs 216; and a genetically increased omega-3/6 ratio induced Nrf2/HO-1 and lowered UVB-induced skin oxidative stress, inflammation and carcinogenesis 219,220. Therefore omega-3 status (and omega-6/3 ratios) may influence redox homeostasis and age-related disease; with a notable abundance of DHA in brain and retina 221.

On the other hand, toxins can disrupt redox homeostasis, as with TMAO above. Note, deep-sea fish also contain particularly high levels of preformed TMAO 222 (e.g. cod vs. salmon 223). Moreover, our oceans are now polluted and the positive health associations with marine omega-3s may be offset by concomitant toxin/PCB intake 209,210. Another important factor may be lifelong accumulation of redox-active metals 9. For instance, in a representative Mediterranean population (n=815), plasma oxidative stress correlated age, heme-iron intake (from meat and fish) and transferrin saturation (i.e. iron status) 224. Further, several toxic metals (e.g. Pb, Cd and Hg) have also been found to accumulate with age in specific organs, where they may promote oxidative stress and disease 225–228. And regarding air pollution, chronic or high exposure to urban particulate matter can inhibit Nrf2 activity 229; as can cigarette smoke via oxidation products 230 and post-translational modifications (prevented by resveratrol) 231.

Advanced glycation end-products (AGEs, aka. glycotoxins) also accumulate with age and disease in relation to inflammation and oxidative stress 232. AGEs can form endogenously and exogenously in food. In particular, heat-processed animal and high-fat foods are rich in AGEs 233, which have partial absorption (~10–30%) and affect host and gut microbiome 234. In healthy US adults, there are correlations between diet and blood AGEs, inflammatory and oxidative markers 235–237; all of which were lowered simply by switching to wet cooking methods 236,237. Further, in a 1-year RCT on older people with MetS (n=138, mean age 61), a low-AGE diet also greatly normalised insulin sensitivity 238. Moreover, the ability of calorie restriction to extend animal health and lifespan has also been linked to lower dietary AGEs, with favourable modulation of GSH redox and oxidative stress 239. Note, AGEs may especially accumulate on long-lived macromolecules (e.g. ECM, esp. cartilage and bone) 232, where they can mediate various deleterious effects 170, including inhibition of SIRT1/Nrf2 activity 101,237,238,240 (prevented by curcumin 241). In animal models, at least arterial AGE accumulation can be quickly reversed (e.g. curcumin 200 and spermidine 242).

Exercise is also associated with healthspan, which may involve hormesis and Nrf2 activity 83,84. Accordingly, an active lifestyle was associated with preserved Nrf2 activity in skeletal muscle of older people 75. Further trials have shown that acute exercise-induced Nrf2 signalling is impaired in PBMCs 76, while aerobic training improved resistance to oxidative stress in forearm 243. Similarly, in old animals, moderate exercise training improved SIRT1/redox in skeletal muscle 244 and Nrf2/redox in heart 245. Note however, exercise and weight loss did not improve leaky gut markers in older people 148, or those with cardiometabolic disease 150, while polyphenols did in MetS 246 and T2D 197. Also, while there is suggestion that (compared to antioxidant vitamins) phytochemicals may not interfere with exercise adaptations 247, or even support them 83, they still can in pharmacologic dose/form in young/aged humans (e.g. epicatechin 248 and resveratrol 249).

Evolution vs. entropy

The biochemistry of life ultimately follows the physics of the universe. In particular, the 2nd law of thermodynamics describes how all isolated systems spontaneously evolve toward equilibrium, with dissipation of all energy—i.e. a state of maximum entropy (S, as joules per kelvin). As open systems, organisms continually take in food/energy, while expelling entropic waste, enabling maintenance of their internal order (i.e. non-equilibrium) indefinitely 250,251. More specifically, aerobic life harnesses non-equilibrium electrochemistry to drive bioenergetics, while coupling the Gibbs free energy to self-organisation (e.g. non-equilibrium biochemistry, biosynthesis and barriers). Order is further protected by homeostatic/defence pathways which commit resources toward clearance of molecular damage, dysfunctional cells and exogenous threats (e.g. toxins and infections).

However, this gradually fails with ageing, which is considered a state of increasing disorder analogous to entropy 251–256 (i.e. energy and information entropy 169,256); characterised by accumulation of stochastic modifications to macromolecules (e.g. via glycation 170,253, DNA methylation 169,256, etc.) and loss of non-equilibrium (e.g. via chemical potentials/gradients and barriers 9), with increasing cell/tissue heterogeneity and dysfunction, and susceptibility to disease and death (i.e. ‘…a rush toward equilibrium’ 251). In essence, ageing may involve a gradual loss of molecular fidelity/clarity due to loss of thermodynamic efficacy (i.e. energy–order) 254. Notably, ageing rate varies widely between species, and in some may be negligible, suggesting the extent of self-maintenance may be determined by ecological niche and evolutionary pressures 250,252,255. As such, in most animals, through reproductive maturation (and parenthood) there may be decreasing selective pressure (i.e. evolutionary value) on such homeostatic/repair systems, thus constraining their function.

On the other hand, while evolution (past environment) shapes the genes which ultimately determine capacity for longevity 252, the present environment still constantly modulates physiology. Accordingly, various redox modulators have rejuvenated age and disease-related organ dysfunction in older humans, animals and cells (Table). Therefore, our current environment may be limiting, and even late optimisation, via removal of disruptive/toxic stressors and re-engagement of homeostatic pathways, may enable some restoration of redox biology, thermodynamic efficacy and physiological function. This may improve/square healthspan and disease-limited lifespan, to more fully express genomic potential; beyond which we may be particularly limited in our ability to clear accumulative extracellular changes and ultimately to maintain aesthetics and function 170,232,253.

Perspective

In summary, much age-related physiological decline seems tied to oxidation of redox status (i.e. oxidative ageing, or ‘oxid-ageing’), in relation to various proximate metabolic and signalling changes, which link to microbial and environmental factors (e.g. diet/lifestyle), and ultimate evolutionary constraints. Importantly, many biological changes seem to remain amenable throughout ageing and disease, where various redox modulators have rejuvenated age-related organ dysfunction. Therefore redox status may be a continuous determinate of physiological function, and regulator of ageing rate and reversibility, which links past and present environment to thermodynamic efficacy and healthspan.

The flexibility of ageing might have some provocative social implications. For instance, to what extent does our notion of ‘normal ageing’ encompass preventable and even reversible functional decline and disease, and therefore suboptimal, unhealthy ageing? Is age-related functional decline an early manifestation of subclinical disease and prodromal to age-related disease? Are the top causes of morbidity and mortality an inevitable consequence of living longer, or environmental mismatch—i.e. our success or failure? Is unhealthy ageing coupled to an unhealthy planet? Moreover, recently the fundamental biological features of spaceflight were revealed, with many notable similarities to ageing, including chronic oxidative stress 257. Therefore optimising our redox biology may be important not just for health on this planet, but for enabling travel to others.

In memory of JM

Resources

References

1.           Singer, L. et al. Trends in multimorbidity, complex multimorbidity and multiple functional limitations in the ageing population of England, 2002-2015. J. comorbidity 9, 2235042X19872030 (2019).

2.           King, D. E., Xiang, J. & Pilkerton, C. S. Multimorbidity Trends in United States Adults, 1988-2014. J. Am. Board Fam. Med. 31, 503–513 (2018).

3.           Zheng, Z. et al. Risk factors of critical & mortal COVID-19 cases: A systematic literature review and meta-analysis. J. Infect. 81, e16–e25 (2020).

4.           Hayflick, L. The greatest risk factor for the leading cause of death is ignored. Biogerontology (2020). doi:10.1007/s10522-020-09901-y

5.           Harman, D. Aging: a theory based on free radical and radiation chemistry. J. Gerontol. 11, 298–300 (1956).

6.           Harman, D. Origin and evolution of the free radical theory of aging: a brief personal history, 1954–2009. Biogerontology 10, 773–81 (2009).

7.           Golubev, A., Hanson, A. D. & Gladyshev, V. N. A Tale of Two Concepts: Harmonizing the Free Radical and Antagonistic Pleiotropy Theories of Aging. Antioxid. Redox Signal. 29, 1003–1017 (2018).

8.           Ristow, M. & Schmeisser, K. Mitohormesis: Promoting Health and Lifespan by Increased Levels of Reactive Oxygen Species (ROS). Dose. Response. 12, 288–341 (2014).

9.           Go, Y.-M. & Jones, D. P. Redox theory of aging: implications for health and disease. Clin. Sci. (Lond). 131, 1669–1688 (2017).

10.        Zhang, H., Davies, K. J. A. & Forman, H. J. Oxidative stress response and Nrf2 signaling in aging. Free Radic. Biol. Med. 88, 314–336 (2015).

11.        Silva-Palacios, A., Ostolga-Chavarría, M., Zazueta, C. & Königsberg, M. Nrf2: Molecular and epigenetic regulation during aging. Ageing Res. Rev. 47, 31–40 (2018).

12.        Barreiro, E. Role of Protein Carbonylation in Skeletal Muscle Mass Loss Associated with Chronic Conditions. Proteomes 4, 18 (2016).

13.        El Assar, M. et al. Frailty Is Associated With Lower Expression of Genes Involved in Cellular Response to Stress: Results From the Toledo Study for Healthy Aging. J. Am. Med. Dir. Assoc. 18, 734.e1-734.e7 (2017).

14.        Belenguer-Varea, Á. et al. Oxidative stress and exceptional human longevity: Systematic review. Free Radic. Biol. Med. 149, 51–63 (2020).

15.        Jayedi, A., Rashidy-Pour, A., Parohan, M., Zargar, M. S. & Shab-Bidar, S. Dietary Antioxidants, Circulating Antioxidant Concentrations, Total Antioxidant Capacity, and Risk of All-Cause Mortality: A Systematic Review and Dose-Response Meta-Analysis of Prospective Observational Studies. Adv. Nutr. 9, 701–716 (2018).

16.        Parohan, M. et al. Dietary total antioxidant capacity and mortality from all causes, cardiovascular disease and cancer: a systematic review and dose-response meta-analysis of prospective cohort studies. Eur. J. Nutr. 58, 2175–2189 (2019).

17.        Bjelakovic, G., Nikolova, D. & Gluud, C. Antioxidant supplements and mortality. Curr. Opin. Clin. Nutr. Metab. Care 17, 40–4 (2014).

18.        Jones, D. P. & Sies, H. The Redox Code. Antioxid. Redox Signal. 23, 734–46 (2015).

19.        Dennis, K. K., Go, Y.-M. & Jones, D. P. Redox Systems Biology of Nutrition and Oxidative Stress. J. Nutr. 149, 553–565 (2019).

20.        Thannickal, V. J. Oxygen in the evolution of complex life and the price we pay. Am. J. Respir. Cell Mol. Biol. 40, 507–10 (2009).

21.        Gacesa, R., Dunlap, W. C., Barlow, D. J., Laskowski, R. A. & Long, P. F. Rising levels of atmospheric oxygen and evolution of Nrf2. Sci. Rep. 6, 27740 (2016).

22.        Go, Y.-M. & Jones, D. P. The redox proteome. J. Biol. Chem. 288, 26512–20 (2013).

23.        Hansen, J. M., Jones, D. P. & Harris, C. The Redox Theory of Development. Antioxid. Redox Signal. 32, 715–740 (2020).

24.        Paiva, C. N. & Bozza, M. T. Are reactive oxygen species always detrimental to pathogens? Antioxid. Redox Signal. 20, 1000–37 (2014).

25.        Martinvalet, D. Mitochondrial Entry of Cytotoxic Proteases: A New Insight into the Granzyme B Cell Death Pathway. Oxid. Med. Cell. Longev. 2019, 9165214 (2019).

26.        Go, Y.-M. et al. Selective targeting of the cysteine proteome by thioredoxin and glutathione redox systems. Mol. Cell. Proteomics 12, 3285–96 (2013).

27.        Mailloux, R. J., Jin, X. & Willmore, W. G. Redox regulation of mitochondrial function with emphasis on cysteine oxidation reactions. Redox Biol. 2, 123–39 (2013).

28.        Dardalhon, M. et al. Redox-sensitive YFP sensors monitor dynamic nuclear and cytosolic glutathione redox changes. Free Radic. Biol. Med. 52, 2254–65 (2012).

29.        Franco, R. & Cidlowski, J. a. Glutathione efflux and cell death. Antioxid. Redox Signal. 17, 1694–713 (2012).

30.        Rebrin, I. & Sohal, R. S. Pro-oxidant shift in glutathione redox state during aging. Adv. Drug Deliv. Rev. 60, 1545–52 (2008).

31.        Maher, P. The effects of stress and aging on glutathione metabolism. Ageing Res. Rev. 4, 288–314 (2005).

32.        Jones, D. P., Mody, V. C., Carlson, J. L., Lynn, M. J. & Sternberg, P. Redox analysis of human plasma allows separation of pro-oxidant events of aging from decline in antioxidant defenses. Free Radic. Biol. Med. 33, 1290–300 (2002).

33.        Sekhar, R. V et al. Deficient synthesis of glutathione underlies oxidative stress in aging and can be corrected by dietary cysteine and glycine supplementation. Am. J. Clin. Nutr. 94, 847–53 (2011).

34.        Nguyen, D., Hsu, J. W., Jahoor, F. & Sekhar, R. V. Effect of increasing glutathione with cysteine and glycine supplementation on mitochondrial fuel oxidation, insulin sensitivity, and body composition in older HIV-infected patients. J. Clin. Endocrinol. Metab. 99, 169–77 (2014).

35.        Nguyen, D., Samson, S. L., Reddy, V. T., Gonzalez, E. V & Sekhar, R. V. Impaired mitochondrial fatty acid oxidation and insulin resistance in aging: novel protective role of glutathione. Aging Cell 12, 415–25 (2013).

36.        Suh, J. H., Wang, H., Liu, R.-M., Liu, J. & Hagen, T. M. (R)-alpha-lipoic acid reverses the age-related loss in GSH redox status in post-mitotic tissues: evidence for increased cysteine requirement for GSH synthesis. Arch. Biochem. Biophys. 423, 126–35 (2004).

37.        Toroser, D. & Sohal, R. S. Age-associated perturbations in glutathione synthesis in mouse liver. Biochem. J. 405, 583–9 (2007).

38.        Dolopikou, C. F. et al. Acute nicotinamide riboside supplementation improves redox homeostasis and exercise performance in old individuals: a double-blind cross-over study. Eur. J. Nutr. 103, 1357–1366 (2019).

39.        Cieslik, K. A. et al. Improved Cardiovascular Function in Old Mice After N-Acetyl Cysteine and Glycine Supplemented Diet: Inflammation and Mitochondrial Factors. J. Gerontol. A. Biol. Sci. Med. Sci. 73, 1167–1177 (2018).

40.        Robillard, J. M., Gordon, G. R., Choi, H. B., Christie, B. R. & MacVicar, B. A. Glutathione restores the mechanism of synaptic plasticity in aged mice to that of the adult. PLoS One 6, e20676 (2011).

41.        Garg, G., Singh, S., Singh, A. K. & Rizvi, S. I. N-acetyl-l-cysteine attenuates oxidative damage and neurodegeneration in rat brain during aging. Can. J. Physiol. Pharmacol. 96, 1189–1196 (2018).

42.        Amatore, D. et al. Glutathione increase by the n-butanoyl glutathione derivative (GSH-C4) inhibits viral replication and induces a predominant Th1 immune profile in old mice infected with influenza virus. FASEB bioAdvances 1, 296–305 (2019).

43.        Kim, H.-J., Barajas, B., Wang, M. & Nel, A. E. Nrf2 activation by sulforaphane restores the age-related decrease of T(H)1 immunity: role of dendritic cells. J. Allergy Clin. Immunol. 121, 1255-1261.e7 (2008).

44.        Sekhar, R. V, Liu, C. W. & Rice, S. Increasing glutathione concentrations with cysteine and glycine supplementation lowers inflammation in HIV patients. AIDS 29, 1899–900 (2015).

45.        De Flora, S., Grassi, C. & Carati, L. Attenuation of influenza-like symptomatology and improvement of cell-mediated immunity with long-term N-acetylcysteine treatment. Eur. Respir. J. 10, 1535–41 (1997).

46.        McCarty, M. F. & DiNicolantonio, J. J. An increased need for dietary cysteine in support of glutathione synthesis may underlie the increased risk for mortality associated with low protein intake in the elderly. Age (Omaha). 37, (2015).

47.        Currais, A. & Maher, P. Functional consequences of age-dependent changes in glutathione status in the brain. Antioxid. Redox Signal. 19, 813–22 (2013).

48.        Ballatori, N. et al. Glutathione dysregulation and the etiology and progression of human diseases. Biol. Chem. 390, 191–214 (2009).

49.        Kumar, P. et al. Supplementing Glycine and N-acetylcysteine (GlyNAC) in Aging HIV Patients Improves Oxidative Stress, Mitochondrial Dysfunction, Inflammation, Endothelial Dysfunction, Insulin Resistance, Genotoxicity, Strength, and Cognition: Results of an Open-Label Clin. Biomedicines 8, (2020).

50.        Kumar, P. et al. Glycine and N-acetylcysteine (GlyNAC) supplementation in older adults improves glutathione deficiency, oxidative stress, mitochondrial dysfunction, inflammation, insulin resistance, endothelial dysfunction, genotoxicity, muscle strength, and cognition: Re. Clin. Transl. Med. 11, e372 (2021).

51.        Wu, D., Meydani, S. N., Sastre, J., Hayek, M. & Meydani, M. In vitro glutathione supplementation enhances interleukin-2 production and mitogenic response of peripheral blood mononuclear cells from young and old subjects. J. Nutr. 124, 655–63 (1994).

52.        Bouamama, S., Merzouk, H., Medjdoub, A., Merzouk-Saidi, A. & Merzouk, S. A. Effects of exogenous vitamins A, C, and E and NADH supplementation on proliferation, cytokines release, and cell redox status of lymphocytes from healthy aged subjects. Appl. Physiol. Nutr. Metab. 42, 579–587 (2017).

53.        Carilho Torrao, R. B. D., Dias, I. H., Bennett, S. J., Dunston, C. R. & Griffiths, H. R. Healthy ageing and depletion of intracellular glutathione influences T cell membrane thioredoxin-1 levels and cytokine secretion. Chem. Cent. J. 7, 150 (2013).

54.        Hajjar, I. et al. Oxidative stress predicts cognitive decline with aging in healthy adults: an observational study. J. Neuroinflammation 15, 17 (2018).

55.        Zhang, Y. et al. Decreased Brain Levels of Vitamin B12 in Aging, Autism and Schizophrenia. PLoS One 11, e0146797 (2016).

56.        Guest, J. et al. Novel relationships between B12, folate and markers of inflammation, oxidative stress and NAD(H) levels, systemically and in the CNS of a healthy human cohort. Nutr. Neurosci. 18, 355–64 (2015).

57.        Aylett, S.-B. et al. Levels of 5-methyltetrahydrofolate and ascorbic acid in cerebrospinal fluid are correlated: implications for the accelerated degradation of folate by reactive oxygen species. Neurochem. Int. 63, 750–5 (2013).

58.        Canfield, C.-A. & Bradshaw, P. C. Amino acids in the regulation of aging and aging-related diseases. Transl. Med. Aging 3, 70–89 (2019).

59.        Hashizume, O. et al. Epigenetic regulation of the nuclear-coded GCAT and SHMT2 genes confers human age-associated mitochondrial respiration defects. Sci. Rep. 5, 10434 (2015).

60.        Mardinoglu, A. et al. Personal model-assisted identification of NAD+ and glutathione metabolism as intervention target in NAFLD. Mol. Syst. Biol. 13, 916 (2017).

61.        Xiao, W., Wang, R.-S., Handy, D. E. & Loscalzo, J. NAD(H) and NADP(H) Redox Couples and Cellular Energy Metabolism. Antioxid. Redox Signal. 28, 251–272 (2018).

62.        Ducker, G. S. & Rabinowitz, J. D. One-Carbon Metabolism in Health and Disease. Cell Metab. 25, 27–42 (2017).

63.        Bradshaw, P. C. Cytoplasmic and Mitochondrial NADPH-Coupled Redox Systems in the Regulation of Aging. Nutrients 11, (2019).

64.        Clement, J., Wong, M., Poljak, A., Sachdev, P. & Braidy, N. The Plasma NAD+ Metabolome Is Dysregulated in ‘Normal’ Aging. Rejuvenation Res. 22, 121–130 (2019).

65.        Chini, C. C. S., Tarragó, M. G. & Chini, E. N. NAD and the aging process: Role in life, death and everything in between. Mol. Cell. Endocrinol. 455, 62–74 (2017).

66.        Fang, E. F. et al. NAD+ in Aging: Molecular Mechanisms and Translational Implications. Trends Mol. Med. 23, 899–916 (2017).

67.        Massudi, H. et al. Age-associated changes in oxidative stress and NAD+ metabolism in human tissue. PLoS One 7, e42357 (2012).

68.        Seyedsadjadi, N. et al. High protein intake is associated with low plasma NAD+ levels in a healthy human cohort. PLoS One 13, e0201968 (2018).

69.        Wu, S. & Zhang, R. CD38-expressing macrophages drive age-related NAD+ decline. Nat. Metab. 2, 1186–1187 (2020).

70.        Pedley, A. M. & Benkovic, S. J. A New View into the Regulation of Purine Metabolism: The Purinosome. Trends Biochem. Sci. 42, 141–154 (2017).

71.        Pissios, P. Nicotinamide N -Methyltransferase: More Than a Vitamin B3 Clearance Enzyme. Trends Endocrinol. Metab. 28, 340–353 (2017).

72.        Sternak, M. et al. Differential involvement of IL-6 in the early and late phase of 1-methylnicotinamide (MNA) release in Concanavalin A-induced hepatitis. Int. Immunopharmacol. 28, 105–114 (2015).

73.        Hamilton, R. T., Walsh, M. E. & Van Remmen, H. Mouse Models of Oxidative Stress Indicate a Role for Modulating Healthy Aging. J. Clin. Exp. Pathol. Suppl 4, (2012).

74.        Kubo, E., Chhunchha, B., Singh, P., Sasaki, H. & Singh, D. P. Sulforaphane reactivates cellular antioxidant defense by inducing Nrf2/ARE/Prdx6 activity during aging and oxidative stress. Sci. Rep. 7, 14130 (2017).

75.        Safdar, A., DeBeer, J. & Tarnopolsky, M. A. Dysfunctional Nrf2-Keap1 redox signaling in skeletal muscle of the sedentary old. Free Radic. Biol. Med. 49, 1487–93 (2010).

76.        Done, A. J., Gage, M. J., Nieto, N. C. & Traustadóttir, T. Exercise-induced Nrf2-signaling is impaired in aging. Free Radic. Biol. Med. 96, 130–8 (2016).

77.        Angulo, J. et al. Short-term pharmacological activation of Nrf2 ameliorates vascular dysfunction in aged rats and in pathological human vasculature. A potential target for therapeutic intervention. Redox Biol. 26, 101271 (2019).

78.        Zhang, H., Zhou, L., Davies, K. J. A. & Forman, H. J. Silencing Bach1 alters aging-related changes in the expression of Nrf2-regulated genes in primary human bronchial epithelial cells. Arch. Biochem. Biophys. 672, 108074 (2019).

79.        Scutt, G. et al. The Association of a Single-Nucleotide Polymorphism in the Nuclear Factor (Erythroid-Derived 2)-Like 2 Gene With Adverse Drug Reactions, Multimorbidity, and Frailty in Older People. J. Gerontol. A. Biol. Sci. Med. Sci. 75, 1050–1057 (2020).

80.        Gao, Y., Yan, Y. & Huang, T. Human agerelated cataracts: epigenetic suppression of the nuclear factor erythroid 2related factor 2mediated antioxidant system. Mol. Med. Rep. 11, 1442–7 (2015).

81.        Suh, J. H. et al. Decline in transcriptional activity of Nrf2 causes age-related loss of glutathione synthesis, which is reversible with lipoic acid. Proc. Natl. Acad. Sci. U. S. A. 101, 3381–6 (2004).

82.        Shenvi, S. V, Smith, E. & Hagen, T. M. Identification of age-specific Nrf2 binding to a novel antioxidant response element locus in the Gclc promoter: a compensatory means for the loss of glutathione synthetic capacity in the aging rat liver? Aging Cell 11, 297–304 (2012).

83.        Done, A. J. & Traustadóttir, T. Nrf2 mediates redox adaptations to exercise. Redox Biol. 10, 191–199 (2016).

84.        Merry, T. L. & Ristow, M. Nuclear factor erythroid-derived 2-like 2 (NFE2L2, Nrf2) mediates exercise-induced mitochondrial biogenesis and the anti-oxidant response in mice. J. Physiol. 594, 5195–207 (2016).

85.        Lin, T.-Y., Cantley, L. C. & DeNicola, G. M. NRF2 Rewires Cellular Metabolism to Support the Antioxidant Response. in A Master Regulator of Oxidative Stress - The Transcription Factor Nrf2 21–48 (InTech, 2016). doi:10.5772/65141

86.        Hayes, J. D. & Dinkova-Kostova, A. T. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem. Sci. 39, 199–218 (2014).

87.        Dinkova-Kostova, A. T. & Abramov, A. Y. The emerging role of Nrf2 in mitochondrial function. Free Radic. Biol. Med. 88, 179–88 (2015).

88.        Mastrocola, R. AGEs and neurodegeneration: the Nrf2/glyoxalase-1 interaction. Oncotarget 8, 5645–5646 (2017).

89.        Kubben, N. et al. Repression of the antioxidant NRF2 pathway in premature aging. Cell 165, 1361 (2016).

90.        Rojo, A. I. et al. NRF2 deficiency replicates transcriptomic changes in Alzheimer’s patients and worsens APP and TAU pathology. Redox Biol. 13, 444–451 (2017).

91.        Huang, D.-D. et al. Nrf2 deficiency exacerbates frailty and sarcopenia by impairing skeletal muscle mitochondrial biogenesis and dynamics in an age-dependent manner. Exp. Gerontol. 119, 61–73 (2019).

92.        Townsend, B. E. & Johnson, R. W. Sulforaphane induces Nrf2 target genes and attenuates inflammatory gene expression in microglia from brain of young adult and aged mice. Exp. Gerontol. 73, 42–8 (2016).

93.        Ray, S. et al. A Role for Nrf2 Expression in Defining the Aging of Hippocampal Neural Stem Cells. Cell Transplant. 27, 589–606 (2018).

94.        Park, G. & Oh, M. S. Acceleration of heat shock-induced collagen breakdown in human dermal fibroblasts with knockdown of NF-E2-related factor 2. BMB Rep. 48, 467–72 (2015).

95.        Pomatto, L. C. D. et al. Deletion of Nrf2 shortens lifespan in C57BL6/J male mice but does not alter the health and survival benefits of caloric restriction. Free Radic. Biol. Med. 152, 650–658 (2020).

96.        Schmidlin, C. J., Dodson, M. B., Madhavan, L. & Zhang, D. D. Redox regulation by NRF2 in aging and disease. Free Radic. Biol. Med. 134, 702–707 (2019).

97.        Bruns, D. R. et al. Nrf2 Signaling and the Slowed Aging Phenotype: Evidence from Long-Lived Models. Oxid. Med. Cell. Longev. 2015, 732596 (2015).

98.        Nóbrega-Pereira, S. et al. G6PD protects from oxidative damage and improves healthspan in mice. Nat. Commun. 7, 10894 (2016).

99.        Pickering, A. M., Lehr, M., Gendron, C. M., Pletcher, S. D. & Miller, R. A. Mitochondrial thioredoxin reductase 2 is elevated in long-lived primate as well as rodent species and extends fly mean lifespan. Aging Cell 16, 683–692 (2017).

100.      Zhou, L., Zhang, H., Davies, K. J. A. & Forman, H. J. Aging-related decline in the induction of Nrf2-regulated antioxidant genes in human bronchial epithelial cells. Redox Biol. 14, 35–40 (2018).

101.      Periyasamy, P. & Shinohara, T. Age-related cataracts: Role of unfolded protein response, Ca2+ mobilization, epigenetic DNA modifications, and loss of Nrf2/Keap1 dependent cytoprotection. Prog. Retin. Eye Res. 60, 1–19 (2017).

102.      Salminen, A. & Kaarniranta, K. AMP-activated protein kinase (AMPK) controls the aging process via an integrated signaling network. Ageing Res. Rev. 11, 230–41 (2012).

103.      Shi, S., Lei, S., Tang, C., Wang, K. & Xia, Z. Melatonin attenuates acute kidney ischemia/reperfusion injury in diabetic rats by activation of the SIRT1/Nrf2/HO-1 signaling pathway. Biosci. Rep. 39, (2019).

104.      Ding, Y.-W. et al. SIRT1 exerts protective effects against paraquat-induced injury in mouse type II alveolar epithelial cells by deacetylating NRF2 in vitro. Int. J. Mol. Med. 37, 1049–58 (2016).

105.      Huang, K. et al. Polydatin promotes Nrf2-ARE anti-oxidative pathway through activating Sirt1 to resist AGEs-induced upregulation of fibronetin and transforming growth factor-β1 in rat glomerular messangial cells. Mol. Cell. Endocrinol. 399, 178–89 (2015).

106.      Zhang, J. et al. SIRT2, ERK and Nrf2 Mediate NAD+ Treatment-Induced Increase in the Antioxidant Capacity of PC12 Cells Under Basal Conditions. Front. Mol. Neurosci. 12, 108 (2019).

107.      Pan, H. et al. SIRT6 safeguards human mesenchymal stem cells from oxidative stress by coactivating NRF2. Cell Res. 26, 190–205 (2016).

108.      Huang, K., Gao, X. & Wei, W. The crosstalk between Sirt1 and Keap1/Nrf2/ARE anti-oxidative pathway forms a positive feedback loop to inhibit FN and TGF-β1 expressions in rat glomerular mesangial cells. Exp. Cell Res. 361, 63–72 (2017).

109.      Kim, E. N. et al. Resveratrol, an Nrf2 activator, ameliorates aging-related progressive renal injury. Aging (Albany. NY). 10, 83–99 (2018).

110.      Yoon, D. S., Choi, Y. & Lee, J. W. Cellular localization of NRF2 determines the self-renewal and osteogenic differentiation potential of human MSCs via the P53-SIRT1 axis. Cell Death Dis. 7, e2093 (2016).

111.      Kulkarni, S. R. et al. Fasting Induces Nuclear Factor E2-Related Factor 2 and ATP-Binding Cassette Transporters via Protein Kinase A and Sirtuin-1 in Mouse and Human. Antioxid. Redox Signal. 20, 15–30 (2014).

112.      Shrishrimal, S. et al. Manganese porphyrin, MnTE-2-PyP, treatment protects the prostate from radiation-induced fibrosis (RIF) by activating the NRF2 signaling pathway and enhancing SOD2 and sirtuin activity. Free Radic. Biol. Med. 152, 255–270 (2020).

113.      Francesco, A. Di et al. NQO1 protects obese mice through improvements in glucose and lipid metabolism. npj Aging Mech. Dis. 2020 61 6, 1–18 (2020).

114.      Ding, L. et al. Nrf2 exerts mixed inflammation and glucose metabolism regulatory effects on murine RAW264.7 macrophages. Int. Immunopharmacol. 71, 198–204 (2019).

115.      Zocchi, E., Franco, L., Guida, L., Calder, L. & De Flora, A. Self-aggregation of purified and membrane-bound erythrocyte CD38 induces extensive decrease of its ADP-ribosyl cyclase activity. FEBS Lett. 359, 35–40 (1995).

116.      Fulop, G. A. et al. Nrf2 deficiency in aged mice exacerbates cellular senescence promoting cerebrovascular inflammation. GeroScience 40, 513–521 (2018).

117.      Fang, J. et al. Melatonin prevents senescence of canine adipose-derived mesenchymal stem cells through activating NRF2 and inhibiting ER stress. Aging (Albany. NY). 10, 2954–2972 (2018).

118.      Joo, M. S. et al. AMPK Facilitates Nuclear Accumulation of Nrf2 by Phosphorylating at Serine 550. Mol. Cell. Biol. 36, 1931–42 (2016).

119.      Fischhuber, K., Matzinger, M. & Heiss, E. H. AMPK Enhances Transcription of Selected Nrf2 Target Genes via Negative Regulation of Bach1. Front. cell Dev. Biol. 8, 628 (2020).

120.      Shao, D. et al. A redox-dependent mechanism for regulation of AMPK activation by Thioredoxin1 during energy starvation. Cell Metab. 19, 232–45 (2014).

121.      Sid, V. et al. Folic acid supplementation during high-fat diet feeding restores AMPK activation via an AMP-LKB1-dependent mechanism. Am. J. Physiol. Regul. Integr. Comp. Physiol. 309, R1215-25 (2015).

122.      Weng, Q. et al. Folate Metabolism Regulates Oligodendrocyte Survival and Differentiation by Modulating AMPKα Activity. Sci. Rep. 7, 1705 (2017).

123.      Yang, H. et al. Dysregulation of glutathione synthesis during cholestasis in mice: molecular mechanisms and therapeutic implications. Hepatology 49, 1982–91 (2009).

124.      Madeo, F., Bauer, M. A., Carmona-Gutierrez, D. & Kroemer, G. Spermidine: a physiological autophagy inducer acting as an anti-aging vitamin in humans? Autophagy 15, 165–168 (2019).

125.      Liu, P. et al. Spermidine Confers Liver Protection by Enhancing NRF2 Signaling Through a MAP1S-Mediated Noncanonical Mechanism. Hepatology 70, 372–388 (2019).

126.      Scholtens, R. M., van Munster, B. C., van Kempen, M. F. & de Rooij, S. E. J. A. Physiological melatonin levels in healthy older people: A systematic review. J. Psychosom. Res. 86, 20–7 (2016).

127.      Hardeland, R. Aging, Melatonin, and the Pro- and Anti-Inflammatory Networks. Int. J. Mol. Sci. 20, (2019).

128.      Perridon, B. W., Leuvenink, H. G. D., Hillebrands, J. L., Van Goor, H. & Bos, E. M. The role of hydrogen sulfide in aging and age-related pathologies. Aging (Albany. NY). 8, 2264–2289 (2016).

129.      Das, A. et al. Impairment of an Endothelial NAD+-H2S Signaling Network Is a Reversible Cause of Vascular Aging. Cell 173, 74-89.e20 (2018).

130.      Lee, H. J. et al. Hydrogen sulfide ameliorates aging-associated changes in the kidney. GeroScience 40, 163–176 (2018).

131.      Hine, C., Zhu, Y., Hollenberg, A. N. & Mitchell, J. R. Dietary and Endocrine Regulation of Endogenous Hydrogen Sulfide Production: Implications for Longevity. Antioxid. Redox Signal. 28, 1483–1502 (2018).

132.      Sen, U., Mishra, P. K., Tyagi, N. & Tyagi, S. C. Homocysteine to hydrogen sulfide or hypertension. Cell Biochem. Biophys. 57, 49–58 (2010).

133.      Solomon, L. R. Functional cobalamin (vitamin B12) deficiency: role of advanced age and disorders associated with increased oxidative stress. Eur. J. Clin. Nutr. 69, 687–92 (2015).

134.      Muratore, C. R. et al. Age-dependent decrease and alternative splicing of methionine synthase mRNA in human cerebral cortex and an accelerated decrease in autism. PLoS One 8, e56927 (2013).

135.      Maddocks, O. D. K., Labuschagne, C. F., Adams, P. D. & Vousden, K. H. Serine Metabolism Supports the Methionine Cycle and DNA/RNA Methylation through De Novo ATP Synthesis in Cancer Cells. Mol. Cell 61, 210 (2016).

136.      Schrier, M., Rose, N., Zhang, Y., Trivedi, M. & Deth, R. The role of Nrf2 in redox-dependent cobalamin processing. Free Radic. Biol. Med. 128, S92–S93 (2018).

137.      Schrier, M. S., Trivedi, M. S. & Deth, R. C. Redox-related epigenetic mechanisms in glioblastoma: Nuclear factor (erythroid-derived 2)-like 2, cobalamin, and dopamine receptor subtype 4. Front. Oncol. 7, 1–14 (2017).

138.      Sbodio, J. I., Snyder, S. H. & Paul, B. D. Regulators of the transsulfuration pathway. Br. J. Pharmacol. 176, 583–593 (2019).

139.      Gopu, V. et al. An accurate aging clock developed from the largest dataset of microbial and human gene expression reveals molecular mechanisms of aging. bioRxiv (2020). doi:10.1101/2020.09.17.301887

140.      Elphick, D. A. et al. Small bowel bacterial overgrowth in symptomatic older people: Can it be diagnosed earlier? Gerontology 51, 396–401 (2005).

141.      Chen, B., Kim, J. J.-W., Zhang, Y., Du, L. & Dai, N. Prevalence and predictors of small intestinal bacterial overgrowth in irritable bowel syndrome: a systematic review and meta-analysis. J. Gastroenterol. 2018 537 53, 807–818 (2018).

142.      Mardinoglu, A. et al. The gut microbiota modulates host amino acid and glutathione metabolism in mice. Mol. Syst. Biol. 11, 834 (2015).

143.      Mardinoglu, A. et al. An Integrated Understanding of the Rapid Metabolic Benefits of a Carbohydrate-Restricted Diet on Hepatic Steatosis in Humans. Cell Metab. 27, 559-571.e5 (2018).

144.      Chan, Y.-M., Aufreiter, S., O’Keefe, S. J. & O’Connor, D. L. Switching to a fibre-rich and low-fat diet increases colonic folate contents among African Americans. Appl. Physiol. Nutr. Metab. 44, 127–132 (2019).

145.      Soto-Martin, E. C. et al. Vitamin Biosynthesis by Human Gut Butyrate-Producing Bacteria and Cross-Feeding in Synthetic Microbial Communities. MBio 11, (2020).

146.      Blacher, E. et al. Potential roles of gut microbiome and metabolites in modulating ALS in mice. Nature 572, 474–480 (2019).

147.      Thevaranjan, N. et al. Age-Associated Microbial Dysbiosis Promotes Intestinal Permeability, Systemic Inflammation, and Macrophage Dysfunction. Cell Host Microbe 21, 455-466.e4 (2017).

148.      Ghosh, S. et al. Elevated muscle TLR4 expression and metabolic endotoxemia in human aging. J. Gerontol. A. Biol. Sci. Med. Sci. 70, 232–46 (2015).

149.      Stehle, J. R. et al. Lipopolysaccharide-binding protein, a surrogate marker of microbial translocation, is associated with physical function in healthy older adults. J. Gerontol. A. Biol. Sci. Med. Sci. 67, 1212–8 (2012).

150.      Kavanagh, K. et al. Biomarkers of leaky gut are related to inflammation and reduced physical function in older adults with cardiometabolic disease and mobility limitations. GeroScience 41, 923–933 (2019).

151.      Carrera-Bastos, P. et al. Serum Zonulin and Endotoxin Levels in Exceptional Longevity versus Precocious Myocardial Infarction. Aging Dis. 9, 317–321 (2018).

152.      Fransen, F. et al. Aged Gut Microbiota Contributes to Systemical Inflammaging after Transfer to Germ-Free Mice. Front. Immunol. 8, 1385 (2017).

153.      Loffredo, L. et al. Oxidative Stress and Gut-Derived Lipopolysaccharides in Neurodegenerative Disease: Role of NOX2. Oxid. Med. Cell. Longev. 2020, 8630275 (2020).

154.      Ke, Y. et al. Gut flora-dependent metabolite Trimethylamine-N-oxide accelerates endothelial cell senescence and vascular aging through oxidative stress. Free Radic. Biol. Med. 116, 88–100 (2018).

155.      Brunt, V. E. et al. Trimethylamine-N-Oxide Promotes Age-Related Vascular Oxidative Stress and Endothelial Dysfunction in Mice and Healthy Humans. Hypertension 76, 101–112 (2020).

156.      Brunt, V. E. et al. The gut microbiome–derived metabolite trimethylamine N-oxide modulates neuroinflammation and cognitive function with aging. GeroScience (2020). doi:10.1007/s11357-020-00257-2

157.      Li, T., Chen, Y., Gua, C. & Li, X. Elevated Circulating Trimethylamine N-Oxide Levels Contribute to Endothelial Dysfunction in Aged Rats through Vascular Inflammation and Oxidative Stress. Front. Physiol. 8, 350 (2017).

158.      Brunt, V. E. et al. Suppression of the gut microbiome ameliorates age-related arterial dysfunction and oxidative stress in mice. J. Physiol. 597, 2361–2378 (2019).

159.      Vogt, N. M. et al. The gut microbiota-derived metabolite trimethylamine N -oxide is elevated in Alzheimer’s disease. Alzheimer’s Res. Ther. 2018 101 10, 124 (2018).

160.      Chen, M.-L. et al. Trimethylamine-N-Oxide Induces Vascular Inflammation by Activating the NLRP3 Inflammasome Through the SIRT3-SOD2-mtROS Signaling Pathway. J. Am. Heart Assoc. 6, (2017).

161.      Pannérec, A. et al. Vitamin B12 deficiency and impaired expression of amnionless during aging. J. Cachexia. Sarcopenia Muscle 9, 41–52 (2018).

162.      Jones, M. K. et al. Loss of parietal cell superoxide dismutase leads to gastric oxidative stress and increased injury susceptibility in mice. Am. J. Physiol. Gastrointest. Liver Physiol. 301, G537-46 (2011).

163.      Sidani, S. et al. AMP-activated protein kinase: a physiological off switch for murine gastric acid secretion. Pflugers Arch. 459, 39–46 (2009).

164.      van der Vliet, A., Tuinstra, T. J. & Bast, A. Modulation of oxidative stress in the gastrointestinal tract and effect on rat intestinal motility. Biochem. Pharmacol. 38, 2807–18 (1989).

165.      Rana, S. V. et al. Relationship of cytokines, oxidative stress and GI motility with bacterial overgrowth in ulcerative colitis patients. J. Crohns. Colitis 8, 859–65 (2014).

166.      Rivera-Chávez, F., Lopez, C. A. & Bäumler, A. J. Oxygen as a driver of gut dysbiosis. Free Radic. Biol. Med. 105, 93–101 (2017).

167.      Yardeni, T. et al. Host mitochondria influence gut microbiome diversity: A role for ROS. Sci. Signal. 12, (2019).

168.      Timmers, P. R. H. J., Wilson, J. F., Joshi, P. K. & Deelen, J. Multivariate genomic scan implicates novel loci and haem metabolism in human ageing. Nat. Commun. 11, 3570 (2020).

169.      Sinclair, D. A. Lifespan: Why We Age – and Why We Don’t Have To. (Thorsons, 2019).

170.      Fedintsev, A. & Moskalev, A. Stochastic non-enzymatic modification of long-lived macromolecules - A missing hallmark of aging. Ageing Res. Rev. 62, 101097 (2020).

171.      Mehdipour, M. et al. Rejuvenation of three germ layers tissues by exchanging old blood plasma with saline-albumin. Aging (Albany. NY). 12, 8790–8819 (2020).

172.      Dong, Y., Sameni, S., Digman, M. A. & Brewer, G. J. Reversibility of Age-related Oxidized Free NADH Redox States in Alzheimer’s Disease Neurons by Imposed External Cys/CySS Redox Shifts. Sci. Rep. 9, 11274 (2019).

173.      Claesson, M. J. et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 488, 178–84 (2012).

174.      Ghosh, T. S. et al. Mediterranean diet intervention alters the gut microbiome in older people reducing frailty and improving health status: the NU-AGE 1-year dietary intervention across five European countries. Gut 69, 1218–1228 (2020).

175.      Bettermann, E. L. et al. Higher Mediterranean Diet Quality Scores and Lower Body Mass Index Are Associated with a Less-Oxidized Plasma Glutathione and Cysteine Redox Status in Adults. J. Nutr. 148, 245–253 (2018).

176.      El Assar, M. et al. Better Nutritional Status Is Positively Associated with mRNA Expression of SIRT1 in Community-Dwelling Older Adults in the Toledo Study for Healthy Aging. J. Nutr. 148, 1408–1414 (2018).

177.      Le Couteur, D. G. et al. Nutritional reprogramming of mouse liver proteome is dampened by metformin, resveratrol, and rapamycin. Cell Metab. (2021). doi:10.1016/j.cmet.2021.10.016

178.      Aune, D. et al. Fruit and vegetable intake and the risk of cardiovascular disease, total cancer and all-cause mortality-a systematic review and dose-response meta-analysis of prospective studies. Int. J. Epidemiol. 14, 207 (2017).

179.      Aune, D. et al. Whole grain consumption and risk of cardiovascular disease, cancer, and all cause and cause specific mortality: systematic review and dose-response meta-analysis of prospective studies. BMJ 353, i2716 (2016).

180.      Carlsen, M. H. et al. The total antioxidant content of more than 3100 foods, beverages, spices, herbs and supplements used worldwide. Nutr. J. 9, 3 (2010).

181.      Pérez-Jiménez, J., Neveu, V., Vos, F. & Scalbert, A. Identification of the 100 richest dietary sources of polyphenols: An application of the Phenol-Explorer database. Eur. J. Clin. Nutr. 64, S112–S120 (2010).

182.      Pall, M. L. & Levine, S. Nrf2, a master regulator of detoxification and also antioxidant, anti-inflammatory and other cytoprotective mechanisms, is raised by health promoting factors. Sheng Li Xue Bao 67, 1–18 (2015).

183.      Senger, D. R., Li, D., Jaminet, S.-C. & Cao, S. Activation of the Nrf2 Cell Defense Pathway by Ancient Foods: Disease Prevention by Important Molecules and Microbes Lost from the Modern Western Diet. PLoS One 11, e0148042 (2016).

184.      Alfieri, A. et al. Sulforaphane preconditioning of the Nrf2/HO-1 defense pathway protects the cerebral vasculature against blood-brain barrier disruption and neurological deficits in stroke. Free Radic. Biol. Med. 65, 1012–1022 (2013).

185.      Khan, N. M. et al. Wogonin, a plant derived small molecule, exerts potent anti-inflammatory and chondroprotective effects through the activation of ROS/ERK/Nrf2 signaling pathways in human Osteoarthritis chondrocytes. Free Radic. Biol. Med. 106, 288–301 (2017).

186.      Bhattacharjee, S. & Dashwood, R. H. Epigenetic Regulation of NRF2/KEAP1 by Phytochemicals. Antioxidants (Basel, Switzerland) 9, (2020).

187.      Bergström, P. et al. Repeated transient sulforaphane stimulation in astrocytes leads to prolonged Nrf2-mediated gene expression and protection from superoxide-induced damage. Neuropharmacology 60, 343–53 (2011).

188.      González-Sarrías, A., Espín, J. C. & Tomás-Barberán, F. A. Non-extractable polyphenols produce gut microbiota metabolites that persist in circulation and show anti-inflammatory and free radical-scavenging effects. Trends Food Sci. Technol. 69, 281–288 (2017).

189.      Kay, C. D., Pereira-Caro, G., Ludwig, I. A., Clifford, M. N. & Crozier, A. Anthocyanins and Flavanones Are More Bioavailable than Previously Perceived: A Review of Recent Evidence. Annu. Rev. Food Sci. Technol. 8, 155–180 (2017).

190.      Ghanim, H. et al. A resveratrol and polyphenol preparation suppresses oxidative and inflammatory stress response to a high-fat, high-carbohydrate meal. J. Clin. Endocrinol. Metab. 96, 1409–14 (2011).

191.      Taub, P. R. et al. Beneficial effects of dark chocolate on exercise capacity in sedentary subjects: underlying mechanisms. A double blind, randomized, placebo controlled trial. Food Funct. 7, 3686–93 (2016).

192.      Annuzzi, G. et al. Diets naturally rich in polyphenols improve fasting and postprandial dyslipidemia and reduce oxidative stress: A randomized controlled trial. Am. J. Clin. Nutr. 99, 463–471 (2014).

193.      Usharani, P., Fatima, N. & Muralidhar, N. Effects of Phyllanthus emblica extract on endothelial dysfunction and biomarkers of oxidative stress in patients with type 2 diabetes mellitus: A randomized, double-blind, controlled study. Diabetes, Metab. Syndr. Obes. Targets Ther. 6, 275–284 (2013).

194.      Ramirez-Sanchez, I. et al. (-)-Epicatechin rich cocoa mediated modulation of oxidative stress regulators in skeletal muscle of heart failure and type 2 diabetes patients. Int. J. Cardiol. 168, 3982–3990 (2013).

195.      Mastroiacovo, D. et al. Cocoa flavanol consumption improves cognitive function, blood pressure control, and metabolic profile in elderly subjects: the Cocoa, Cognition, and Aging (CoCoA) Study--a randomized controlled trial. Am. J. Clin. Nutr. 101, 538–48 (2015).

196.      Munguia, L. et al. High Flavonoid Cocoa Supplement Ameliorates Plasma Oxidative Stress and Inflammation Levels While Improving Mobility and Quality of Life in Older Subjects: A Double-Blind Randomized Clinical Trial. J. Gerontol. A. Biol. Sci. Med. Sci. 74, 1620–1627 (2019).

197.      Yang, H. et al. Curcumin attenuates urinary excretion of albumin in type ii diabetic patients with enhancing nuclear factor erythroid-derived 2-like 2 (Nrf2) system and repressing inflammatory signaling efficacies. Exp. Clin. Endocrinol. Diabetes 123, 360–367 (2015).

198.      Li, Q., Zhao, H., Zhao, M., Zhang, Z. & Li, Y. Chronic green tea catechins administration prevents oxidative stress-related brain aging in C57BL/6J mice. Brain Res. 1353, 28–35 (2010).

199.      Moreno-Ulloa, A. et al. Recovery of Indicators of Mitochondrial Biogenesis, Oxidative Stress, and Aging With (-)-Epicatechin in Senile Mice. J. Gerontol. A. Biol. Sci. Med. Sci. 70, 1370–8 (2015).

200.      Fleenor, B. S. et al. Curcumin ameliorates arterial dysfunction and oxidative stress with aging. Exp. Gerontol. 48, 269–76 (2013).

201.      Yokozawa, T. et al. Amla (Emblica officinalis Gaertn.) attenuates age-related renal dysfunction by oxidative stress. J. Agric. Food Chem. 55, 7744–52 (2007).

202.      Ren, B. et al. Protective Effects of Sesamol on Systemic Inflammation and Cognitive Impairment in Aging Mice. J. Agric. Food Chem. 68, 3099–3111 (2020).

203.      Li, B. et al. Lactobacillus helveticus KLDS1.8701 alleviates d-galactose-induced aging by regulating Nrf-2 and gut microbiota in mice. Food Funct. 9, 6586–6598 (2018).

204.      Ho, S.-T., Hsieh, Y.-T., Wang, S.-Y. & Chen, M.-J. Improving effect of a probiotic mixture on memory and learning abilities in d-galactose-treated aging mice. J. Dairy Sci. 102, 1901–1909 (2019).

205.      Walsh, M. E. et al. The histone deacetylase inhibitor butyrate improves metabolism and reduces muscle atrophy during aging. Aging Cell 14, 957–70 (2015).

206.      Si, H. et al. Dietary epicatechin improves survival and delays skeletal muscle degeneration in aged mice. FASEB J. 33, 965–977 (2019).

207.      Choi, S. J. et al. Biological effects of rutin on skin aging. Int. J. Mol. Med. 38, 357–63 (2016).

208.      Zhang, Y. et al. Association of fish and long-chain omega-3 fatty acids intakes with total and cause-specific mortality: prospective analysis of 421 309 individuals. J. Intern. Med. 284, 399–417 (2018).

209.      Jayedi, A., Shab-Bidar, S., Eimeri, S. & Djafarian, K. Fish consumption and risk of all-cause and cardiovascular mortality: a dose-response meta-analysis of prospective observational studies. Public Health Nutr. 21, 1297–1306 (2018).

210.      Donat-Vargas, C. et al. Cardiovascular and cancer mortality in relation to dietary polychlorinated biphenyls and marine polyunsaturated fatty acids: a nutritional-toxicological aspect of fish consumption. J. Intern. Med. 287, 197–209 (2020).

211.      Hall, W. L. The future for long chain n-3 PUFA in the prevention of coronary heart disease: do we need to target non-fish-eaters? Proc. Nutr. Soc. 76, 408–418 (2017).

212.      Kris-Etherton, P. M. et al. Recent Clinical Trials Shed New Light on the Cardiovascular Benefits of Omega-3 Fatty Acids. Methodist Debakey Cardiovasc. J. 15, 171–178 (2019).

213.      Heshmati, J. et al. Omega-3 fatty acids supplementation and oxidative stress parameters: A systematic review and meta-analysis of clinical trials. Pharmacol. Res. 149, 104462 (2019).

214.      Golpour, P. et al. Improvement of NRF2 gene expression and antioxidant status in patients with type 2 diabetes mellitus after supplementation with omega-3 polyunsaturated fatty acids: A double-blind randomised placebo-controlled clinical trial. Diabetes Res. Clin. Pract. 162, 108120 (2020).

215.      Gao, L. et al. Novel n-3 fatty acid oxidation products activate Nrf2 by destabilizing the association between Keap1 and Cullin3. J. Biol. Chem. 282, 2529–37 (2007).

216.      Nakagawa, F. et al. 4-Hydroxy hexenal derived from dietary n-3 polyunsaturated fatty acids induces anti-oxidative enzyme heme oxygenase-1 in multiple organs. Biochem. Biophys. Res. Commun. 443, 991–6 (2014).

217.      Shati, A. A. & El-Kott, A. F. Resolvin D1 protects against cadmium chloride-induced memory loss and hippocampal damage in rats: A comparison with docosahexaenoic acid. Hum. Exp. Toxicol. 9603271211038740 (2021). doi:10.1177/09603271211038739

218.      Kang, G. J., Kim, E. J. & Lee, C. H. Therapeutic Effects of Specialized Pro-Resolving Lipids Mediators on Cardiac Fibrosis via NRF2 Activation. Antioxidants (Basel, Switzerland) 9, (2020).

219.      Yum, H.-W. et al. Endogenous ω-3 Fatty Acid Production by fat-1 Transgene and Topically Applied Docosahexaenoic Acid Protect against UVB-induced Mouse Skin Carcinogenesis. Sci. Rep. 7, 11658 (2017).

220.      Yum, H.-W., Kim, S. H., Kang, J. X. & Surh, Y.-J. Amelioration of UVB-induced oxidative stress and inflammation in fat-1 transgenic mouse skin. Biochem. Biophys. Res. Commun. 502, 1–8 (2018).

221.      Weiser, M. J., Butt, C. M. & Mohajeri, M. H. Docosahexaenoic Acid and Cognition throughout the Lifespan. Nutrients 8, 99 (2016).

222.      Yin, X. et al. The Relationship between Fish Intake and Urinary Trimethylamine-N-Oxide. Mol. Nutr. Food Res. 64, 1–9 (2020).

223.      Hagen, I. V et al. TMAO, creatine and 1-methylhistidine in serum and urine are potential biomarkers of cod and salmon intake: a randomised clinical trial in adults with overweight or obesity. Eur. J. Nutr. 59, 2249–2259 (2020).

224.      Romeu, M. et al. Diet, iron biomarkers and oxidative stress in a representative sample of Mediterranean population. Nutr. J. 12, 102 (2013).

225.      Pamphlett, R., Bishop, D. P., Kum Jew, S. & Doble, P. A. Age-related accumulation of toxic metals in the human locus ceruleus. PLoS One 13, e0203627 (2018).

226.      Pamphlett, R., Kum Jew, S., Doble, P. A. & Bishop, D. P. Elemental Analysis of Aging Human Pituitary Glands Implicates Mercury as a Contributor to the Somatopause. Front. Endocrinol. (Lausanne). 10, 419 (2019).

227.      Erie, J. C., Good, J. A. & Butz, J. A. Excess lead in the neural retina in age-related macular degeneration. Am. J. Ophthalmol. 148, 890–4 (2009).

228.      Wills, N. K. et al. Cadmium accumulation in the human retina: effects of age, gender, and cellular toxicity. Exp. Eye Res. 86, 41–51 (2008).

229.      Pardo, M., Qiu, X., Zimmermann, R. & Rudich, Y. Particulate Matter Toxicity Is Nrf2 and Mitochondria Dependent: The Roles of Metals and Polycyclic Aromatic Hydrocarbons. Chem. Res. Toxicol. 33, 1110–1120 (2020).

230.      Fratta Pasini, A. et al. Serum oxidative stress-induced repression of Nrf2 and GSH depletion: a mechanism potentially involved in endothelial dysfunction of young smokers. PLoS One 7, e30291 (2012).

231.      Kode, A. et al. Resveratrol induces glutathione synthesis by activation of Nrf2 and protects against cigarette smoke-mediated oxidative stress in human lung epithelial cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 294, L478-88 (2008).

232.      Chen, J.-H., Lin, X., Bu, C. & Zhang, X. Role of advanced glycation end products in mobility and considerations in possible dietary and nutritional intervention strategies. Nutr. Metab. (Lond). 15, 72 (2018).

233.      Uribarri, J. et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J. Am. Diet. Assoc. 110, 911–16.e12 (2010).

234.      Snelson, M. & Coughlan, M. T. Dietary Advanced Glycation End Products: Digestion, Metabolism and Modulation of Gut Microbial Ecology. Nutrients 11, (2019).

235.      Uribarri, J. et al. Circulating glycotoxins and dietary advanced glycation endproducts: two links to inflammatory response, oxidative stress, and aging. J. Gerontol. A. Biol. Sci. Med. Sci. 62, 427–33 (2007).

236.      Vlassara, H. et al. Protection against loss of innate defenses in adulthood by low advanced glycation end products (AGE) intake: role of the antiinflammatory AGE receptor-1. J. Clin. Endocrinol. Metab. 94, 4483–91 (2009).

237.      Uribarri, J. et al. Suppression of native defense mechanisms, SIRT1 and PPARγ, by dietary glycoxidants precedes disease in adult humans; relevance to lifestyle-engendered chronic diseases. Amino Acids 46, 301–9 (2014).

238.      Vlassara, H. et al. Oral AGE restriction ameliorates insulin resistance in obese individuals with the metabolic syndrome: a randomised controlled trial. Diabetologia 59, 2181–92 (2016).

239.      Cai, W. et al. Oral glycotoxins determine the effects of calorie restriction on oxidant stress, age-related diseases, and lifespan. Am. J. Pathol. 173, 327–36 (2008).

240.      Chen, X.-J. et al. Advanced glycation end-products induce oxidative stress through the Sirt1/Nrf2 axis by interacting with the receptor of AGEs under diabetic conditions. J. Cell. Biochem. (2018). doi:10.1002/jcb.27524

241.      Tang, Y. & Chen, A. Curcumin eliminates the effect of advanced glycation end-products (AGEs) on the divergent regulation of gene expression of receptors of AGEs by interrupting leptin signaling. Lab. Invest. 94, 503–16 (2014).

242.      LaRocca, T. J., Gioscia-Ryan, R. A., Hearon, C. M. & Seals, D. R. The autophagy enhancer spermidine reverses arterial aging. Mech. Ageing Dev. 134, 314–20 (2013).

243.      Done, A. J. & Traustadóttir, T. Aerobic exercise increases resistance to oxidative stress in sedentary older middle-aged adults. A pilot study. Age (Dordr). 38, 505–512 (2016).

244.      Koltai, E. et al. Exercise alters SIRT1, SIRT6, NAD and NAMPT levels in skeletal muscle of aged rats. Mech. Ageing Dev. 131, 21–8 (2010).

245.      Gounder, S. S. et al. Impaired transcriptional activity of Nrf2 in age-related myocardial oxidative stress is reversible by moderate exercise training. PLoS One 7, e45697 (2012).

246.      Moreno-Indias, I. et al. Red wine polyphenols modulate fecal microbiota and reduce markers of the metabolic syndrome in obese patients. Food Funct. 7, 1775–87 (2016).

247.      Bruns, D. R. et al. Differential effects of vitamin C or protandim on skeletal muscle adaptation to exercise. J. Appl. Physiol. 125, 661–671 (2018).

248.      Schwarz, N. A. et al. (-)-Epicatechin Supplementation Inhibits Aerobic Adaptations to Cycling Exercise in Humans. Front. Nutr. 5, 132 (2018).

249.      Gliemann, L., Nyberg, M. & Hellsten, Y. Effects of exercise training and resveratrol on vascular health in aging. Free Radic. Biol. Med. 98, 165–176 (2016).

250.      Mitteldorf, J. Aging is not a process of wear and tear. Rejuvenation Res. 13, 322–6 (2010).

251.      Bortz, W. M. Aging as entropy. Exp. Gerontol. 21, 321–8 (1986).

252.      Hayflick, L. Entropy explains aging, genetic determinism explains longevity, and undefined terminology explains misunderstanding both. PLoS Genet. 3, e220 (2007).

253.      Yin, D. The Essential Mechanisms of Aging: What Have We Learnt in Ten Years? Curr. Top. Med. Chem. 16, 503–10 (2016).

254.      Chatterjee, A., Georgiev, G. & Iannacchione, G. Aging and efficiency in living systems: Complexity, adaptation and self-organization. Mech. Ageing Dev. 163, 2–7 (2017).

255.      Aledo, J. C. & Blanco, J. M. Aging is neither a failure nor an achievement of natural selection. Curr. Aging Sci. 8, 4–10 (2015).

256.      Zhang, B. & Gladyshev, V. N. How can aging be reversed? Exploring rejuvenation from a damage-based perspective. Genet. Genomics Next 1, e10025 (2020).

257.      Afshinnekoo, E. et al. Fundamental Biological Features of Spaceflight: Advancing the Field to Enable Deep-Space Exploration. Cell 183, 1162–1184 (2020).

258.      Meydani, S. N., Lewis, E. D. & Wu, D. Perspective: Should Vitamin E Recommendations for Older Adults Be Increased? Adv. Nutr. 9, 533–543 (2018).

259.      Choi, S.-W., Yeung, V. T. F., Collins, A. R. & Benzie, I. F. F. Redox-linked effects of green tea on DNA damage and repair, and influence of microsatellite polymorphism in HMOX-1: results of a human intervention trial. Mutagenesis 30, 129–37 (2015).

260.      Ho, C. K., Choi, S., Siu, P. M. & Benzie, I. F. F. Effects of single dose and regular intake of green tea (Camellia sinensis) on DNA damage, DNA repair, and heme oxygenase-1 expression in a randomized controlled human supplementation study. Mol. Nutr. Food Res. 58, 1379–83 (2014).

261.      Hachmo, Y. et al. Hyperbaric oxygen therapy increases telomere length and decreases immunosenescence in isolated blood cells: a prospective trial. Aging (Albany. NY). 12, (2020).

262.      Ferrández, M. D., Correa, R., Del Rio, M. & De la Fuente, M. Effects in vitro of several antioxidants on the natural killer function of aging mice. Exp. Gerontol. 34, 675–85 (1999).

263.      Ismail, I. A., El-Bakry, H. A. & Soliman, S. S. Melatonin and tumeric ameliorate aging-induced changes: implication of immunoglobulins, cytokines, DJ-1/NRF2 and apoptosis regulation. Int. J. Physiol. Pathophysiol. Pharmacol. 10, 70–82 (2018).

264.      Stanhewicz, A. E., Alexander, L. M. & Kenney, W. L. Folic acid supplementation improves microvascular function in older adults through nitric oxide-dependent mechanisms. Clin. Sci. 129, 159–167 (2015).

265.      Romero, S. A. et al. Folic acid ingestion improves skeletal muscle blood flow during graded handgrip and plantar flexion exercise in aged humans. Am. J. Physiol. Circ. Physiol. 313, H658–H666 (2017).

266.      Heiss, C. et al. Impact of cocoa flavanol intake on age-dependent vascular stiffness in healthy men: a randomized, controlled, double-masked trial. Age (Dordr). 37, 9794 (2015).

267.      Heiss, C. et al. Improvement of endothelial function with dietary flavanols is associated with mobilization of circulating angiogenic cells in patients with coronary artery disease. J. Am. Coll. Cardiol. 56, 218–24 (2010).

268.      Aviram, M. et al. Pomegranate juice consumption for 3 years by patients with carotid artery stenosis reduces common carotid intima-media thickness, blood pressure and LDL oxidation. Clin. Nutr. 23, 423–33 (2004).

269.      Ramirez-Sanchez, I. et al. (-)-Epicatechin induced reversal of endothelial cell aging and improved vascular function: underlying mechanisms. Food Funct. 9, 4802–4813 (2018).

270.      Monahan, K. D., Eskurza, I. & Seals, D. R. Ascorbic acid increases cardiovagal baroreflex sensitivity in healthy older men. Am. J. Physiol. Heart Circ. Physiol. 286, H2113-7 (2004).

271.      Manzella, D., Barbieri, M., Ragno, E. & Paolisso, G. Chronic administration of pharmacologic doses of vitamin E improves the cardiac autonomic nervous system in patients with type 2 diabetes. Am. J. Clin. Nutr. 73, 1052–7 (2001).

272.      Ozcamdalli, M. et al. Comparison of Intra-articular Injection of Hyaluronic Acid and N-Acetyl Cysteine in the Treatment of Knee Osteoarthritis: A Pilot Study. Cartilage 8, 384–390 (2017).

273.      Ghoochani, N., Karandish, M., Mowla, K., Haghighizadeh, M. H. & Jalali, M. T. The effect of pomegranate juice on clinical signs, matrix metalloproteinases and antioxidant status in patients with knee osteoarthritis. J. Sci. Food Agric. 96, 4377–81 (2016).

274.      Ruiz-Ramos, M., Vargas, L. A., Fortoul Van der Goes, T. I., Cervantes-Sandoval, A. & Mendoza-Nunez, V. M. Supplementation of ascorbic acid and alpha-tocopherol is useful to preventing bone loss linked to oxidative stress in elderly. J. Nutr. Health Aging 14, 467–72 (2010).

275.      Ornstrup, M. J., Harsløf, T., Kjær, T. N., Langdahl, B. L. & Pedersen, S. B. Resveratrol increases bone mineral density and bone alkaline phosphatase in obese men: a randomized placebo-controlled trial. J. Clin. Endocrinol. Metab. 99, 4720–9 (2014).

276.      Geng, L. et al. Chemical screen identifies a geroprotective role of quercetin in premature aging. Protein Cell 10, 417–435 (2019).

277.      Lamport, D. J. et al. The effect of flavanol-rich cocoa on cerebral perfusion in healthy older adults during conscious resting state: a placebo controlled, crossover, acute trial. Psychopharmacology (Berl). 232, 3227–34 (2015).

278.      Brickman, A. M. et al. Enhancing dentate gyrus function with dietary flavanols improves cognition in older adults. Nat. Neurosci. 17, 1798–803 (2014).

279.      Neshatdoust, S. et al. High-flavonoid intake induces cognitive improvements linked to changes in serum brain-derived neurotrophic factor: Two randomised, controlled trials. Nutr. Heal. aging 4, 81–93 (2016).

280.      Richer, S. et al. Resveratrol based oral nutritional supplement produces long-term beneficial effects on structure and visual function in human patients. Nutrients 6, 4404–20 (2014).

281.      Diyana, I., Stuart, R. & Anish, B. Improved Visual Acuity and Retinal Integrity with Resveratrol Based Supplementation in Patients with Macular Degeneration. Int. J. Ophthalmol. Clin. Res. 4, 1–7 (2017).

282.      Feher, J. et al. Improvement of visual functions and fundus alterations in early age-related macular degeneration treated with a combination of acetyl-L-carnitine, n-3 fatty acids, and coenzyme Q10. Ophthalmologica 219, 154–166 (2005).

283.      Herman, J. Case Report of Dietary Supplements Improving Macular Pigment and Visual Function. Adv. Ophthalmol. Vis. Syst. 6, 1–11 (2017).

284.      Yoon, H.-S. et al. Cocoa Flavanol Supplementation Influences Skin Conditions of Photo-Aged Women: A 24-Week Double-Blind, Randomized, Controlled Trial. J. Nutr. 146, 46–50 (2016).

285.      Weschawalit, S., Thongthip, S., Phutrakool, P. & Asawanonda, P. Glutathione and its antiaging and antimelanogenic effects. Clin. Cosmet. Investig. Dermatol. 10, 147–153 (2017).

286.      Sohn, E.-J. et al. Restoring Effects of Natural Anti-Oxidant Quercetin on Cellular Senescent Human Dermal Fibroblasts. Am. J. Chin. Med. 46, 853–873 (2018).

 

No comments:

Post a Comment