Skip to main content
Aging and Disease logoLink to Aging and Disease
. 2022 Dec 1;13(6):1664–1714. doi: 10.14336/AD.2022.0414

Resolving Geroplasticity to the Balance of Rejuvenins and Geriatrins

Siamak Tabibzadeh 1,*
PMCID: PMC9662275  PMID: 36465174

Abstract

According to the cell centric hypotheses, the deficits that drive aging occur within cells by age dependent progressive damage to organelles, telomeres, biologic signaling pathways, bioinformational molecules, and by exhaustion of stem cells. Here, we amend these hypotheses and propose an eco-centric model for geroplasticity (aging plasticity including aging reversal). According to this model, youth and aging are plastic and require constant maintenance, and, respectively, engage a host of endogenous rejuvenating (rejuvenins) and gero-inducing [geriatrin] factors. Aging in this model is akin to atrophy that occurs as a result of damage or withdrawal of trophic factors. Rejuvenins maintain and geriatrins adversely impact cellular homeostasis, cell fitness, and proliferation, stem cell pools, damage response and repair. Rejuvenins reduce and geriatrins increase the age-related disorders, inflammatory signaling, and senescence and adjust the epigenetic clock. When viewed through this perspective, aging can be successfully reversed by supplementation with rejuvenins and by reducing the levels of geriatrins.

Keywords: geroplasticity, balance, rejuvenins, geriatrins

Introduction

At the organismal level, aging is evident in all human beings by loss of the ability to reproduce, and dysfunction or loss of function in organs, tissues, and cells (Tables 1-2). There is myriad of cell-centric damages that are associated with aging. In 1981, Harman ascribed aging to be due to a progressive accumulation of temporal changes that lead to an ever-increasing susceptibility to disease and death [1]. According to Hayflick, the common denominator that underlies all modern theories of biological aging is change in molecular structure(s) and, hence, function of cells [2]. This includes damage to bioinformational macromolecules, proteins, carbo-hydrates and lipids as well as concomitant hyperfunction of certain pathways, most notably, of mTOR and NF-κB signaling [3]. Other changes include DNA mutations and genomic instability, glycation, side reactions, and loss of proteostasis (protein folding and proteolysis). Aging is associated with replication-associated DNA damage that perturbs normal cell function as a result of altered specific or global gene expression. changes in transcription and RNA processing modifying protein production. Damage to proteins contributes to cellular aging as a result of formation of mis-folded and aggregated proteins that cause loss of function of proteins and loss of a stable and functional proteome [4-5]. Ever increasing levels of radical oxygen species (ROS) cause an increase in oxidative stressogeneic mutations.

Table 1.

Impact of aging and senescence on cellular parameters.

Item Effect Reference
Integrity of membrane barriers
Mitochondrial membrane (membrane bound ATP) 19
Nuclear membrane 20
Lysosome membrane 16, 17
Plasma membrane 16, 17
DNA
Integrity 21
Global DNA methylation 16, 17
Site specific DNA methylation ↑↓ 16, 17
Histone modifications
Global acetylation 22-24
H3K4me3 16, 17
H3K9me 16, 17, 25
H3K9me2 ↑↓ 25
H3K9me3 26-30
H3K9ac 25, 28, 31-34
H3S10phospho 25, 34
H3K27me3 35-36
H3K56ac 25, 37-38
H4K16ac 16, 17, 25, 37
H4K20me 25
H4K20me2 ↓↑ 16, 17, 25, 39
H4K20me3 ↓↑ 39
H3.1 25, 38, 40, 41, 1999
H3.2 25, 38, 40 41
H3.3 40, 41,
H4 25
H2A.1 38, 40, 41
H2A.2 25, 40, 41,
γH2AX 25, 42
Chromatin remodeling
HP1α 16-17
NuRD 16-17
Epigenome
Epigenetic clock 43
Other changes 44
Telomere
Telomere shortening 45
Telomere elongation 45
Mitochondria
Mitochondrial biogenesis 16-17, 46
Mitochondrial function 16-17, 47-48
Bioenergetics 49
ΔΨm 50
ATP 51
NAD+ 52-53
TCA cycle 54
Glycolysis 55-56
Deregulated Signaling
mTOR 57-58
NFκB 59
Nutrient signaling 16-17, 48
Omics
Transcriptome ↓↑ 60-61
Acetylome ↓↑ 60
Proteome ↓↑ 16-17, 60, 62
NAD+ metabolome 54
αKetoglutarate metabolome 54
Robustness of Housekeeping Function
Ubiquitin Proteasome System (UPS) 63
Autophagy 64-66
Mitophagy 66-67
Robustness of ER Stress Response
Unfolded protein response (UPR) 68-69
Cell fitness
Robustness of circadian rhythms 70
Proliferation, regeneration and increased population doublings 71-73
Lost functions 73
Cell signaling ↓↑ 74-75
Damage Response
Oxidative-ROS 76-77
DNA 67, 78-80
Cancer 81-82
Repair Response
Cell and tissue repair 83
PARP activity 84
Inflammatory Response
Inflammatory signaling 85-86
Senescence
Hallmarks of senescence 87-89
SASP 90
Stem cells
Exhaustion 16-17, 48
Cell Death
Apoptosis 91

↑ Improved or protected, ↓ Deteriorated or reversed, Mutations or other changes that are unlikely to be reversible are not included.

Table 2.

Impact of aging and senescence on clinical parameters

Item Effect Reference
Blood and Bone Marrow
Acid base balance 92
Blood Lipidome ↓↑ 93-94
Blood Metabolome ↓↑ 95-96
Blood pressure 97
Bone marrow function 98-100
Integrity of endothelial barriers 16-17
Bone
Bone mass 101
ECM
ECM biosynthesis and organization 102-103
ECM degradation 104
Energy and Metabolism
Glucose tolerance 105-106
Energy utilization - Appetite and stamina 107-110
Fat depots 111
GI tract
Integrity of GI barrier (leaky gut) 16-17
Immune Response
Immune response (immunosenescence) 16-17, 112
Inflammation 113-116
Liver
Liver metabolism 117-118
Musculoskeletal System
Muscle mass and physical frailty 119
Nervous System
Memory 120
Neural plasticity 121
Integrity of BBB 122
Cognition 110, 122
Sleep 124
Thermoregulation 107, 125, 126
Reproductive System
Fertility 127, 128
Respiratory System
Integrity of respiratory tract barrier 16-17
Oxygenation 107, 130-131
Skin
Integrity of Skin Barrier 16-17
Stem Cells and Tissue Regeneration
Stem cell function 16-17, 132-134
Tissue regeneration 132, 135, 136
Organ Protection
Organ, tissue, and metabolic protection 16-17, 110
Senescence
Senescence 115, 137
Aging
DNAm Age 44
Age-related disorders and disease 88, 110
Cancer
Risk of cancer 81, 138
Health-span and Lifespan
Health-span 139
Lifespan 139, 140

↑ Improved or protected, ↓ Deteriorated or reversed, Mutations or other changes that are unlikely to be reversible are not included.

Aging is also associated with mitochondrial DNA damage and dysfunction. As a result of telomere shortening, actively proliferating cells stop their cell division (replicative senescence) by the engagement of p16INK4a/Rb and p19ARF/p53, creating a diseased tissue landscape [6-12]. Changes that contribute to aging also manifest by loss of cellular, tissue and organ function in long lived cells such as neurons and muscle fibers. Occurrence of chronological and replicative damage in stem cells and their loss impairs the tissue regeneration which is essential to the replacement of short-lived cells that are continuously lost with time [13]. Aging causes emergence of inflammation (inflammaging), and increased susceptibility to the development of cancer [14-15].

There is emerging evidence that the cellular changes that occur in aged tissues can be reversed, and aging cells can be rejuvenated. In this paper, the current evidence that supports that aging is plastic (geroplasticity), the conceptual framework of an eco-centric model for this plasticity and data that support for the existence of rejuvenating (rejuvenin) and age inducing (geriatrin) factors that drive such a plasticity are discussed.

Eco-centric model of aging

Eco-centric model of aging is founded on the basis of two principles (Fig. 1A-D). First, that homeostasis of organs, tissues, and cells after birth is required to be continuously maintained by an on-going rejuvenation of damaged cells and tissues by a host of rejuvenating factors (rejuvenins) (Fig. 1). A host of age promoting factors (geriatrins) oppose the rejuvenating effects of rejuvenins and induce an aging landscape (Fig. 1). Following production from a host of sources, to be disseminated throughout the body, rejuvenins and geriatrins first enter the interstitium of all tissues and then they transit to peripheral blood, and by passing the Blood-Brain-Barrier (BBB), enter cerebrospinal fluid (CSF) and brain tissues. Second, rejuvenins are abundant early in life, and their age dependent progressive decline with age, leads to the loss of their impact on cell and organ health and a steady increase in epigenetic age (Fig. 1). On the other hand, concomitant with reducing levels of rejuvenins in aging, the levels of geriatrins increase with age exacerbating the effect of rejuvenin loss. This allows for emergence of an onslaught of cell-centric damages that are hallmarks of age-related changes that ultimately manifest at organismal levels (Tables 1-2) [16-18]. If aging is viewed through this lens, treatment of aging becomes feasible by the ensemble substitution of lost rejuvenins and reduction of geriatrins. This strategy, reverses, the age mediated cell mal-functions, preferably resets the epigenetic clock to an earlier time-point, restores homeostasis in cells, tissues and organs and extends health-span and lifespan.

Figure 1.

Figure 1.

Geroplasticity resolved to the balance of rejuvenins and geriatrins. (A) At the beginning of life rejuvenins provide cell homeostasis whereas, later in life, loss of rejuvenins and escalating levels of geriatrins lead to aging and age-related disorders. (C) Akin to atrophy caused by withdrawal of trophic factors, aging is caused by declined levels of rejuvenins and increased levels of geriatrins. For example, increase in Hcys leads to cortical atrophy (16). (D) Aging leads to an increase in epigenetic age, whereas the diverse rejuvenation strategies drive down the epigenetic age (17-18).

Rejuvenins, might be considered as part of the innate defense mechanisms that have been evolutionary designed and evolved to prolong life as much as possible and to prevent catastrophic early failures of cells, tissues, and organs, as they occur in progeroid syndromes. In contradistinction, late loss of these molecules can significantly prolong life in octogenarians (80-89 years), nonagenarians (90-99 years), centenarians (100-109 years) and super-centenarians (110 years and older).

There are several conditions that lead to cell rejuvenation and resetting of biologic and epigenetic clocks (Fig. 1D). In all such cases, restoration of old cells to more youthful cells and tissue phenotypes have been associated with the molecular signatures of youthfulness.

Rejuvenation by fertilization and parthenogenesis

The most pronounced form of rejuvenation occurs after each fertilization. The cytoplasm of a zygote holds factors that are capable of resetting the epigenetic clock. In mouse, within 6-8 hours after fertilization, paternal genome, actively undergoes extensive demethylation, whereas the demethylation of maternal genome is initiated after several cell divisions [141,142]. The reprogramming in DNA methylation continues in early stages of embryogenesis, leading to rejuvenation and allowing the organism to start life at an earliest feasible epigenetic time-point (Fig. 1D) [143]. The rejuvenation also occurs when the egg is artificially inseminated or even when the animal is cloned by introducing somatic cell nuclei into a surrogate egg (somatic cell nuclear transfer; SCNT) and then the hybrid cell receives electric shock (parthenogenesis) instead of sperm (fertilization) to initiate embryogenesis [144-146]. The renewal after each fertilization, therefore, shows, that the biologic age of a cell can be completely reset and that most, if not all evidence of damage to cellular compartments, that are detriment to a normal lifespan, can be erased.

It has been known that chronological and biological age do not necessarily match, and some individuals appear and function much better than others who are at the same age. The proof for this observation was achieved by the pioneering work of Horvath. Horvath et al examined the DNA methylation status of 353 genes in 8,000 samples from 51 healthy tissues and various cell types and showed that the epigenetic age can accurately estimate the age of any tissue within a narrow 2-year margin and can be used as a great predictor of heritable acceleration of age [44, 147, 148]. Multi-tissue clock of Horvath also showed that, in human embryonic stem cells (ESCs), the epigenetic age is near zero even after extensive passaging [44]. More recently, Kerepesi et al confirmed these findings and using rDNAm determined the epigenetic age of these cells to be mostly below zero [143]. These cells did not age even after 100 passages indicating that cells, equivalent to those in the early stages of embryogenesis, are essentially immune to aging. The epigenetic age in embryonic stem cells which has been consistently found to be below zero, starts ticking during mid-embryonic development in mouse and humans [143]. In mouse, for example, at E6.5 and E7.5, the epigenetic age was significantly lower than the age of cells at E13.5 (primordial germ cells, the direct progenitors of sperms and oocytes). Thus, the epigenetic age is initially diminished and then it increases from birth, and it consistently advances throughout life in virtually all tissues [143]. These findings show that it is possible to stop and reverse the aging processes in somatic cells if the conditions that exist in embryonic cells can be defined and applied to aging cells.

Rejuvenation by genetic reprogramming

The epigenetic rejuvenation by fertilization can be simulated and the epigenetic age can be set to near zero by inducing genetic reprogramming (iPSCs) [41]. However, one of the side effects of therapeutically using iPSCs is that these cells form teratomas in vivo [149]. Moreover, C-Myc gene is an oncogene that reduces lifespan, and its decrease is associated with increased health-span [148]. Using such an insight, it was recently shown that introduction of Oct4, Sox2, and Klf4 genes (OSK) without cMyc to old mice was sufficient to erase signs of aging [150]. This reprogramming induced in the recipient old mice, a more youthful gene expression pattern. Moreover, after optic nerve crush injury, the treatment promoted axon regeneration and restored vision in a mouse model of glaucoma [151]. Finally, there was evidence of resetting of the DNA methylation age of retinal ganglion cells. The reprogramming was DNA demethylase (Tet1 and Tet2) dependent suggesting that DNA methylation patterns might not be merely co-incidental effects rather they play a causative role in aging. It is also becoming apparent that aberrations of DNA methylation are initiated by double stranded DNA breaks (DSB) that cause persistent age dependent epigenetic drifts in yeast to mammals [152]. Together, these studies have demonstrated that, the genetic reprogramming simultaneously erases differentiation [de-differentiation), causes telomeric rejuvenation and leads to resetting of the epigenetic clock to an earlier time-point. Thus, it appears that, similar to differentiation, aging is plastic and subject to reversal to a more youthful state [153-156]. Interestingly, a backup record of youthful epigenetic information, encoded in part by DNA methylation, appears to exist in mammalian tissues, that is used for restoration of a youthful epigenome [151, 155]. A clear question that arises is whether the de-differentiation can completely be separated from the epigenetic clock reprogramming.

Due to the fear of inducing tumors, Ocampo et al, introduced Yamanaka OSKM factors [Oct4, Sox2, Klf4, and c-Myc) to mouse and human cells under conditions that allowed their cyclic expression [156]. This partial reprogramming was shown to lead to rejuvenation, reversed the cellular and physiologic hallmarks of aging and prolonged the lifespan of a progeroid mouse model that undergoes premature aging [156]. This landmark study showed that, not only individual cells, but the entire organism can be successfully rejuvenated, giving credibility to the notion that rejuvenation of organisms is quite feasible and is within reach. Moreover, this type of reprogramming was not associated with an increased risk of tumorigenesis. Besides genetic reprogramming, the conditioned media or the so-called secretome of stem cells of diverse origins appear to include factors that can rejuvenate tissues. For example, the conditioned media of human umbilical cord or bone marrow derived mesenchymal stem cells rejuvenated human skin [157-158]. Such findings clearly indicate that rejuvenating factors are released and exist within the stem cell secretome. Identifying such factors, therefore, offers the opportunity for rejuvenation of aging human cells.

The studies on epigenetic clock have made it clear that the trajectory of the epigenetic clock differs in different individuals and can be accelerated in certain conditions such as progeria. If the same variation would exist in different tissues of the same individual, the divergence of epigenetic age in different tissues would have ultimately emerged with time [41]. However, aging occurs uniformly throughout the body as evident by the similarity of aging of skin in the left and right sides of the body. The definitive proof for such a uniformity, was provided only recently by examining the epigenetic age of 30 anatomic sites in young people to supercentenarians. The findings confirmed that only the cerebellum and, to a lesser extent, the occipital cortical regions, exhibit negative epigenetic age acceleration in the supercentenarians [157]. DNA methylation (DNAm) age, in a host of sorted cell types (CD4+ T cells, monocytes, B cells, glial cells, and neurons, osteoblasts and osteocytes) as well as diverse organs (brain, breast, kidney, liver, lung, bone), and even the body fluids (whole blood, and saliva) that harbor DNA showed that the “epigenetic clock, uniformly applies to all of these cells, organs and body fluids [159]. Thus, there is a synchronicity in the epigenetic clock throughout the entire body and in the brain. Despite this knowledge, it has remained unclear as what accounts for the precision of the clock, or why its speed varies in different individuals or why and how the uniformity of age in all organs and tissues occurs. One possible explanation is that there are factors that circulate the blood to ensure that the epigenetic clock tick rate is the same throughout the entire body. Moreover, given that brain also ages coordinately with the rest of the body, we argue that the rejuvenating factors must also pass the BBB.

Rejuvenation by parabiosis

The idea that circulating rejuvenating factors exist in blood and CSF is supported by studies that join two animals by their vasculature (parabiosis). Parabiosis is a technique that surgically joins the circulatory systems of two living organisms such as rats and mice so that they will have a single shared circulatory system [160-162]. In rats, the total blood that is exchanged between the two co-joined animals, reaches 10 times of the total volume of blood per day. This ensures that the two animals continuously receive the same level of blood constituents [163]. The pair of joined animals may be of the same age (isochronic parabionts) or can differ in age (heterochronic parabionts) (Fig. 2). Parabiosis defined as “living besides or next to” is one of the oldest surgical techniques which was first carried out close to two centuries ago by Paul Bert by surgically joining two rats, to simulate a condition, similar to the shared circulation of “Siamese twins” [164, 165]. Parabiosis has been used for over 150 years to test whether systemic or circulatory factors from one animal can affect another animal. For example, the discovery of leptin is owed to parabiosis experiments designed to identify whether a “satiety factor” circulates the blood [166]. Parabiosis was neglected until it gained popularity nearly hundred years later. McKay et al showed that in the heterochronic parabiosis, old parabionts gain life-extension, a finding that was validated by Ludwig et al in 1972 who showed a 20% increase in lifespan in old parabionts [167-168]. In these studies, young parabionts suffered negatively from the joined circulatory system perhaps because old blood might dilute the concentration of rejuventating factors present in young blood. However, a more likely possibility is that there are gero-inducing factors (geriatrins) that are produced by aging that exert negative effects on cell health causing the rampant aging deficits. Consistent with the latter possibility, the administration of plasma from old mice inhibited hippocampal neurogenesis in young mice in vivo and inhibited the function and proliferation of neural stem cells (NSC) in vitro [162]. These findings underscore the existence of molecules in the blood of old organisms that oppose or reverse the effect of rejuvenins and induce an aging phenotype (geriatrins).

Figure 2.

Figure 2.

Isochronic and heterochronic parabiosis in mice. In young-old pairings, the blood of young mice has putative rejuvenating factors (rejuvenins) that lead to the rejuvenation of old mice whereas the blood of old mice has putative age inducing factors (geriatrins) that induce age-related disorders.

The idea to join the circulatory systems of a young and an old animal made the parabiosis an impressive tool to examine the aging and age-related diseases in such contexts as diabetes, cardiovascular disease, Alzheimer’s disease, multiple sclerosis, and osteoarthritis [161, 162,169, 170]. These studies have repeatedly led to the conclusion that co-joining of young and old mice causes the rejuvenation of old animals and aging of the young organisms. These studies give credence to the existence of humoral factors in the circulatory system of young organisms that have rejuvenating (rejuvenins) or age inducing (geriatrin) effects [169, 171].

The effects of heterochronic parabiosis on aging involves every organ and tissue in the aged animal. The wide range of rejuvenating activity in heterochronic parabiosis is evident in rejuvenation of aged progenitor and stem cells and skeletal muscle stem (satellite) cells, to an increased density of the dendritic spine of mature neurons and synaptic plasticity in the hippocampus [161, 162, 172]. Heterochronic parabiosis of young and old mouse pairings led to restoration of the cEBP-alpha complex in old parabionts to levels that are seen in young animals and resulted in an increase in proliferation of hepatocytes in aged mice [161]. In a similar model, when compared with satellite cells from aged-isochronic controls, satellite cells in aged heterochronic parabionts exhibited improved myogenic differentiation and lower levels of DNA damage [173].

Using a limiting-dilution, competitive trans-plantation method, it was observed that the frequency of marrow competitive repopulation units (CRUs) increased approximately 2-fold in mice from 2 months to 2 years of age. However, the homing efficiency of old CRUs was approximately 3-fold lower than that of young CRUs. Moreover, the increased age of HSC donors and recipients promoted myelopoiesis [174-177]. However, heterochronic parabiosis led to the restoration of bone marrow function and dramatically reduced long-term HSCs to their normal ‘youthful’ levels.

The notion that young environments have a rejuvenating potential was demonstrated by restoring the regenerative potential of old muscles when transplanted to young rats. However, the young muscles showed declined regeneration when they were grafted to old rats [178]. In both isochronic and heterochronic parabioses, only five days after induction of muscle injury, muscles in young animals regenerated. Whereas injured muscles from old isochronic parabionts failed to regenerate, the heterochronic parabiosis of old and young mice restored the regenerative potential of aged muscles, an effect that was attributed to notch ligand that was shown to be required for muscle regeneration [179].

Rejuvenation by reactivation of telomerase

One of the key features of aging is progressive attrition of telomeres, mainly due to the loss of telomerase activity and their uncapping, which then leads to stem cell depletion and consequently, to progressive tissue atrophy, organ system failure and impaired response to tissue injury [183-181]. In aging mice that harbored short telomeres, increased DNA damage signalling and degenerative phenotypes, telomerase reactivation, reduced the DNA damage signalling and cellular checkpoint responses and restored lost proliferative potential in quiescent cells. This reactivation also eliminated degenerative phenotypes in multiple organs including testes, spleens, and intestines. Moreover, the treatment also led to the reversal of age-related neurodegeneration as evidenced by the restoration of proliferating Sox2+ neural progenitors, and Olig2+ oligodendrocyte populations [181]. Unfortunately, the reactivation of telomerase is not clinically suitable for human use since telomerase activation following telomere dysfunction can lead to carcinogenesis [182].

Rejuvenation by modifying cell signaling

Molecular approaches have allowed pinpointing the cell signaling pathways that have gone awry in aging. Reversal of these aberrant signalings is sufficient to restore homeostasis and reverse age-related declines. Most notably among these, is the activation of NFκB that drives inflammatory response and of mTOR which senses the cellular nutrient levels and regulates the rate of protein synthesis and energy utilization.

Conditional inhibition of NFκB in the epidermis of chronologically aged mice for 2 weeks, restored the gene expression programs that are found in young mice and erased the molecular signature of aging including SA-β-gal activity, p16 down-regulation, and p16INK4A protein expression [183]. This blockade also promoted the proliferation of skin progenitors and their normal differentiation and induced a more youthful skin phenotype that was associated with an increased skin thickness [183].

Similarly, inhibition of mTOR in old mice with rapamycin prevented the age-related increase in hematopoeitic stem cells or their expression of aging biomarkers, reversed aging-related declines in the function of mouse HSCs and enhanced the immune response to infection with influenza virus [184-185].

Rejuvenation by chemical, diet, and life-style changes

Aging reversal has also been tested with a cocktail of specific factors including recombinant human growth hormone (hGH), combined with metformin and DHEA. The participants, initially, received for a week, hGH alone (0.015 mg/kg/day) and then 50 mg/day DHEA in the second week and finally, these were administered jointly with 500 mg/day metformin in the third week. By the fourth week, all doses were individualized based on particular response of each participant [186]. This treatment led to the improved immunological response and risk indices and reversed the epigenetic clock. The rate of reversal of the epigenetic aging relative to the actual chronological age increased from -1.6 year/year during the 0-9th months to -6.5 year/year in the 9-12th months of the study. This is the first report that the epigenetic age estimator of human lifespan can be reversed by an anti-aging strategy by drugs and hormones. Epigenetic age also responded to diet and life-style changes and dropped by an average of 1.96 years by the end of an 8-week treatment [187].

Rejuvenation by extracellular matrix (ECM)

ECM does not merely act as a scaffold, it actively participates in the regulation of cellular fate, behavior, and function. Moreover, to maintain tissue functionality, the tissue microenvironment may have rejuvenating or age inducing effects in the cellular repertoire of that tissue [161]. For example, ECMs that were deposited by the young fibroblasts derived from either newborn or a 6-year-old boy, restored the functions that were lost in senescent fibroblasts [188]. Exposure of senescent fibroblasts to young ECM reduced the level of the intracellular ROS levels and senescent markers such as β-gal, and inhibitors of proliferation namely, p21Waf1, p16INK4a, p53 and caveolin-1. The treatment also led to the telomere extension, reduced the level of DNA breaks, increased mitochondrial membrane potential (ΔΨm) and restored a youthful morphology. Restored cells regained the telomere related DNA repair protein dimer, Ku, in a SIRT1 dependent manner, and started to proliferate within 7 days. In contrast to senescent cells that fail to respond to stimulation by epidermal growth factor (EGF), the rejuvenated cells regained their EGF responsiveness and exhibited phosphorylation of signal-regulated kinase (ERK) in response to EGF [188].

Together, these findings support the idea that there are rejuvenating factors [rejuvenins) in young organsisms that are lost upon aging and there are humoral factors that lead to a diseased landscape. The properties that can be gleaned from the available data and ascribed to rejuvenins and geriatrins include effects on various cell and clinical parameters that determine a healthy or aging status (Tables 1-2).

Rejuvenins and geriatrins

We have adopted the terms, rejuvenins and geriatrins, for small endogenously produced factors, that circulate blood and are released from bone marrow, stem cell niches, ECM, muscle, nervous system, or other sites. Rejuvenins have rejuvenating and geriatrins exhibit age inducing effects. We use increase in health-span and lifespan as primary and other features of health and aging as secondary criteria to choose a host of rejuvenins and geriatrins discussed here. We discuss carnosine, polyamines, NAD+ and its precursors, melatonin, and alpha ketoglutarate (AKG) as rejuvenins and review the homocysteine (Hcys) and reactive oxygen species (ROS) as geriatrins.

Support for existence of rejuvenating factors that circulate blood is recently provided by the administration of young plasma to old rats that more than halved the epigenetic age of their blood, heart, and liver tissue and reduced senescence in various tissues [189]. Such studies confirmed that rejuvenins can reset the peripheral epigenetic clocks. Existence of circulating factors that can pass the Blood-Brain Barrier (BBB) was shown by heterochronic parabiosis experiments since a statistically significant rejuvenation also occurs in the brain of heterochronic parabionts, manifested by increased density of the dendritic spine of mature neurons and synaptic plasticity in the hippocampus [161, 162, 172]. In similar pairings, the old parabionts showed an increase in volume and blood flow in cerebral blood vessels, a higher self-renewal and differentiation of neural stem cell population in subventricular zone and exhibited improvement in olfactory discrimination [190]. Consistent with these findings, administration of plasma from young mice into aged mice reversed cognitive decline, including contextual fear conditioning and spatial learning and memory [162]. Thus, entry of rejuvenins to the brain ensures that the epigenetic clocks in the brain and the rest of the body are in tune and synchronized in terms of their tick rate. The idea that rejuvenins or geriatrins must pass BBB, restricts the type of molecules that have age altering property to small soluble factors. The size of molecules that pass the BBB, is estimated to be as small as 100-400 dalton (Da) to a maximum of 1000 Da. Small molecules generally cross the BBB in pharmacologically significant amounts only if the molecular mass of the drug is less than 400-500 Da, and the compound forms less than 8-10 hydrogen bonds with solvent water [192]. To modify the cell function, they must also be sufficiently small that either can pass the plasma membrane freely or enter the cell by transporters that exist within cell membrane [193]. In fact, the molecular weights of our chosen candidates all fall below the maximum of 1000 Da (Table 3). If any of these factors is a metabolite, then, the predicted molecular weight is estimated to be less than 1500 Da [194-196]. However, large polypeptides and proteins due to their size, charge and reduced mobility are unlikely to act as a rejuvenin or geriatrin.

Table 3.

Molecular mass and chemical composition of rejuvenating factors.

graphic file with name AD-13-6-1664-g3.jpg

Rejuvenation is short-lived and continuous release of rejuvenating factors is required to maintain a youthful phenotype and their gradual loss can progressively result in the development of an aging phenotype. In other words, rejuvenation is plastic, requiring active maintenance, and withdrawal of rejuvenins promotes aging. Thus, aging might be equivalent to atrophy which occurs when the trophic factors are withdrawn with the exception that geriatrins further exacerbate the impact of rejuvenin withdrawal (Fig. 1B). OKSM factors also are required to be continuously administered since only days after their withdrawal, their rejuvenating activity ceases [156]. In a rejuvenation trial in humans that were treated with rGH, metformin and DHEA, consistent with the transient nature of rejuvenation requiring active maintenance, when the treatment was discontinued, the GrimAge predictor of human morbidity and mortality persisted only for six more months [186]. Similarly, the aging phenotype also appears to require constant maintenance. For example, continuous NF-κB activity is sufficient to induce, in the mouse skin, a gene expression pattern that is reminiscent of those induced by aging [183]. More importantly, blocking NF-κB activity reverses many of the characteristics of aging in skin of old animals. Thus, continuous activity of NF-κB is required to actively maintain the aging phenotype and withdrawal of such a blockade is sufficient to erase the aging phenotype rendered by continuous NF-κB activity [183]. This suggests that continuous presence of geriatrins that induce chronic NF-κB activation and nuclear translocation would be required for sustaining the inflammaging. The fragile nature of youth and aging requiring active maintenance is also evident in experiments that used young ECM. The proliferation of rejuvenated old cells that were deposited on young ECM was arrested after approximately 25 population doublings, and these cells developed senescence as evident by flattened and enlarged cell shapes and high SA-β-gal activity showing that youth must be maintained, and that loss of youthful factors permits a down-hill fall into the abyss of an aging phenotype [188]. These findings support the idea that the significant fall in serum levels of all rejuvenins with aging and coordinate increase in geriatrins is likely permissive for aging to be settled (Fig. 1).

Rejuvenins are expected to show a broad range of rejuvenating properties that reverse those functions that deteriorate with age. The age induced decline includes loss of cell and organellar fitness and cell functions, homeostasis, frailty, loss of cognitive functions, increase in blood pressure, disturbed glucose tolerance, altered barrier integrity, eroded functional omics, decreased proliferation responsive to local needs, attrition of telomeric length, decreased bioenergetics, oxidative and ER stress or DNA damage responses, reduced or loss of repair mechanisms, inflammatory signaling, senescence and increase in epigenetic age (Fig. 1, Tables 1-2).

Both circadian clock and methyl cycle evolved early in evolution in species as diverse as unicellular algae to humans [197]. Circadian clocks and methyl cycles are tightly linked. For example, circadian rhythms modify the global DNA methylation daily whereas inhibition of methyl cycle disrupts these rhythms [197]. Aging leads to progressive erosion of robustness of circadian rhythms and by progressive accumulation of cytosine modifications in DNA, aging likely modifies global and site-specific DNA methylations (Fig. 1) [115, 116]. This age induced loss of maintenance of the DNA methylation might be caused by the activity of TET in open chromatin, enhancers and CpGs which show dynamic circadian oscillations [148]. Thus, the rejuvenating effect of rejuvenins might, at least, partially, be due to their impact on regulation of circadian rhythms, epigenetic modifications, or both. For example, the secretion of melatonin is rhythmic, and its nocturnal release allows it to have rhythmic effect on circadian rhythms, sleep, and body temperature. Moreover, melatonin exerts effects on DNA methylations that are opposite of those which are inducible by light [198]. It appears that melatonin exerts its effect on methylation and regulates epigenome by inhibition of DNA methyltransferase [199]. Similarly, the levels of enzymes that participate in generation of polyamines and their levels undergo circadian oscillations. Polyamines, in turn, adjust the circadian period and their loss leads to longer periods [200]. Thus, rejuvenins are expected to regulate and oppose the changes in methylome, and circadian rhythms that occur with age, and should potentially be able to reset the epigenetic age and extend health-span and lifespan.

Although the rejuvenin and geriatrin candidates that we discuss here, fulfill many of expected requirements, they do not exhibit identical properties, because data on some of their effects are still required (Tables 4-8). Below, we briefly discuss the salient rejuvenating features of some rejuvenins (carnosine, polyamines, NAD+, melatonin, AKG) and although there are other examples of geriatrins such as iron, for the purpose of brevity, we will discuss only two geriatrins (homocysteine, reactive oxgen species) [330-331]. Dyshomeostasis of iron in aging is associated with neuroinflammation, abnormal protein aggregation, neurodegeneration, and neuro-behavioral deficits [330].

Table 4.

Restoration of cell responses by treatment with Carnosine, NAD+, polyamines, and melatonin.

Cell Response Carnosine NAD+ Polyamines Melatonin
Genome and epigenome
DNA methylation
Histone acetylation
Telomere
Telomere shortening
Mitochondria
Mitochondrial biogenesis
Mitochondrial function
Bioenergetics
ΔΨm
ATP *
TCA cycle
Omics
Transcriptome
Acetylome
Proteome
Metabolome
Housekeeping Function
Ubiquitin Proteasome System *
Autophagy/Mitophagy ↑↓
ER
Stress
Cell fitness
Circadian rhythms
Proliferation
Population doublings
Restoration of lost functions
Cell Signaling
Damage
ROS stress
DNA damage
Oxidative damage
Senescence
Sirtuin activation
Cell damage and death
Repair
DNA repair
PARP activity
Repair and regeneration
Inflammatory Response
Inflammatory signaling
NFκB activation and nuclear translocation
SASP *

↑ Improved or protected ↓ Deteriorated or reversed * Not consistent with rejuvenation

Table 8.

Clinical impact of chronic oxidative stress.

Item Effect Reference
Barrier Inegrity
•BBB dysfunction 303-304
Bone
•Bone mass 305
ECM
•ECM biosynthesis and organization 306
•ECM degradation 307
Energy and Metabolism
•Insulin resistance 308
•Appetite 309
•Adipose tissue dysfunction 310
GI tract
•Epithelial injury 311
Immune Response
•Immune response (immunosenescence) 312
•Inflammation 312
Liver
•Liver disease 313, 314
Musculoskeletal System
•Contractile function 315
Nervous System
•Memory 316
•Cognition 317
•Neurodegeneration 289
Reproductive System
•Fertility 318
Skin
•Pigmentation, wrinkles, aging 319
Stem Cells and Tissue Regeneration
•Stem cell function 320
•Stem cell senescence 321
Senescence
•Senescence 322-324
Urinary tract
•Kideny pathology 325
Disease risk
•Aging 323, 326, 327
•Disease 289, 328
•Cancer 328
Health-span and Life-span
•Health-span 323, 326
•Lifespan 329

↑ Increased ↓ Decreased

Carnosine

Carnosine is a small di-peptide (AH or β-alanyl-l-histidine) that was first discovered in 1900 by V.S. Gulewitch [332] (Fig. 3A). Carnosine is synthesized from β-alanine and L-histidine, by carnosine synthase (ATPGD1) which is present mainly in the heart and skeletal muscle, olfactory bulb and epithelium and olfactory receptor neurons and oligodendrocytes [333-335]. Carnosine is widely distributed in the body and is present in excitable tissues, most notably, the heart, the kidney, spleen, nervous system, olfactory bulb, choroid plexus, cerebral cortex, plasma, and CSF [332, 336]. Skeletal muscle is a depot and most prominent site for production of carnosine that releases it during exercise into the tissue interstitium [337].

Figure 3.

Figure 3.

Structure of carnosin and polyamins and effects of spermidine. (A) Molecular structure of carnosine. (B) Molecular structure of polyamines. (C) Cellular and clinical effects of spermidine.

Following synthesis, carnosine undergoes modifications including methylation, acetylation and decarboyxlation. A methylated variant of carnosine, either anserine (β-alanyl-Npi-methyl-histidine) or ophidine/balenine (β-alanyl-Ntau-methyl-histidine) are found in most animals but not in humans [332]. The production of methyl-anserine involves transfer of a methyl group from S-Adenosyl-Methionine (SAM) by N-methyltransferase (CMT) or by direct condensation of β-alanine with methyl-L-histidine [333-334]. The ophidine is produced by the enzymatic condensation of β-alanine with Npi-methyl-histine (or Ntau-methyl-histidine) by carnosine synthase. Acetylated form of carnosine (acetyl-carnosine) is produced by substitution of L-histidine with histamine. There are also different variants of carnosine that include homo-carnosine, and carnicine which is produced by the decarboxylation of carnosine [332].

Carnosine and its methylated analogs are transported through the cellular membrane by the proton-coupled oligo-peptide transporters (POT-family or SLC15). The mammalian family members include PEPT1 and PEPT2 (oligo-peptide transporter 1 and 2) and PHT1 and PHT2 (peptide-histidine transporter 1 and 2) [338]. Besides carnosine, the PEPT1 and PEPT2, transport 400 di-peptides and 8,000 tripeptides, however, these members do not transport amino-acids or longer peptides. In contrast, the PHTs, in addition to di-peptides and trans-peptides can transfer L-histidine [339-340]. Carnosine is ultimately degraded by serum carnosinase (CN1) and by tissue carnosinase (CN2) or by a non-specific cytosolic di-peptidase [341-343].

Carnosine is a major player of health and is essential to cellular homeostasis by protection from modifications of a large array of cellular functions that erode with aging (Tables 4-5). Carnosine and anserine are anti-fatigue agents, which, in honor of the discovery of the effect by Severin, is named the “Severin's phenomenon”. Carnosine protects muscles from fatigue during rhythmic nerve-stimulated muscle contractions by acting as a pH buffer. By such an action, carnosine limits the extent of blood acidosis that results from excess lactic acid which is produced by glycolysis during persistent muscle contractions (Boldyrev, 2013).

Table 5.

Restoration of clinical responses by treatment with Carnosine, NAD+, polyamines, and melatonin.

Clinical Response Carnosine NAD+ Polyamines Melatonin
Blood
•Blood and serum profile
•Blood pressure
•Bone marrow function
ECM
•ECM biosynthesis
•ECM degradation
Energy and Metabolism
•Glucose tolerance *
•Energy
Immune Response
•Immune response
•Inflammatory signaling
Liver
•Liver metabolism
Musculo-skeletal System
•Bone mass
•Muscle mass
Nervous System
•Cognition
•Memory
•Neural plasticity
•Neuroprotection
•Sleep
Reproductive System
•Fertility
Stem Cells and Tissue Regeneration
•Stem cell function
•Tissue regeneration
Organ Protection
•Organ protection
Senescence
•Senescence
Age-related Disorders
•Age-related disorders
Disease
•Risk of disease
Cancer
•Risk of cancer

↑ Improved or protected ↓ Deteriorated or reversed * Not consistent with rejuvenation

Carnosine is protective of cell and tissue functions, effects that are gradually diminished as concentration of muscle carnosine and its serum levels decline with age or in age-related diseases such as Alzheimer’s disease (AD) [216, 217, 218, 220, 344-348] (Fig. 4A). However, such a drop can be compensated for by taking carnosine as a supplement since carnosine from food sources is absorbed by intestinal absorption by peptide transporter, PEP1 and its levels in blood is maintained by re-adsorbtion from renal tubules by PEP2 [332, 349-350]. In healthy volunteers, the daily ingestion of L-carnosine at 13 grams per day for 4 weeks elevated the muscle carnosine content by 65% or by 80-85% when taken orally for 10-12 weeks [332].

Figure 4.

Figure 4.

Day and age-related changes in the levels of rejuvenins. (A) Serum levels of NAD+, ATP and carnosine throughout life (349-353). (B) Serum levels of spermine and spermidine throughout life (404). (C) Serum levels of melatonin throughout life (499). (D) Serum levels of melatonin throughout a single circadian rhythm (354-355). (E) Plasma levels of Hcys throughout life in males and females (356). F. Plasma levels of Hcys during a 24 hr cycle (357).

The wide range of biological activities of carnosine is attributable to its property in resetting the clock genes, (Bmal1, Dec1, Cry1). Carnosine also accelerates the resetting rate of the myocardial clocks by virtue of acting on the autonomic nervous system [351]. Carnosine influences the transcription of the pyruvate dehydrogenase kinase 4 (PDK4), an action that requires epigenetic regulation of PDK4 through enhanced histone acetylation of its promoter [352]. In eukaryotes, PDKs (PDK1-4) regulate the pyruvate dehydrogenase complex in the mitochondrial matrix that converts the pyruvate derived from glycolysis in the cytosol to acetyl-coenzyme A, that its oxidation by the Krebs cycle, produces energy in the mitochondria [361]. Carnosine leads to de-acetylations which are mediated by sirtuins [353].

Carnosine protects against ROS induced damage since it is an antioxidant, and defends the tissues against oxidative stress, peroxynitrite-dependent reaction, hypochlorite (HOCl) and ischemia-reperfusion damage in multiple organs such as brain, kidney and testis and regulates the blood pressure and systemic glucose levels [332]. Carnosine, at physiological concentrations, directly reacts with superoxide anions like superoxide dismutase (SOD) and quenches peroxyl radicals, O2·-, and ROS such as hydroxyl radicals (·OH) [354, 355].

The anti-aging mechanisms of carnosine are attributed to its suppression of protein oxidation, glycation, AGE formation, and cross-linking, reducing the effects of reactive carbonyls, modulating mitochondrial function and autophagy, inhibiting the cellular respiration regulatory complex, and by acting as natural antioxidant, anti-inflammatory, and anti-senescence agent [356-360] (Tables 3-4). Carnosine prevents the age induced glycation of proteins that alters protein structure and decreases their biological activity. Carnosine prevents glycation and enhances the thermal un-folding of glycated proteins as validated by a decreased T (0.5) and a lowered Gibbs free energy barrier and enthalpy of denaturation, consistent with the idea that carnosine promotes hydration during heat denaturation of glycated proteins [361].

Carnosine protects tissues against protein carbonylation and glyco-oxidation, preventing the formation of advanced lipo-oxidation end-products (ALEs) and advanced glyco-oxidation end-products (AGEs) which are involved in oxidative damage in age-related pathologies including those which occur in diabetes, atherosclerosis, and Alzheimer’ disease. Carnosine reacts and quenches in vitro and in vivo, α, β-unsaturated aldehydes, such as HNE, the most abundant class of ALE precursors [332]. Carnosine, provided as supplement, reduces protein carbonylation in the kidney, lowers the level of urinary AGEs, and diminishes the collagen cross linking as evident by an increase in collagen solubility [362]. By reversal of protein glycation and inhibition of formation of AGEs and ALEs, carnosine prevents and reduces diabetic complications such as diabetic neuropathy and abnormal sensory perception. Carnosine also contributes to the removal of damaged proteins by acceleration of protein turnover by a rapid step involving a localized thermal un-folding, and then a slower proteolytic event [361, 363].

One of the age-related pathologies is the development of cataract, that results from cross linking of the major structural lens protein, crystallin (predominantly α-crystallin) and formation of insoluble aggregates. Carnosine prevents the self-assembly of alpha-crystallin, into fibrillar structures, under mild denaturing conditions in vitro, dis-aggregates glycated α-crystallin, ensures the lens transparency, protects the lens from the development of cataract and delays the progression of cataract in diabetic rats [364-366]. Application of ophtalmic solution of N alpha-acetyl-carnosine (NAC), which is resistant to the enzymatic hydrolysis by human serum carnosinase, has shown improvement in visual acuity and glare sensitivity [367, 368].

Carnosine protects against diabetes by its anti-oxidative property, exerts glycemic control, reduces plasma glucose and hyperglycemia, by increasing the insulin levels and by preservation or increase in the number of β-cells in the pancreas [332]. The protecting effect of carnosine on myocardial fibers is due to increases in neutral (non-lysosomal) protease activity [369-370]. In cardiovascular system, carnosine enhances cardiac contractility and shows anti-hypertensive and hypotensive effect, likely by vasodilation, through carnosine-histidine-histamine pathway, or NO-cGMP mechanism, or directly by the modulation of the autonomic nervous system [332]. Carnosine protects kidneys from podocyte loss and from diabetic nephropathy and by reducing the expression of Bcl-2-associated X protein (bax) and cytochrome c protects glomeruli from apoptosis [371-373]. Carnosine also protects tissues from damage such as prevention of mucosal ulcerations or Helicobacter pylori induced gastritis and of age-related pathologies in various organs [332].

Carnosine has protective effects against AD as evidenced by inhibition of polymerization of β-amyloid, and the neurotoxicity of amyloid-β [374]. In 3xTg-AD mice, that develop a neuro-degenerative phenotype, that is similar to AD, carnosine reduced the hippocampal intra-neuronal accumulation of β-amyloid (Aβ) and fully restored mitochondrial dysfunctions [375]. Similarly, in transgenic AD mice, on a high fat diet, carnosine prevented cognitive decline [376]. In animal models of Parkinson’s disease, carnosine inhibited the oligomerization of α-synuclein and, by virtue of its anti-oxidative and anti-inflammatory protection of the striatum, prevented the development of Parkinson’s disease [377-379]. In a pilot study, it has been shown that carnosine (1.5 grams per day) increases the efficiency of DOPA therapy in patients with Parkinson's disease, decreases plasma protein carbonyls, increases SOD and improves clinical symptoms such as rigidity of hands and legs, leading to increased hand and leg movements [380].

Carnosine substantially improves the rate of wound healing by increasing cytokines and growth factors, collagen biosynthesis and fibroblast proliferation and increases the speed of healing of bleomycin-induced and irradiation-induced pulmonary wounds and in a model of type 2 diabetes [332].

Carnosine prevents senescence, induces rejuvenation, and has geroprotective effect, likely by its wide spectrum of actions, including protein carbonylation and degradation of damaged proteins [381-383]. Carnosine also reduces the damage to telomeres and their shortening rate in fibroblasts [384]. Carnosine, at 50 mM, increases plating efficiency, the population doublings (PDs) and Hayflick limit, reduces senescence, and rejuvenates late passages of fibroblasts, a phenotype that is reversed upon carnosine withdrawal [356]. Carnosine mimics the geroprotecting action of rapamycin by inhibition of Akt/mTOR/p70S6K signaling [358, 385-386]. Carnosine extended lifespan in Drosophila and increased the lifespan by 20% in senescence-accelerated mice—prone 1 (SAMP1), leading to an increase in the number of animals that reached old age. These changes were associated with a decrease in the level of thiobarbituric acid reactive substances (TBARS), monoamine oxidase B (MAO B), and Na/K-ATPase activity [387-389].

Carnosine has been shown in mice to inhibit tumor growth and mortality, likely by inhibition of glycolysis which is essential to cancer growth (Warburg’s effect) by depletion of metabolic intermediates (glyceraldehydes phosphate and dihydroxyacetone phosphate), carbonyl quenching and reduced generation of ATP [390-393].

Together, the available data show that a small dipeptide has sweeping rejuvenating effects. These effects are found in cellular biology in terms of significant improvement in cellular fitness and damage control and repair. The clinical impacts of carnosine ensure maintenance of a youthful phenotype, by promoting longer health-span and lifespan expected from a true rejuvenin (Tables 4-5).

Polyamines

Sperm and crystalline substances formed by semen were initially identified in 1678 by Antonie van Leeuwenhoek. These crystals were later shown to consist of a group of substances with multiple amine groups known as polyamines which include a diamine (putrescine; 1,4-diaminobutane), triamine (spermidine) and tetraamines (spermine and thermospermine) [394-395] (Fig. 3B). These natural polyamines are produced from ornithine by the action of ornithine decarboxylase (ODC) that converts it to putrescine which is then modified to spermidine by decarboxylation by S-adenosyl-L-methionine; (SPDSY). Spermine is produced by the action of spermidine synthase (SPMSY), and spermine synthase [396]. These polyamines are regulated by spermidine/spermine N1-acetyltransferase (SSAT) that causes their ubiqiutination and proteasomal degradation, an effect which can be blocked by the binding to polyamines [397].

Besides SSAT, other enzymes of the biosynthetic pathway of polyamines, all exhibit a rapid turnover ensuring that polyamines are produced in a time-limited fashion [397]. Moreover, the transcript levels of enzymes that participate in the biosynthesis of polyamines including ornithine decarboxylase 1 (Odc1), S-adenosylmethionine decarboxylase 1 (Adm1), and spermidine synthase (Srm), show circadian oscillations [398]. The hepatic circadian changes in the abundance of these enzymes as well as the circadian fluctuations of polyamines are lost in Per1-/-Per2-/- mice, that lack a functional circadian clock, showing that their circadian rhythms rely on Per. The level of the enzymes of the polyamine biosynthetic pathway starts to increase at ZT12, peaks at ZT16 and their levels drops significantly at ZT20 and ZT0 [399]. The daily oscillation of intracellular levels of polyamines has been confirmed in vitro in NIH/3T3 mouse fibroblasts and in vivo in the mouse liver [399]. In fact, coordinate with the levels of the enzymes participating in their synthesis (Odc1, Amd1, and Srm), the intracellular levels of putrescine and spermidine and not spermine increased and peaked at ZT16 [399]. The daily levels of enzymes that participate in polyamine synthesis are regulated through light driven rhythmic binding of BMAL1:CLOCK heterodimers to conserved DNA elements and in response to feeding. The polyamines, in turn, adjust the circadian period by regulating the interaction with and stabilizing PER2-CRY1 heterodimers and for this reason, their loss during aging leads to longer circadian periods, a deficit that can be adjusted with polyamine supplementation [200, 399]. Also, spermidine regulates BMAL1:CLOCK-dependent transcription by virtue of inhibiting acetyltransferase E1A binding protein, p300 (EP300) [400]. By acting as a calorie restriction (CR) mimetic, spermidine also induces the beneficial effects of CR such as activating autophagy [401]. These data show an intimate relationship that exists between the daily circadian oscillations and the spermidine/spermine synthetic pathway and their polyamine output. Loss of this delicate balance artificially or by aging unleashes adverse impacts that can be abrogated only by supplementation of the diet with polyamines.

Due to their polycationic nature, polyamines have a wide range of effects since they readily interact with negatively charged molecules, including DNA, RNA, and lipids (Fig. 3C). There is evidence that polyamines regulate DNA methylation, and their effects have the potential to reverse the impact of aging on DNA. High polyamine intake that elevated polyamine concentrations in whole blood, prevented the development of age-associated genome-wide DNA methylations and coordinately reduced the age-associated pathological changes, inflammatory state, and mortality and extended lifespan in aged mice [402-404] (Fig. 4). Polyamines override aberrant DNA methylations and other changes that are caused by inhibiting ODC including increased decarboxylated levels of S-adenosylmethionine and decrease in DNA methyltransferase activity [404]. By virtue of its inhibitory effect on histone H3 acetylation by histone acetyl transferases (HATs), polyamines reduce ROS load and oxidative stress [405].

Polyamines participate in tissue regeneration by halting translation, increased cell proliferation and growth and by virtue of enhancing regenerative potential [406-408]. Inhibition of polyamine synthesis leads to disturbed cell cycle progression, within a single cell cycle, first affecting S phase and then G1 and G2/M phases [410]. In the yeast, Schizosaccharomyces pombe, spermidine, at a minimum concentration of 10-6 M was found to be an absolute requirement for growth and cell cycle progression [409]. Three of six spermidine homologs supported cell growth in presence of a specific inhibitor of putrescine biosynthesis, alpha-difluoromethylornithine (DFMO) [407]. As compared to α-methyl-ornithine, which is a competitive inhibitor of ornithine decarboxylase, its irreversible inhibitor, α-Difluoro-methyl-ornithine, which more potently inhibits polyamine production, coordinately inhibits growth [411]. L-α-Difluoro-methyl-ornithine that depletes the polyamines, reduces protein synthesis by inhibiting translation and thus, it seems that both translation initiation and elongation are dependent on presence of polyamines [412]. Due to their importance to tissue regeneration, coinciding with increase in DNA synthesis, the uptake of putrescine and the biosynthesis of spermidine, were accelerated in the liver within 2 hours after partial hepatectomy. Spermidine also promoted proliferation in stem cells of hair follicles, leading to hair shaft elongation and prolonged hair growth phase (anagen) showing that the effect of polyamines on cell growth and regeneration is not tissue dependent [408].

Autophagy represents cellular self-cannibalism, by which, non-nuclear cellular parts are transferred to lysosomes for degradation, and recycling, facilitating renewal of damaged molecules, organelles, and cytoplasmic and plasma membrane constituents. Aging leads to the loss of autophagy and genetic inhibition of autophagy has been shown to induce degenerative changes that are reminiscent of age-related pathologies [413-415]. Thus, it is clear that maintenance of such an important housekeeping function is required for tissue homeostasis and in preventing aging and age-related decline in autophagy as well as decrease in polyamine levels [416, 417] (Fig. 4B). Polyamines regulate autophagy by phosphorylating AMPK, which sustains ATP levels and by regulation of MFN1, MFN2, DRP1, and COX IV and by actions that involve both acetylation and SIRT1 independent deacetylation events by inhibition of EP300 [396, 400, 418]. Spermidine promotes autophagy by increasing ATG5 protein and LC3B-II levels, and by decreasing p62 protein expression [419]. The loss of cardiac autophagy leads to impaired heart function. Spermidine exerts cardio-vascular protective effects by improving the mechano-elastical properties of cardiomyocytes, by autophagy, mitophagy and by decreased vessel stiffness and reducing the mechanical load on the heart by lowering of the blood pressure [420, 421]. The cardio-protective effects of spermidine, at least in part, seem to be due to enhanced autophagy since such effects were not seen in ATG5 deficient mice [422]. In the brain, both spermidine and spermine induce autophagy in senescence accelerated mouse-8 (SAMP8) mice and delay brain aging as evidenced by increased activity of superoxide dismutase (SOD) that scavenges superoxide anions and by reducing malondialdehyde (MDA) in aging brains that is generated by ROS induced degradation of poly-unsaturated fatty acids [396]. Moreover, spermidine maintains brain health in older individuals who experience subjective cognitive decline [423].

Polyamines have anti-inflammatory and anti-apoptotic effects, e.g., spermidine supplementation was shown to reduce accumulation of neurotoxic soluble Aβ in early as well as late stages of Alzheimer’s disease [396, 424, 425]. In thioacetamide (TAA)-induced acute liver injury, spermine exerted its anti-inflammatory action by inhibiting M1 polarization, while promoting M2 polarization of Kupffer cells (KCs), decreasing their STAT1 activation, reducing the levels of IL1-β, and iNOS, and increasing STAT6 activation. The anti-inflammatory effect of spermidine in LPS activated macrophages is evident by reducing nuclear translocation of phorphorylated form of p65, and decreased production of ROS, NO, PGE2, TNF-α and IL1-β [426]. Similarly, in LPS treated microglial cells, spermidine blocked the nuclear translocation of p65, production of NO, and expression of TNF-α and IL-6 [427]. In skin, the anti-inflammatory impact of polyamines was shown by reducing edema, NO, TNF-α and IL1-β in 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced dermal edema in mice [428]. In collagen-induced arthritis, spermidine was shown to reduce joint macrophage activation and inflammation in mice [429].

In the lungs, spermidine increased autophagy, reduced endoplasmic reticulum stress and cell death, and prevented bleomycin induced senescence as exemplified by decreased expression of p16 and p21, β-gal, apoptosis, and fibrosis [430]. Spermidine afforded protection, retained locomotor activity, and increased survival in Drosophila melanogaster from toxic levels of superoxide generator, Paraquat, an action that required intact autophagy since these effects were absent in flies that lacked ATG7 [431]. Spermidine supplementation afforded more resistance to heat and hydrogen peroxide in yeasts and reduced oxidative stress in mouse fibroblasts in a glutathione independent manner [407, 432]. Spermine protected cells from DNA damage induced by Fenton reaction and at concentrations ranging from 0.1 to 1 mM, spermidine reduced release of 8-oxo-7,8-dihydroguanine and 5-hydroxycytosine after radiation and protected cells from radiation induced DNA damage [433, 434]. Spermidine, reduced retinal ganglion cell (RGC) death, prevented retinal degeneration and augmented optic nerve regeneration after optic nerve damage [435, 436]. Spermidine stabilized mitochondrial genome and membrane potential, reduced apoptosis and protected cells against D-galactose (gal)-induced mitochondrial damage [437].

Polyamines protect cells and organs against stress, prevent kidney from the damaging effect of diabetes, and reduce bone loss [438, 439]. In spermine oxidase mutant (Δspe3 Δfms1) strain of Saccharomyces cerevisiae, supplementation with spermidine or spermine led to the induction of transport-related genes and genes involved in methionine, arginine, lysine, NAD and biotin biosynthesis and down-regulated genes which are involved in nucleic acid metabolism and various stress responses [410]. Polyamines promoted wound healing through urokinase-type plasminogen activator (uPA)/uPA receptor (uPAR) signaling in vitro and in a skin wound repair in vivo [440].

Spermidine increases the lifespan of human peripheral blood mononuclear cells (PBMC) [405]. After 12 days in culture, only 15% of control PBMC survived, whereas up to 50% of the cells survived after treatment with 20 nM spermidine [405]. Similarly, supplementation of food with polyamines successfully increased the concentration of polyamines in blood, reduced the pro-inflammatory state that is typical of aged tissues [402]. This approach also prevented global alterations in DNA methylation, reduced age-related pathologies and decreased mortality in aged mice. The polyamine supplementation also increased lifespan and prevented liver fibrosis and development of hepatocellular carcinoma by activating MAP1S-mediated autophagy [402, 441]. In humans, high spermidine intake was associated with a lower rate of mortality and polyamine supplementation was shown to prolong health-span and lifespan in yeasts, flies, worms, to humans [402, 405, 431, 442, 443]. Polyamine depletion inhibited growth and caused cellular arrest in G1 phase, an event which was associated with increased p53 protein and other inhibitors including p21Waf1/Cip1 and p27Kip1. The treatment, within five hours, also led to up to 150% increase in the stress-activated protein kinase/c-Jun NH2-terminal kinase (JNK) type of mitogen-activated protein kinase (MAPK) and caused a sustained increase in the activity of ERK-2 isoform [444]. These findings suggest that the prolonged life in Nona-Centenarians, at least to some extent, might be owed to the high levels of polymaines in their blood [445].

The effects of spermidine on aging appears to depend on insuring DNA stability, fostering autophagy, promoting cellular fitness, cell growth and tissue regeneration, enhancing adipogenesis, reducing apoptosis, protecting against organ mal-function, and in enhancing anticancer immune surveillance [442]. The effectd of polyamines are ultimately translated to reducing age-related decline by promoting health-span and lifespan [431].

NAD+ and its precursors

NAD+ is a central and essential metabolite and an important co-factor in all living cells. For this reason, the NAD+ synthesis and degradation are exquisitely controlled. In mammals, NAD+ is synthesized from one or more of its major precursors including its principal substrate, nicotinamide (NAM), as well as tryptophan (Trp), nicotinic acid (NA), nicotinamide mononucleotide (NMN), and nicotinamide riboside (NR) (Fig. 5A). NMN is synthesized from nicotinamide, by the rate-limiting enzyme, nicotinamide phosphoribosyl transferase (Nampt), and 5′-phosphoribosyl-1-pyrophosphate (PRPP), or in the salvage pathway generated, by NR kinases (NRKs), from NR involving a phosphorylation reaction which allows it to be converted to NAD+ by NMN adenylyl transferases (NMNATs) [446].

Figure 5.

Figure 5.

Biosynthesis of NAD+ (+ is superscripted) and homocysteine and radical oxygen species. (A) Biosynthetic pathways of NAD+, (B) Biosynthetic pathways of homocysteine, (C) Structure of Radical Oxygen Species (ROS).

NAD+ participates in numerous biological processes ranging from production of ATP via anaerobic glycolysis, OXPHOS, fatty acid β-oxidation, and tricarboxylic acid cycle metabolism (Krebs cycle). By virtue of engaging NAD+-dependent sirtuins, NAD+ regulates diverse cell signaling, gene expression profiles, and DNA damage repair [446]. NAD+ is essential to metabolism and induces expression of metabolic genes through its action on circadian clock [447]. NAD+ is required for the actions of poly (ADP-ribose) polymerase proteins (PARPs), namely PARP1 and PARP2 in sensing and responding to DNA damage which involves protein de-acetylation and poly-ADP-ribosylation (PARylation) [448-450]. In C. elegans, increase in levels of NAD+ by PARP inhibitors, activated the mitochondrial unfolded protein response (UPRmt), which is a mitochondrial proteostasis pathway, known to promote longevity and improved mitochondrial homeostasis through the activation of the sirtuin homolog, sir-2.1 [451-453]. The increase in NAD+ also activates the pro-longevity, FOXO transcription factor (daf-16), which engages antioxidant protection programs [454, 455].

It has been shown that aging leads to decreased levels of ATP and NAD+ in tissues and serum in C. elegans, to mice and humans [456-461] (Fig. 4A). Besides, aging and diseases, metabolic disorders, cancer, and neurodegenerative diseases also reduce intracellular NAD+ levels by reducing its synthesis or by increasing its consumption. Such a decrease reduces lifespan in C elegans whereas the genetic increase in NAD+ or restoration of its levels prevents metabolic changes that are associated with aging, leading to prolonged lifespan in C. elegans [455, 462]. The administration of NAD+ precursors, such as NAM, NMN or NR is an efficient way to substitute the lowered levels of NAD+ that occur with age [463, 464]. Long-term oral administration of NMN (up to 300 mg/kg), without inducing any toxicity, increases NAD+ in various peripheral tissues including brain tissues since NMN readily passes the BBB [465-467].

Aging is associated with disturbed circadian rhythms and sleep-wake cycles. Supplementation with NR erases such defects by inhibition of PER2 by its deacetylation at lysine 680 and its subsequent phosphorylation that regulates its entry into the nucleus [447]. Restoration of NAD+ to its youthful levels by supplementation with NR, reverses the age-related decrease in chromatin binding by BMAL1 which is required for oscillations of transcription, mitochondrial respiration and increased late evening activity [447].

In humans, administration of NR in 400 mg/kg produced dose-dependent increases in the blood levels of NAD+, and its metabolome, without inducing any adverse effect [468, 469]. NAM supplementation, protects the heart and brain against ischemia-induced damage, prevents age and high fat diet induced DNA damage, inhibits formation of glucoma, and restores diet-induced hepato-steatosis and glucagon storage in age-matched mice [470-473]. NR supplementation also reverses progressive muscle dysfunction and loss of muscle fiber integrity which occurs in old mice as a result of diminished NAD+ levels [468].

Administration of NMN maintains lipid and energy metabolism, increases insulin sensitivity, counteracts aberrant glucose tolerance, and immune functions, maintains bone density and affords protection in animals against age-associated functional decline [474, 475]. Similarly, NR improves glucose tolerance, and in models of diabetes and high-fat diet, improves metabolic function, prevents obesity, and reduces fat deposition [455, 476-479]. NMN supplementation reduces oxidative stress and inflammatory responses. NMN also maintains the population of neural stems and progenitor cells, prevents synaptic loss and protects aged mice against neuronal cell death, neurodegeneration and pathological damage in Alzheimer’s disease associated with Aβ, and the associated decline in the cognitive function [455, 488-490]. Similarly, NR supplementation delays the decline in cognitive function, improves learning and memory, maintains motor functions, and reduces neuronal cell death in animal models of AD and Parkinson's disease (PD) and prevents the development and progression of Aβ pathology in AD mice and C. elegans [484-487].

NMN supplementation provides protection against premature aging and extends lifespan and health-span [465, 488-490]. NAD+ dependent increase in health-span has been shown in normal aging mice, Nampt+/- mice, β cell-specific Sirt1-overexpressing (BESTO) mice, hypomorphic BubR1 (a mitotic check-point kinase) mice, mice with age or diet-induced diabetes, and glucoma-prone mice [472-474, 491- 493]. NR prevents MuSC senescence in the mdx (C57BL/10ScSn-Dmdmdx/J) mouse model of muscular dystrophy, prevents the senescence of neural and melanocyte SCs, protects against aging and diseases and increases lifespan and health-span in many model systems [455, 476, 477, 494-496].

Thus, the available evidence shows that NAD+, is not only required in a large array of cellular activities, it has many rejuvenating features that promote a longer life-span and health-span. Replenishing reduced levels of NAD+ in aging by administration of NAM, NMN or NR restores homeostasis and provides protection against aging and age-related declines.

Melatonin

Melatonin has a a wide range of health promoting effects including improving cell fitness and providing cell protection and survival [497]. Particularly, because of its nocturnal production by pineal gland which is controlled by light-dark cycles, melatonin participates in circadian oscillations [196] (Tables 4-5). The nightly increase in melatonin in humans starts at the 6th to 8th week of life, reaches its peak levels in 4th-7th year of life. Moreover, its peak night levels progressively fall so that by the 7th decade of life, there is no appreciable difference between the day and night-time levels [498-502] (Fig. 4C-D). Rhythmic secretion of melatonin at night defines biological night and endows this molecule to have rhythmic effect on a wide range of cell and body responses. Melatonin regulates sleep, circadian body temperature, and circadian rhythms [195, 196]. Light drives non-visual responses, including phase shifting the internal circadian clock, and increasing alertness, and heart rate. Melatonin also causes phase shifting of human circadian rhythms and protects against phase shifts which are artificially introduced by night-time exposure to light [195]. The therapeutic efficacy of melatonin hinges on increasing the dietary melatonin intake, sometimes using it at supra-physiological dosages [503]. The phase shift in the human circadian rhythms can be achieved by appropriate timing of administration of exogenous melatonin and light for the successful treatment of sleep disorders resulting from night shift work, circadian sleep disorders as well as jet lag induced by travel across substantially different time zones [195].

Melatonin increases mitochondrial respiratory efficiency, improves mitophagy, promotes mitochondrial biogenesis, and has been shown to protect mitochondria in the senescence-accelerated mouse strain, SAMP8 [504]. Melatonin enters mitochondria where it regulates the mitochondrial bioenergetic function. Melatonin increases the mitochondrial mass, mtDNA copy numbers and the energy supply, while decreasing the oxygen consumption. Melatonin protects mitochondria against DNA damage including those that occur as a result of intra-mitochondrial Aβ aggregates that accumulate in Alzheimer’s disease (AD) [505-508).

Melatonin provides resistance to oxidative stress by its antioxidant effect in countering generation of reactive oxygen species, and stimulation of antioxidant enzymes including superoxide dismutase, glutathione peroxidase, glutathione reductase, and catalase [501, 507]. Being both lipophilic and hydrophilic, melatonin readily passes cellular membranes and provides protection against ROS induced damage in various cell organelles [501]. Meltaonin prevents oxidatively induced telomere attrition, reduces damage to bioinformational molecules, proteins and lipids, and organelles, reduces the mitotically inactive-DNA damaged cells and associated SASP phenotype, prevents apoptosis, while increasing proliferation in leukocytes, stem cells and progenitor cells [508-510]. Melatonin also reduces inflammatory signaling and the development of senescence and immunoscenescence [508]. The pro-survival effects of melatonin have been attributed to its regulation of SIRT1 [511].

Senescence was delayed and lifespan was prolonged when the pineal glands of young mice were grafted into old animals [498]. Clinical trials show that melatonin prevents cell damage, in such conditions as sepsis, metabolic and neuro-degenerative diseases, cancer, inflammation, and aging [503]. Melatonin reduced lipid peroxidation, restored the locomoter activity, and increased lifespan in Transgenic Knockdown Parkin Drosophila Melanogaster exposed to paraquat [512]. Chronic administration of melatonin improved mitochondrial function and increased lifespan in senescence prone (SAMP8) mice [513].

Melatonin is also used in adjuvant therapy for age-related disorders including diabetes, neuro-degenerative diseases such as Alzheimer’s disease, glaucoma, macular degeneration, arterial hypertension, and insomnia associated with aging [514]. Together, the available evidene show that melatonin prevents age-related declines and increases lifespan and health-span [508].

Alpha-ketoglutarate (AKG)

AKG, also referred to as 2-ketoglutaric acid, is an essential rate-determining intermediate in the Krebs cycle with a crucial role in cellular energy metabolism, protein, amino acid, mineral and lipid metabolism, and circadian patterns of lipid peroxidation [515-518]. AKG promotes higher protein and lower triacylglyceride levels in Drosophila [519). AKG also improves nitrogen balance, decreases protein catabolism and glutamine degradation, enhances protein synthesis, and for this reason, is used to reduce the protein catabolism during recovery from trauma, severe infection, burn, or surgery [520, 521]. AKG is a a mitochondrial redox sensor, that stabilizes redox homeostasis and affords protection against cyanogens, oxidative stress and ROS damage, and carbohydrate-induced cell death in Saccharomyces cerevisiae [522-527]. AKG modifies the metabolism by impacting levels of circulating hormones. Supplemention of the diet with AKG significantly increases circulating plasma levels of insulin, growth hormone (GH) and insulin like growth factor-1 (IGF-1) [528-529].

AKG maintains the pluripotency of the embryonic stem cells (ESC) and their NANOG expression and promotes the differentiation of primed human pluripotent cells [530-532]. Consistent with such a role in pluripotent cells, AKG significantly increases the number of inner cell mass (ICM) cells and promotes fetal growth [533].

AKG is neuroprotective and prevents induced motor dysfunction [534]. AKG significantly promotes proliferation and differentiation of mouse myoblasts, by impacting the glutamine metabolism, oxidative stress, and energy metabolism [535]. In the murine heart, AKG reduces pressure overload in chronic cardiac dysfunction and maintains elasticity of arterial wall in elderly mice [536, 537]. In middle aged mice, AKG causes adipose tissue rejuvenation by increasing the de-methylation and by increasing the expression of Prdm16 promoter, promoting beige adipogenesis, and reducing gain in body weight and obesity [538].

AKG is involved in collagen metabolism by acting as a cofactor of prolyl-4-hydroxylase (P4H) that catalyzes the formation of 4-hydroxyproline, a crucial step in the formation of the collagen triple helix, and by facilitating collagen synthesis by increasing proline derived from glutamate [516]. AKG increases procollagen production in human dermal fibroblasts and decreases formation of wrinkles which are inducible by UVB exposures in skin of hairless mice [528, 529, 539, 540]. AKG increases BMP, is pro-osteogenic, supports bone development in rats and lambs, increases bone mass, overcomes age-related osteoprosis and protects bone loss due to hormone deficiency caused by ovariectomy [541-546].

AKG improves epithelial recovery induced by stress, protects the epithelial metabolic transition, and prevents dextran sulfate sodium (DSS) induced colitis [547, 548]. In pigs challenged with endotoxin, AKG improves the integrity of intestinal mucosa, alleviates oxidative stress and injury in intestinal mucosal cells, and maintains the absorption of nutrients [549].

AKG regulates major histone demethylases including ten-eleven translocation (TET) hydroxylases and the Jumonji C domain containing lysine demethylases, which catalyze the removal of repressive and age-related histone methylation marks (H3K9me3 and H3K27me3), and decreases their accmulation [532, 546, 550, 551]. By regulation of histone methylation, AKG reverses age-related osteoporosis [532]. AKG delays age-related decline in fertility in mammals, and partially prevents the age-related decline in locomotor activity and cold tolerance in Drosophila [519, 552]. AKG delays age-related phenotypes including hypercholestrolemia, increases lifespan in Drosophila and worms and reduces age induced morbidity in mice [553-556]. AKG reduced frailty scores by 46% in females and by 41% in males with higher frailty score correlating negatively with life expectancy [556]. The mechanism of action of AKG appears to depend on reducing NFκB mediated inflammatory pathway, hence, reducing the level of inflammatory cytokines. AKG also reduces ATP synthesis, by binding and inhibiting ATP synthase, and by inhibition of oxygen consumption. Its further actions are directed at activating autophagy and by decreasing the activity of TOR pathway [554, 556, 557]. The impact of AKG supplementation on increasing lifespan might also be related to its pleiotropic action on metabolism, reducing oxidative stress, and by endowing stress resistance. In a sense, the administration of AKG mimics calorie restriction, which is known to prolong life [558].

Together, the available evidence shows that AKG promotes cell fitness, health, and homeostasis, offers cell and organ protection, prevents, and protects cells against damage and prolongs health-span and lifespan in animals. For these reasons, AKG has rejuvenating features expected from a rejuvenin.

Homocysteine

Homocysteine (Hcys) is a sulfur containing amino acid involved in one-carbon metabolism, that is derived from the essential amino acid, methionine via methionine cycle (Fig. 5B) Hcys is catabolized into cysteine through transsulfuration, a pathway that regulates cellular redox state by regulating levels of glutathione (GSH) and hydrogen sulfide (H2S) [559]. Hcys biosynthesis requires the participation of a host of enzymes including ATP-L-Methionine S-Adenosyltranferase (MAT), S-adenosylmethionine-dependent methyltransferases such as glycine N-methyltransferase, and S-adenosyl-homocysteine hydrolase. Homocysteine is converted by cystathionine β-synthase (CBS) to cystathionine which is coverted by cystathionine γ-lyase (CSE) to cysteine, ammonium and α-ketobutyrate. Homocysteine can be remethylated back to methionine by the participation of methionine synthase, methionine synthase reductase, vitamin B12, and 5-methyl-tetra-hydrofolate [560] (Fig. 5B).

Hcys is a methyl donor, and its functions are important to the synthesis of methylated factors, folate metabolism, choline catabolism and in regulating methionine activity [561]. Hcys is also a potent oxidant and in plasma, by rapid auto-oxidation, Hcys generates stong oxidative products including superoxide and hydrogen peroxide [223]. The normal levels of Hcys in humans is between 5-15 μmol/l, and an increase above such levels causes hyperhomocysteinemia and homocystinuria, conditions that are associated with a host of cellular deficits and health issues [562, 563] (Fig. 4E, Table 6). Hcys levels in mild, moderate, and severe forms of hyperhomocysteinemia are, respectively, in the range of 15-30, 30-100 and above 100 μmol/l [252, 564-566]. Hyperhomocysteinemia leads to ROS induced oxidative stress, ER stress, angio- and neurotoxicity, and inhibition of cross-linking of collagen (Table 6) [567]. Excess Hcys increases the level of its highly oxidant metabolite, homocysteine thiolactone (HTL). HTL is highly reactive with ε-NH2 groups of the lysinic residues of proteins and changes their structure, activity, and function by protein homo-cysteinylation [568-570].

Table 6.

Associations of hyperhomocysteinemia with cellular and clinical features of aging.

Barrier Integrity
•Impaired blood retinal barrier 201
Cell Degeneration and Death
•Apoptosis 202
•Neural apoptosis 203
•Apoptosis of mesenchymal stem cells 204
Damage
•Loss of function of cell membranes 205
•Impaired genergy metabolism 206
•Metabolome changes 207
•Abnormal lipid metabolism 208
•Alterations of serum VLDL 209
•Defective DNA synthesis 210
•Hypomethylation 211
•Neural DNA damage 212
•Inhibition of NO synthase (eNOS) expression 213
•Inactivation of eNOS by over-expression of caveolin-1 214
•Aberrant stem cell function 215
Housekeeping Function
•Decreased autophagy 216
•Altered Ubiquitin proteasome composition 217
Immune Response and Inflammation
•Induction of inflammation and inflammatory markers 218, 219
•Activation of NFκB 220
•Release of inflammatory cytokines 221
•Induction of monocyte chemoattractant protein-1 (MCP-1) 220
•Induction of IL-8, chemokines, RAGE, MMP9 and adhesion molecules 220, 222
•Monocyte proliferation 221
•Monocyte activation 221
•Immune activation 222
Stress
•Increase in ROS and oxidative stress 223, 224
•Lipid peroxidation 223
•Oxidative modification of low-density lipoproteins 225
•Oxidative degradation of endothelial membrane lipids 224
•Increased ER stress 226
Induction of a thrombotic environment
•Platelet activation 227
•Induction of expression of tissue factor 228
•Activation of factor V and VII 222, 229
•Reduced protein C activation 230
•Inactivation of expression of thrombomodulin 231
•Formation of thrombus 227
•Suppressing anticoagulant heparin sulfate expression 232
•Blocking tissue plasminogen activator binding to endothelial cells 233
•Depletion of endothelium-derived nitric oxide 203
•Stimulation of N-methyl-D-aspartate (NMDA) receptors, resulting in calcium influx and excitotoxicity 234
Bone
•Decreased bone blood flow 235
•Decreased formation of collagen cross links in bone 236
•Decreased bone mass 237
•Osteoporosis 238
Barrier Integrity
•Impaired blood retinal barrier 201
Cell Degeneration and Death
•Apoptosis 202
•Neural apoptosis 203
•Apoptosis of mesenchymal stem cells 204
Damage
•Loss of function of cell membranes 205
•Impaired genergy metabolism 206
•Metabolome changes 207
•Abnormal lipid metabolism 208
•Alterations of serum VLDL 209
•Defective DNA synthesis 210
•Hypomethylation 211
•Neural DNA damage 212
•Inhibition of NO synthase (eNOS) expression 213
•Inactivation of eNOS by over-expression of caveolin-1 214
•Aberrant stem cell function 215
Housekeeping Function
•Decreased autophagy 216
•Altered Ubiquitin proteasome composition 217
Immune Response and Inflammation
•Induction of inflammation and inflammatory markers 218, 219
•Activation of NFκB 220
•Release of inflammatory cytokines 221
•Induction of monocyte chemoattractant protein-1 (MCP-1) 220
•Induction of IL-8, chemokines, RAGE, MMP9 and adhesion molecules 220, 222
•Monocyte proliferation 221
•Monocyte activation 221
•Immune activation 222
Stress
•Increase in ROS and oxidative stress 223, 224
•Lipid peroxidation 223
•Oxidative modification of low-density lipoproteins 225
•Oxidative degradation of endothelial membrane lipids 224
•Increased ER stress 226
Induction of a thrombotic environment
•Platelet activation 227
•Induction of expression of tissue factor 228
•Activation of factor V and VII 222, 229
•Reduced protein C activation 230
•Inactivation of expression of thrombomodulin 231
•Formation of thrombus 227
•Suppressing anticoagulant heparin sulfate expression 232
•Blocking tissue plasminogen activator binding to endothelial cells 233
•Depletion of endothelium-derived nitric oxide 203
•Stimulation of N-methyl-D-aspartate (NMDA) receptors, resulting in calcium influx and excitotoxicity 234
Bone
•Decreased bone blood flow 235
•Decreased formation of collagen cross links in bone 236
•Decreased bone mass 237
•Osteoporosis 238
•Risk of osteoporotic fracture 239
Pancreas
•Type 2 diabetes 240
Muscle
•Loss of muscle mass and function 241
•Skeletal muscle atrophy 242
•Inhibition of satellite regeneration 243
Nervous System
•Aberrant sleep 244
•Neurotoxicity 245
•Enhanced glutamine excitotoxicity 246
•Enhanced β-amyloid neurotoxicity 247
•Tau hyperphosphorylation 248
•Cerebral cortical atrophy 249
•Hipocampal atrophy 249
•Disruption of frontal subcortical circuits 250
•Disturbance in gait balance 251
•Loss of motivation 252
•Cognitive decline 252
•Depression 252
•Dementia 252
Reproduction
•Recurrent pregnancy loss 253
Retina
•Retinopathy 254
Vessel Dysfunction and Disease
•Hypertension 255
•Angiotoxicity 246
•Endothelial dysfunction 222, 227
•Endothelial injury 203
•Vascular smooth muscle cell proliferation 256
•Depletion of endothelial dervied NO 203
•Vascular disease (arteriosclerotic changes) 257
•Cerebral micro-angiopathy 258
•Cerberal macro-angiopathy 258
•Proliferation of vascular smooth muscle cells 256
•Coronary heart disease 259
•Peripheral artery disease 260
•Homocysteine-induced arteriosclerosis 227
•Myocardial infarction 261
Senescence
•Induction of senescence 262, 263
Risk of Disease and Cancer
•Increased risk of degenerative disease 264
•Increased risk for cancer 265-266
Mortality
•Increased risk of death 267
•Increased risk of cardiac death 268

↑ Increased or deteriorated

Mildest forms of hyperhomocysteinemia occur by deficiency of folate and vitamin B12, and in the advanced stages of renal disease [560, 571]. Interestingly, the folate deficiency which leads to hyperhomocysteinemia, coordinately dampens PER2 and vasopressin circadian rhythms and reduces responsiveness to photic resetting of the liver clock in mice [572]. Moreover, the pharmacological inhibition or knockdown of cystathionine β-synthase (CBS), which is involved in metabolism of Hcys and its deficiency, leads to hyperhomocysteinemia, significantly increases the amplitude of oscillations and baseline expression of Per2, suggesting an intimate link between the systemic levels of Hcys and circadian rhythms [573].

The impact of excess Hcys that leads to hyperhomocysteinemia and homocystinuria due to inborn errors in metabolism were first described in 1962 [562, 574]. Ten percent of the world population suffers from hyperhomocysteinemia due to reduced activity of the folate-metabolizing enzyme, MTHFR, which is caused by homozygous C677T polymorphism of MTHFR gene [561]. One of the most common forms of error in Hcys metabolism occurs due to the deficiency of CBS that prevents the conversion of Hcys to cystathionine and leads to deficient re-methylation of Hcys to methionine [560). Patients with hyperhomocysteinemia are at risk of developing multi-system disorders including vascular disease, vessel occlusion, cerebral ischemia, thrombo-embolic events such as pulmonary embolism, myocardial infarction, pancreatitis, seizures, osteoporosis, and mental retardation [562, 575]. Persistent hyperhomocysteinemia promotes endothelial cell dysfunction, inflammation, atherogenesis, and development of a thrombophilic profile [561].

Such findings have helped pave the way to the understanding of the role of Hcys in aging. Aging and age-related diseases lead to the aberrant metabolism of Hcys and cause hyperhomocysteinemia [559]. Aging leads to the progressive increase in the plasma levels of Hcys with values reaching over 10 μmole/L, particularly in males over age 50 [252, 565] (Figure 4E). The prevalence of hyperhomocysteinemia is 5-10% but might be as high as 30% in individuals over 65 with levels reaching 40 µmol/L [566, 576, 577]. Even in old but healthy individuals, hyperhomocysteinemia can contribute to the regional or widespread cortical atrophy associated with cognitive decline [578]. In fact, hyperhomocysteinemia, is a known risk factor for age-related diseases and a strong contributor and risk factor for cardiovascular diseases, atherosclerosis, Alzheimer’s disease, and other dementias as well as Parkinson's disease [561, 579]. The most severe forms of age-related hyperhomocysteinemia are associated with multi-system disorders that lead to the decline in organ function, peripheral vascular disease, atherogenesis, coronary artery disease, stroke, cognitive impairment, dementia, depression, osteoporosis and osteoporotic fractures, skeletal muscle myopathy, inflammatory syndromes, and rheumatism [252, 561] (Tables 5-6).

Together, the available evidence show that Hcys levels rise with aging and that such a rise leads to a host of age-related disorders

Reactive Oxygen Species (ROS)

Denham Harman proposed oxidative stress as one of the most important drivers of aging [580, 581]. His hypothesis received further support when the superoxide dismutase (SOD), was identified as an enzyme that generates superoxide anions (O-2.) in vivo [582]. Identification of defense mechanisms against the oxidative stress and damage further boosted his original hypothesis. His idea was also compatible with the “rate of living” hypothesis, since it was shown that species with high metabolic rates, have faster aging rates and shortened lifespan, linking energy consumption to senescence and aging [583]. Ultimately, it was realized that higher respiration rates of mitochondria were the principal cause of production of reactive oxygen radicals, that were capable of inducing significant damage to cell constituents [584-587]. In mammalian cells, ROS are comprised of (.O2), Hydroxy Radical (.OH), Hydroxyl Ion (OH-), and Hydrogen Peroxide (H2O2). These species are generated within mitochondria by electron transport chain (ETC), through metabolism of peroxisomal fatty acid and xenobiotic compounds, mostly from plants, and by phagocytic and cytocidal release of hypochlorite as a “respiratory burst” for killing foreign pathogens [588] (Fig. 5C).

ROS are ubiquitous second messengers and although, low levels of ROS are required for normal signaling and cell function, the rate of generation of ROS progressively increases with age, until oxidative stress prevails, inducing widespread damage to cellular and organ functions [288, 589]. Interestingly, humans or species with long lives, have better antioxidant mechanisms that protect them from such damages [590]. In addition, aging leads to the loss of enzymes such as catalase and glutathione peroxidase which break down hydrogen peroxide, further compounding the oxidative damage [591]. Sustained increase in ROS and lowered level of protection against ROS, ultimately, leads to activation of inflammatory signaling including those inducible by early growth response protein-1 (Egr-1), activator protein 1 (AP-1) and NF-κB [592]. The final outcome of such oxidative and inflammatory damages are oxidized molecules such as proteins and polyunsaturated fatty acids, declined cell function such as those related to mtDNA mutation, decreased mtDNA copy number, and compromised bioenergetics, senescence, fibrosis, and cell death. Ultimately, dysfunction appears in virtually every organ such as heart, liver, brain, and kidneys.

Together, the available evidence shows that, although certain levels of ROS are required for normal cell function, any increase above such levels causes chronic oxidative stress that manifest in aging and in age-related diseases (Tables 7-8).

Table 7.

Cellular impact of chronic oxidative stress.

Item Effect Reference
DNA
•Integrity 269
•Global DNA methylation 270
•Site specific DNA Methylation ↑↓ 270
Histone modifications
• Global acetylation 271
•γH2AX 272
Telomere
•Telomere length 273-276
•Telomere dysfunction 277
Mitochondria
•Function and energy balance 278, 279
•ΔΨm 278
•NAD+ ↓↑ 280
•TCA cycle 282
•Glycolysis 282
Deregulated Signaling
•mTOR 283
•NFκB 284
Robustness of ER Stress Response
•Unfolded protein response (UPR) 285-286
Cell fitness
•Proliferation, regeneration and number of population doublings 273
•Cell signaling ↓↑ 287
Damage Response
•Oxidative-ROS 288
•DNA (single & double strand breaks) 272, 289
•Protein 289
•Lipid 289
•Cancer 290-291
Repair Response
•Cell and tissue repair 292-294
•PARP activity 295-296
Inflammatory Response
•Inflammatory signaling 297
Senescence
•Hallmarks of senescence 273, 288, 298, 299
•SASP 289
Stem cells
• Exhaustion 299-300
Cell Death
•Apoptosis 301-302
•Necrosis 288

↑ Increased ↓ Decreased

Discussion

Diverse lines of evidence show that youth and aging are plastic, requiring active maintenance. According to eco-centric hypothesis of aging, rejuvenation, and aging, are properties that emerge from the biologic impact of endogenous small molecules that circulate the blood, transit the basal laminae and BBB, enter the interstitium and ultimately pass the plasma membranes to initiate their rejuvenating or age inducing effect on cells. Most notably, parabiosis experiments show of existence of both rejuvenating (rejuvenin) and age inducing (geriatrin) factors. The labile nature and plasticity of youth and aging might be related to the diminishing levels of rejuvenins and escalating levels of geriatrins in the blood of aging organisms (Fig. 1). Consistent with such a hypothesis, whereas the centenerians do not suffer from a nocturnal loss in melatonin levels, all other aging individuals experience a flattened circadian profile for melatonin by the time they reach the 7th decade of life [502]. Similarly, in contrast to falling levels of spermidine in old individuals, healthy nonagenarians and centenerians do not suffer from the loss of optimal spermidine concentration in blood and have levels that match those found in younger individuals [445].

There is increasing evidence that rejuvenation without undesirable effects can be achieved by parabiosis pairings of young and old organisms or by injection of plasma from young organisms. However, it is unlikely that similar approaches can lead to routine clinical treatment of aging in humans. The current methods of genetic reprogramming carry the risk of compromising the genetic integrity due to the possibility of insertional mutagenesis. Moreover, iPSCs or reactivation of telomerase have limited clinical use because of fear of tumorigenesis. Thus, although it is desirable to reset the epigenetic clock to an earlier time-point, it is not advisable to set it to zero. Clearly, cyclic genetic reprogramming is the leading method of choice since it was free of side effects [153]. Yet, this approach does not yield itself to clinical use of OKSM factors for rejuvenation. A method that has the likelihood of success for rejuvenation is the chemical induction of pluripotency. The early methods for chemically induced pluripotent stem cells or CiPSc still required introduction of OCT4, making such approaches to be clinically impractical [593-596]. However, using a combination of seven small-molecule compounds, Hou et al described successful reprogramming in mouse somatic cells, at a frequency of up to 0.2%, suggesting that reprogramming can be achieved merely by chemical rather than genetic means [597]. Thus, search for rejuvenins can include screening for chemical reprogramming, and/or factors that are competent to induce pluripotency and can reset the epigenetic clock to an earlier time-point (Table 9). Search for geriatrins can include identifying the small endogenous molecules that cause NF-κB or mTOR activation and a phenotype and molecular signature that are consistent with aging or those factors that can induce acceleration of epigenetic clock [183].

Table 9.

The epigenetic and other conditions required for pluripotency.

Gene or epigenetic change Pluripotent cells and ESC Remark Reference
Methylation and Methyltransferases
Heterochromatin Reduction of heterochromatin promotes reprogramming. Heterochromatin represses gene expression. 598 599
Global DNA methylation (5mC) Naïve pluripotency is associated with global DNA hypomethylation DNA methylation silences somatic genes and chromatin remodeling during iPSC generation. DNA demethylation reactivates pluripotency genes, which are hypermethylated and silenced in somatic cells. Inhibitors of DNA methyltransferase improve the overall efficiency of the reprogramming. 600-603
H3K9me2 &3 A barrier to reprogramming. Reduction promotes reprogramming. H3K9me2 &3 cover only 4% of the hESC genome, but over 10% of the human fibroblast genome. Dominant chromatin feature at the differentially bound regions (DBR)s, heterochromatic regions enriched for H3K9me3 in Fibroblasts.Repressive on Gene Expression. Megabase domains of H3K9me3 impair OSKM binding and reprogramming. Knockdown of relevant methyltransferases allows OSKM binding and enhances reprogramming. H3K9 methylation restricts late reprogramming events 598, 604-606
DNMT1 Impedes conversion of pluripotent to omnipotent state transition. Totipotent zygote is essentially devoid of DNA methylation. DNA methylation is not required for pluripotency. Dnmt1 suppression leads to loss of DNA methylation on pluripotency-related genes and their expression is up-regulated, promoting pluripotency 607-609
EzH2 Presence is required for reprogramming Catalyzes histone H3 methylation at lysine 27. Inhibition of PRC1 (Bmi1, Ring1) and PRC2 components (Ezh2, Eed, Suz12) significantly decreases reprogramming efficiency. EZH2 null blastocysts fail to generate ESC lines. The reprogramming capacity of Ezh2-depleted ESCs was severely impaired and several genes were either not induced (NANOG, REX1, TERT) or only partially induced (CRIPTO, OCT4). Long-term removal of Ezh2 (Δ/Δ) also resulted in compromised reprogramming. Ezh2 mediated H3K27me3 activity facilitates human pluripotent reprogramming and maintenance of bivalent chromatin domains in Pluripotent cells. 610-611
SUV39H1 Negative regulation or knockdown promotes pluripotency Suv39h lysine methyltransferases (KMTs) generates H3K9me3 - a mark that is recognized by HP1 to maintain the heterochromatin 612
KAT7/HBO1 Dispensable for H4 acetylation
Demethylation and demethylases
5mhC 5hmC increases expression in ESCs and pluripotent cells. 5hmC promotes the demethylation of pluripotency genes during reprogramming. 5hmC enrichment is involved in the demethylation and reactivation of genes and regulator regions important to pluripotency. Promotes H3K36me2/3 demethylation in mouse embryonic fibroblasts in culture and during reprogramming. 598, 613, 614
KDM2A/JHDM1a Dispensable for pluripotency maintenance in ESCs The expression of pluripotency genes in Kdm2a-/-ESCs is downregulated and show defective lineage commitment upon differentiation. Regulates the expression of a subset of genes in ESCs. Promoters bound by Kdm2a are enriched in H3K4me3. Overexpression of potently enhances reprogramming, whereas knockdown impairs iPSC generation. 614-616
KDM2B/JHDM1b AKG dependent protein involved in self renewal. Kdm2b is a histone H3 Lys 36 dimethyl (H3K36me2)-specific demethylase, with the capacity to promote iPSC generation binds unmethylated CpG islands via a ZF-CXXC domain. Kdm2b is part of the PRC1.1 complex that mediates gene repression. Demethylases of histone H3K36 methylation. Increases gene expression and is required for generation of pluripotency. Overexpression of potently enhances reprogramming, whereas knockdown impairs iPSC generation. 598, 615, 616
Tet1, Tet2, TDG Indispensable for maintaining pluripotency. Expression enhances reprogramming. 5-methylcytosine modifiers that cause 5mC to change to 5hydroxymC (5hmc), then 5-formylcytosine (5fC) to 5-carboxylcytosine (5caC). 5hmC, 5fc, and 5caC are enriched at regions bound by pluripotency factors. Oxidative demethylation is required to promote gene activation, to cause mesenchymal to epithelial transition and reprogramming of fibroblasts to pluripotency. Tet1 can replace Oct4 to generate fully pluripotent iPSCs. Tet2 mediated 5mC to 5hmC conversion on NANOG occurs in OKSM reprogramming. Loss of TDG leads to a significant decrease in pluripotency. 598, 613, 617-619
Acetylation and acetyl transferases
H4ac Critical regulator of pluripotency in embryonic stem cells 599, 620-623
H3K9ac Increased H3K9ac predicts increased reprogramming to pluripotency. Enriched in mouse and human ESCs H3K9ac is enriched at gene promoters and highly correlates with gene expression. H3K9ac level correlates with binding of transcription factors and (HDAC) inhibitors (HDACi such as valproic acid) that increase H3K9ac levels, restore reprogramming capacity. H3K9ac deposition enhances Sox2 binding. Most genes with high acetylation levels are bound by c-Myc. Increase in H3K9ac and decrease in H3K27me3 in promoters causes an active chromatin state. 599, 603, 622, 623
KAT1/HAT1 Coordinately increases with OCT4 and NANOG at 1 cell, 2 cell stage and blastocysts. Acetylates histone H2A on lysine 5 624, 625
KAT6A and KAT6B Acetylate both histone H3 and non-histone proteins. KAT6A modulates Oct4 and Nanog binding to chromatin in ESC. 626, 627
KAT8/hMOF Induces Generation of Pluripotent Stem Cells MOF protein is dramatically upregulated following reprogramming/Kat8 plays key roles in embryonic stem cell core transcriptional network. 628, 629
p300/EP300 Proximal regulatory regions of pluripotency genes, such as OCT4 and Nanog shows H3K9ac Recruited and directly interacts with Nanog to induce gene activation of Nanog-targeted genes in mESC. 630
Deacetylation and deacetylases
HDAC1 Regulates and is essential to pluripotency. HDAC1 occupies critical genes involved in maintaining self-renewal of ES and TS cells. Binds predominantly to pluripotency genes (OCT4, SOX2, NANOG, KLF4). 630, 632
SIRT1 Required for deacetylation of Oct4 and maintenance of naive pluripotency. Oct4 is hyper-acetylated during the transition to pluripotency. SirT1-knockout reverses the induction of the primed pluripotency network. 633
SIRT6 Addition to Yamanaka factors improves reprogramming efficiency of HDFs Highly expressed in pluripotent cells. Regulates the efficiency of mouse somatic reprogramming. Binds to the promoters of pluripotency genes. A H3K56 deacetylase controls embryonic stem cell fate via TET-mediated production of 5-hydroxymethylcytosine. 634-637
Others
Lamin B1 638
FOXO3a An essential regulator of pluripotency in hESC. Contributes to reprogramming. 638, 639
NRF2/NFE2L2-KEAP1 Controls self renewal and pluripotency of hESC. NRF2 inhibition directly disrupts self-renewal of hESCs and cellular reprogramming, and Nrf2 activation delays differentiation of hESCs. 640
cMyc Prevents downregulation of pluripotent genes and plays crucial roles in generation of iPSC. Enhances the binding of the other pluripotency factors to chromatin. cMyc expression leads to efficient induction of iPSC. cMyc alone can keep the mESC cells in pluripotent state. 614, 641-643
AKG High level alters chromatin. Maintains pluripotency. A cofactor for several chromatin-modifying enzymes, including histone demethylases and the Tet family of enzymes that are involved in DNA demethylation. Increased gene expression 598 644
HIRA Dispensable for pluripotency in mESCs 645

ESC: embryonic stem cells. hESC: human embryonic stem cells, mESC: mouse embryonic stem cells, HDF: Human dermal fibroblast, iPSC: induced pluripotent stem cells, H4ac: Acetylated Histone 4, 5mC: 5 methyl cytosine, 5hmC: 5 hydroxy-methylcytosine

There is increasing evidence of the interplay between rejuvenins (NAD+, carnosine, spermidine, and melatonin), and geriatrins (Hcys). The idea that a geriatrin (Hcys) and a rejuvenin (NAD+), are metabolically linked, is clear from the fact that the NAD+ and Hcys biosynthesis are intertwined which directly links one-carbon metabolism and methylation balance to the levels of NAD+ [646]. Moreover, excess Hcys causes the depletion of NAD+ pools and significant fall in the NAD+ levels, likely due to an increase in the ROS since such a side effect could be prevented by radical scavengers [647]. Hyperhomocysteinemia is a risk factor in normal pregnancy. The administration of carnosine (100 mg/Kg) to pregnant rats that their pups were suffering from toxic effects of methionine induced prenatal hyper-homocysteinemia, restored the superoxide dismutase levels and prevented the cognitive decline and protected cerebellar neurons in the pubs from oxidative stress [648]. Carnosine prevented the oxidative stress in human erythrocytes that is induced by homocysteine and its by-product, homocysteic acid [649]. The suppression of liver S-adenosylmethionine which, leads to reduced spermidine levels, coordinately increased the plasma Hcys levels [650]. The key to the link between Hcys and melatonin, at least, to some extent, is related to the fact that melatonin synthesis requires methylation of acetyl-serotonin which is donated by S-adenosylmethionine and the by-product of this reaction, S-adenosylhomocysteine, is a substrate for homocysteine production [651]. In line with this, the levels of Hcys showed circadian rhythms and these oscillations are coordinated with the circadian melatonin release [652] (Fig. 4F). In addition, hyperhomocysteinemia, due to folate deficiency, is accompanied with decreased melatonin levels in the pituitary gland, and urine [651]. On the other hand, the removal of pineal gland, which reduces melatonin levels, leads to hyperhomocysteinuria [653]. Whereas pinealectomy did not change the circadian rhythms, it led to a substantial increase in mean plasma levels of Hcys [653]. Administration of melatonin to pinealectomized rats restored homocysteine to its normal levels and enhanced the antioxidant defense by raising the total glutathione levels [653]. Melatonin has been used to treat the adverse effects of a geriatrin (Hcys) in aged human population who suffer from hyperhomocysteinuria. The normal treatment for hyperhomocysteinuria is administration of folate (1 mg/day), vitamin B12 (400 µg/day) and vitamin B6 (10 mg/day), often, with mixed and not necessarily desirable results [654]. However, it was recently shown that melatonin reduces the oxidative stress induced by elevated levels of Hcys in nervous system and vessels [651]. Moreover, supplementation of melatonin to rats with chronic hyperhomocysteinemia, at a dose of 10 mg/kg, restored the decreased glutathione levels and significantly reduced the lipid peroxidation, and decline in cognition, learning and memory performance [655].

The key to the successful treatment of aging population is the parenteral rather than oral administration of rejuvenins since their absorption from gastrointestinal tract might be impeded by the degeneration of villi and decline in the number of nerve cells of the myenteric plexus in old individuals [656]. Moreover, the treatment should be tailored in such a way that the normal youthful circadian values of the rejuvenins and geriatrins are restored [446].

We presented the portraits of potential rejuvenins that show a wide range of rejuvenating effects as well as sample geriatrins whose effects simulate many of age-related declines and disorders. The age dependent decline in rejuvenins appear to underlie many features of aging which are further exacerbated by a coordinate increase in putative geriatrins. To this end, we truly hope that this primer can motivate those in the field to add their own rejuvenins and geriatrins to the rejuvenome and geriatrome atlases. The data and tools are publicly available at our Web Portal at www.bioscience.org/RGmoics. As shown by examples here, the rejuvenins and geriatrins might create interacting networks, akin to cellular molecular repertoires. We also hope that once the identity of all rejuvenins and geriatrins are revealed, the ensemble supplementation of lost rejuvenins alone, or in conjunction with strategies that suppress geriatrins, can lead to effective treatment of aging.

References

  • [1].Harman HD (1981). The aging process. Proc Natl Acad Sci, 78(11):7124-7128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Hayflick L (2007). Entropy explains aging, genetic determinism explains longevity, and undefined terminology explains misunderstanding both. PLoS Genet, 3:e220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Gems D,de la Guardia Y (2013). Alternative Perspectives on Aging in Caenorhabditis elegans: Reactive Oxygen Species or Hyperfunction? Antioxid Redox Signal, 19(3):321-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Koga H,Kaushik S,Cuervo AM (2011). Protein homeostasis and aging: The importance of exquisite quality control. Ageing Res Rev, 10:205-215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Douglas PM,Dillin A (2010). Protein homeostasis and aging in neurodegeneration. J Cell Biol, 190:719-729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Fraga MF,Esteller M (2007). Epigenetics and aging: the targets and the marks. Trends in genetics, 23(8):413-418. [DOI] [PubMed] [Google Scholar]
  • [7].Brunet A,Rando TA (2007). Ageing: from stem to stern. Nature, 449(7160):288. [DOI] [PubMed] [Google Scholar]
  • [8].Vijg J,Campisi J (2008). Puzzles, promises and a cure for ageing. Nature, 454(7208):1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Campisi J,Vijg J (2009). Does damage to DNA and other macromolecules play a role in aging? If so, how? J Gerontology, 64:175-178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Haigis MC,Yankner BA (2010). The aging stress response. Mol Cell, 40:333-344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Seviour EG,Lin SY (2010). The DNA damage response: Balancing the scale between cancer and ageing. Aging, 2:900-907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Rodier F,Campisi J (2011). Four faces of cellular senescence. J Cell Biology, 192(4):547-556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Charville GW,Rando TA (2011). Stem cell ageing and non-random chromosome segregation, Philos Trans R Soc Lond B Biol Sci, 366:85-93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Rando TA (2006). Stem cells, ageing and the quest for immortality. Nature, 441:1080-1086. [DOI] [PubMed] [Google Scholar]
  • [15].Liu L,Rando TA (2011). Manifestations and mechanisms of stem cell aging. J Cell Biol, 193:257-266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].López-Otín C,Kroemer G (2020). Hallmarks of health. Cell, 184, 33-63. [DOI] [PubMed] [Google Scholar]
  • [17].López-Otín C,Blasco MA,Partridge L,Serrano M,Kroemer G (2013). The hallmarks of aging. Cell, 153(6):1194-1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Gordon LB, Rothman FG, López-Otín C, Misteli T, (2014). Progeria: a paradigm for translational medicine. Cell, 156(3):400-407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Savitha S,Panneerselvam C (2006). Mitochondrial membrane damage during aging process in rat heart: potential efficacy of L-carnitine and DL α lipoic acid. Mech ageing Dev, 127(4):349-355. [DOI] [PubMed] [Google Scholar]
  • [20].Robijns J,Houthaeve G,Braeckmans K,De Vos WH (2018). Loss of nuclear envelope integrity in Aging and disease. Intl Rev Cell Mol Biol, 336:205-222. [DOI] [PubMed] [Google Scholar]
  • [21].Niedernhofer LJ,Gurkar AU,Wang Y,Vijg J,Hoeijmakers JH,Robbins PD (2018). Nuclear genomic instability and aging. Ann Rev Biochem, 87:295-322. [DOI] [PubMed] [Google Scholar]
  • [22].Ryan JM,Cristofalo VJ (1972) Histone acetylation during aging of human cells in culture. Biochem Biophys Res Commun, 48(4):735-742. [DOI] [PubMed] [Google Scholar]
  • [23].Huang JC,Yan LY,Lei ZL,Miao YL,Shi LH,Yang JW,Wang Q,Ouyang YC,Sun QY,Chen DY (2007). Changes in histone acetylation during postovulatory aging of mouse oocyte. Biol Reprod, 77(4):666-670. [DOI] [PubMed] [Google Scholar]
  • [24].Piña B,Martinez P,Suau P (1988). Differential acetylation of core histones in rat cerebral cortex neurons during development and aging. Eur J Biochem, 174(2):311-315. [DOI] [PubMed] [Google Scholar]
  • [25].O'sullivan RJ,Kubicek S,Schreiber SL,Karlseder J (2010). Reduced histone biosynthesis and chromatin changes arising from a damage signal at telomeres. Nature, Struct Mol Biol, 17(10):1218-1225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Narita M,Nuñez S,Heard E,Narita M,Lin AW,Hearn SA,Spector DL,Hannon GJ, Lowe SW (2003). Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell, 113(6):703-716. [DOI] [PubMed] [Google Scholar]
  • [27].Braig M,Lee S,Loddenkemper C,Rudolph C,Peters AH,Schlegelberger B,Stein H,Dörken B,Jenuwein T,Schmitt CA (2005). Oncogene-induced senescence as an initial barrier in lymphoma development. Nature, 436(7051):660-665. [DOI] [PubMed] [Google Scholar]
  • [28].Scaffidi P,Misteli T (2006). Lamin A-dependent nuclear defects in human aging. Science, 312(5776):1059-1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Di Micco R,Sulli G,Dobreva M,Liontos M,Botrugno OA,Gargiulo G,Dal Zuffo R,Matti V,d'Ario G,Montani E,Mercurio C (2011). Interplay between oncogene-induced DNA damage response and heterochromatin in senescence and cancer. Nature Cell Biol, 13(3):292-302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Larson K,Yan SJ,Tsurumi A,Liu J,Zhou J,Gaur K,Guo D,Eickbush TH,Li WX (2012). Heterochromatin formation promotes longevity and represses ribosomal RNA synthesis. PLoS Gen, 8(1): e1002473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Shi LH,Yang JW,Wang Q,Ouyang YC,Sun QY,Chen DY (2007). Changes in histone acetylation during postovulatory aging of mouse oocyte. Biol Reprod, 77(4):666-670. [DOI] [PubMed] [Google Scholar]
  • [32].Mostoslavsky R,Chua KF,Lombard DB,Pang WW, et al. (2006). Genomic instability and aging-like phenotype in the absence of mammalian SIRT6. Cell, 124(2):315-329. [DOI] [PubMed] [Google Scholar]
  • [33].Michishita E,McCord RA,Berber E, et al. (2008). SIRT6 is a histone H3 lysine 9 deacetylase that modulates telomeric chromatin. Nature, 452(7186):492-496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Kawakami K,Nakamura A,Ishigami A,Goto S,Takahashi R (2009). difference of site-specific histone modifications in rat liver. Biogerontology, 10(4):415-421. [DOI] [PubMed] [Google Scholar]
  • [35].Bracken AP,Kleine-Kohlbrecher D,Dietrich N, et al. (2007). The Polycomb group proteins bind throughout the INK4A-ARF locus and are disassociated in senescent cells. Genes Dev, 21(5):525-530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Dhawan S,Tschen SI,Bhushan A (2009). Bmi-1 regulates the Ink4a/Arf locus to control pancreatic β-cell proliferation. Genes & Dev, 23(8):906-911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Dang W,Steffen KK,Perry R,Dorsey JA, et al. (2009). Histone H4 lysine 16 acetylation regulates cellular n. Nature, 459(724): 802-807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Feser J,Truong D,Das C,Carson JJ,Kieft J,Harkness T,Tyler JK (2010). Elevated histone expression promotes life span extensioN, Molecular Cell, 39(5):724-735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Sarg B,Koutzamani E,Helliger W,Rundquist I,Lindner HH, (2002). Postsynthetic trimethylation of histone H4 at lysine 20 in mammalian tissues is associated with aging. J Biol Chem, 277(42):39195-39201. [DOI] [PubMed] [Google Scholar]
  • [40].Pina B,Suau P (1985) Core histone variants and ubiquitinated histones 2A and 2B of rat cerebral cortex neurons. Biochem Biophys Res Commun, 133(2):505-510. [DOI] [PubMed] [Google Scholar]
  • [41].Rogakou EP,Sekeri-Pataryas KE (1999) Histone variants of H2A and H3 families are regulated during in vitro aging in the same manner as during differentiation. Exp Gerontol, 34(6):741-754. [DOI] [PubMed] [Google Scholar]
  • [42].di Fagagna FDA,Reaper PM,Clay-Farrace L,Fiegler H,Carr P,Von Zglinicki T,Saretzki G,Carter NP,Jackson SP (2003). A DNA damage checkpoint response in telomere-initiated senescence. Nature, 426(6963):194-198. [DOI] [PubMed] [Google Scholar]
  • [43].Horvath S (2013). DNA methylation age of human tissues and cell types. Genome Biol, 14(10):3156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Hunt NJ,Kang SWS,Lockwood GP, et al. (2019). Hallmarks of aging in the liver. Comput Struct Biotech J, 17:1151-1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Blasco MA (2007). Telomere length, stem cells and aging. Nature Chem Biol, 3(10). 640-649. [DOI] [PubMed] [Google Scholar]
  • [46].López-Lluch G,Irusta PM,Navas P,de Cabo R (2008). Mitochondrial biogenesis and healthy aging. Exp Gernotol, 43(9):813-819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Bratic A,Larsson NG (2013). The role of mitochondria in aging. J Clin Invest, 123(3):951-957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Folgueras AR,Freitas-Rodríguez S,Velasco G,López-Otín C (2018). Mouse models to disentangle the hallmarks of human aging. Circ Res, 123(7):905-924. [DOI] [PubMed] [Google Scholar]
  • [49].Lenaz G,D’Aurelio M,Pich MM,Genova ML,Ventura B,Bovina C,Formiggini G,Castell GP (2000). Mitochondrial bioenergetics in aging. Bioch Biophy Acta (BBA)-Bioenerg, 1459(2-3):397-404. [DOI] [PubMed] [Google Scholar]
  • [50].Girotra M,Naveiras O,Vannini N (2020). Targeting mitochondria to stimulate hematopoiesis. Aging (Albany NY), 12(2):1042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Owen L,Sunram-Lea SI (2011). Metabolic agents that enhance ATP can improve cognitive functioning: a review of the evidence for glucose, oxygen, pyruvate, creatine, and L-carnitine. Nutrients, 3(8):735-755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Verdin E (2015). NAD+ in aging, metabolism, and neurodegeneration. Science, 350(6265):1208-1213. [DOI] [PubMed] [Google Scholar]
  • [53].Poljsak B,Kovač V,Milisav I (2020). Healthy Lifestyle Recommendations: Do the Beneficial Effects Originate from NAD+ Amount at the Cellular Level? Ox Med Cell Longevity, 2020:1-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Sharma R,Ramanathan A (2020). The aging metabolome—Biomarkers to hub metabolites. Proteomics, 20(5-6):1800407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Wiley CD,Campisi J (2016). From ancient pathways to Aging Cells—connecting metabolism and cellular senescence. Cell Metab, 23(6):1013-1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Peterson CM,Johannsen DL,Ravussin E (2012). Skeletal muscle mitochondria and aging: a review. J Aging Res, 2012:1-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Carroll B,Nelson G,Rabanal-Ruiz Y,Kucheryavenko O, et al. (2017). Persistent mTORC1 signaling in cell senescence results from defects in amino acid and growth factor sensing. J Cell Biol, 216(7):1949-1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Papadopoli D,Boulay K,Kazak L,Pollak M,Mallette FA,Topisirovic I,Hulea L (2019). mTOR as a central regulator of lifespan and aging. F1000Research, 8:1-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Sebastian-Valverde M,Pasinetti GM, (2020). The NLRP3 inflammasome as a critical actor in the inflammaging process. Cells, 9(6):1552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Liu JF,Wu SF,Liu C,Chen HZ,Yang J (2020). Integrated transcriptome, proteome, acetylome, and metabolome profiling of mouse liver during normal aging. Res Square, 1-26. [Google Scholar]
  • [61].Ham S,Lee SJV (2020). Advances in transcriptome analysis of human brain aging. Exp Mol Med, 52(11):1787-1797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Johnson AA,Shokhirev MN,Wyss-Cora T, Lehallier, B (2020). Systematic review and analysis of human Proteomics aging studies unveils a novel proteomic aging clock and identifies key processes that change with age. Ageing Res Rev, 60:101070. [DOI] [PubMed] [Google Scholar]
  • [63].Löw P (2011). The role of ubiquitin-proteasome system in ageing. Gen Comp Endocrinol, 172(1):39-43. [DOI] [PubMed] [Google Scholar]
  • [64].Rajawat YS,Hilioti Z,Bossis I (2009). Aging: central role for autophagy and the lysosomal degradative system. Ageing Res Rev, 8(3):199-213. [DOI] [PubMed] [Google Scholar]
  • [65].Cuervo AM,Bergamini E,Brunk UT,Dröge W,Ffrench M,Terman A (2005). Autophagy and aging: the importance of maintaining" clean" cells. Autophagy,1(3):131-140. [DOI] [PubMed] [Google Scholar]
  • [66].Nakamura S,Oba M,Suzuki M,Takahashi A,Yamamuro T, et al. (2019). Suppression of autophagic activity by Rubicon is a signature, of aging. Nature Commun, 10(1):1-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Shmulevich R,Krizhanovsky V (2021). Cell senescence, DNA damage, and metabolism. Antioxid Redox signal, 34(4):324-334. [DOI] [PubMed] [Google Scholar]
  • [68].Naidoo N (2009). ER and aging—protein folding and the ER stress response. Ageing Res Rev. 8(3): 150-159. [DOI] [PubMed] [Google Scholar]
  • [69].Naidoo N,Brown M (2012). The endoplasmic reticulum stress response in aging and diseases. Front Physiol, 3:263-274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Tabibzadeh S (2021). CircadiOmic medicine and aging. Ageing Res Rev, 101424. [DOI] [PubMed] [Google Scholar]
  • [71].Silva H,Conboy IM (2021). Aging and stem cell renewal. Intl [J] Biomed Health Sci. 9(2):75-88 [Google Scholar]
  • [72].Ogrodnik M (2021). Cellular aging beyond cellular senescence: Markers of senescence prior to cell cycle arrest in vitro and in vivo. Aging Cell, 20(4): e13338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Rando TA,Jones DL (2021). Regeneration, Rejuvenation, and Replacement: Turning Back the Clock on Tissue Aging. Cold Spring Harb Perspect Biol, a040907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Santos MA,Franco FN,Caldeira CA, et al. (2021). Antioxidant effect of Resveratrol: Change in MAPK cell signaling pathway during the aging process. Arch Gernotol Geriatric, 104266:1-8 [DOI] [PubMed] [Google Scholar]
  • [75].Wicher SA,Roos BB,Teske JJ,Fang YH,Pabelick C,Prakash YS (2021). Aging increases senescence, calcium signaling, and extracellular matrix deposition in human airway smooth muscle. PloS one, 16(7): e0254710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Hussain F,Kayani HUR (2020). Aging-Oxidative stress, antioxidants and computational modeling. Heliyon, 6(5):e04107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Thoma A,Akter-Miah T,Reade RL,Lightfoot AP (2020). Targeting reactive oxygen species (ROS) to combat the loss of muscle mass and function. BioGerontology, 21:475-484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Yousefzadeh M,Henpita C,Vyas R,Soto-Palma C,Robbins P,Niedernhofer L (2021). DNA damage—how and why we age? Elife,10:e62852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Bardin AJ (2020). Stem cell DNA damage and genome mutation in the context of aging and cancer initiation. Cold Spring Harb Perspect Biol, 12(10):a036210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Kahroba H,Sadeghzadeh H,Wilson DM III,Maadi H,Samadi N,Hejazi MS,Farajpour H,Onari BN,Sadeghi MR, (2020). DNA damage repair response in mesenchymal stromal cells: from cellular senescence and aging to apoptosis and differentiation ability. Ageing Res Rev, 101125. [DOI] [PubMed] [Google Scholar]
  • [81].Pitot HC (1977) Carcinogenesis and aging--two related phenomena? A review. Am [J] Pathol. 87(2): 444. [PMC free article] [PubMed] [Google Scholar]
  • [82].Fane M,Weeraratna AT (2020). How the ageing microenvironment influences tumour progression. Nature Rev Cancer, 20(2):89-106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Sameri S,Samadi P,Dehghan R,Salem E,Fayazi N,Amini R (2020). Stem cell aging in lifespan and disease: A state-of-the-art review. Curr Stem Cell Res Therap, 15(4):362-378. [DOI] [PubMed] [Google Scholar]
  • [84].Mendelsohn AR,Larrick JW (2017). The NAD+/PARP1/SIRT1 axis in aging. Rejuven Res, 20(3):244-247. [DOI] [PubMed] [Google Scholar]
  • [85].Santoro A,Martucci M,Conte M,Capri M,Franceschi C,Salvioli S (2020). Inflammaging, hormesis and the rationale for anti-aging strategies. Ageing Res Rev, 101142. [DOI] [PubMed] [Google Scholar]
  • [86].Gritsenko A,Green JP,Brough D,Lopez-Castejon G (2020). Mechanisms of NLRP3 priming in inflammaging and age-related diseases. Cytokine Growth Factor Rev, 55:15-25 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Amaya-Montoya M,Pérez-Londoño A,Guatibonza-García V,Vargas-Villanueva A,Mendivil CO (2020). Cellular senescence as a therapeutic target for diseases: a review. Adv Therap, 37(4):1407-1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Song S,Lam EWF,Tchkonia T,Kirkland JL,Sun Y (2020). Senescent cells: emerging targets for human aging and diseases. Trends Biochem Sci, 45(7):578-592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Wang AS,Dreesen O (2018). Biomarkers of cellular senescence and skin aging. Front Genetics. 9:247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Newgard CB,Sharpless NE (2013). Coming of age: molecular drivers of aging and therapeutic opportunities. J Clin Invest, 123(3): 46-950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Lu B,Chen HD,Hong-Guang HG (2012). The relationship between apoptosis and aging. Adv BioSci Biotechnol. 3(06):705. [Google Scholar]
  • [92].Huo J,Li D,McKay C,Hoke M,Worcester E,Coe F (2021). Relative contributions of urine sulfate, titratable urine anion, and GI anion to net acid load and effects of age. Physiol Rep, 9(10):e14870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Wong MWK,Braidy N,Pickford R,Vafaee F,Crawford J,Muenchhoff J,Schofield P,Attia J,Brodaty H,Sachdev P,Poljak A (2019). Plasma lipidome variation during the second half of the human lifespan is associated with age and sex but minimally with BMI. PloS one, 14(3):e0214141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Carrard J,Gallart-Ayala H,Infanger D,Teav T,Wagner J,Knaier R,Colledge F,Streese L,Königstein K,Hinrichs T,Hanssen H (2021). Metabolic view on human healthspan: A lipidome-wide association study. Metabolites, 11(5):287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Sharma R,Ramanathan A (2020). The aging metabolome-Biomarkers to hub metabolites. Proteomics. 20(5-6):1800407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Kondoh H,Kameda M,Yanagida M (2021). Whole blood metabolomics in aging research. International Journal Mol Sci, 22(1):175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Preuss HG,Kaats GR,Mrvichin N,Bagchi D (2021). Analyzing blood pressure ascent during aging in non-diabetics: Focusing on links to insulin resistance and body fat mass. J Am College of Nut, 40(4):317-326. [DOI] [PubMed] [Google Scholar]
  • [98].Lin H,Sohn J,Shen H,Langhans MT,Tuan RS (2019). Bone marrow mesenchymal stem cells: Aging and tissue engineering applications to enhance bone healing. Biomater, 203:96-110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Groarke EM,Young NS (2019). Aging and hematopoiesis. Clinics Geriatric Med, 35(3):285-293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Kovtonyuk L,Fritsch K,Feng X,Manz MG,Takizawa H (2016). Inflamm-aging of hematopoiesis, hematopoietic stem cells, and the bone marrow microenvironment. Front Immunol, 7:502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Corrado A,Cici D,Rotondo C,Maruotti N,Cantatore FP (2020). Molecular basis of bone aging. International J Mol Sci, 21(10):3679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Levi N,Papismadov N,Solomonov I,Sagi I,Krizhanovsky V (2020). The ECM path of senescence in aging: components and modifiers. FEBS J, 287(13):2636-2646. [DOI] [PubMed] [Google Scholar]
  • [103].Gao P,Gao P,Choi M,Chegireddy K,Slivano OJ,Zhao J,Zhang W,Long X (2020). Transcriptome analysis of mouse aortae reveals multiple novel pathways regulated by aging. Aging, 12(15):15603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Ewald CY (2020). The Matrisome during aging and longevity: a systems-level approach toward defining Matreotypes promoting healthy aging. Gerontology, 66(3):266-274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Shimokata H,Muller DC,Fleg JL,Sorkin J,Ziemba AW Y,Andres R (1991). Age as independent determinant of glucose tolerance. Diabetes, 40(1):44-51. [DOI] [PubMed] [Google Scholar]
  • [106].Szoke E,Shrayyef MZ,Messing S,Woerle HJ,Van Haeften TW,Meyer C,Mitrakou A,Pimenta W,Gerich JE (2008). Effect of aging on glucose homeostasis: accelerated deterioration of β-cell function in individuals with impaired glucose tolerance. Diabetes Care, 31(3):539-543. [DOI] [PubMed] [Google Scholar]
  • [107].Ayres JS (2020). The biology of physiological health. Cell, 181(2):250-269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Wilson MG,Philpot C, Morley JE (2007). Anorexia of aging in long term care: is dronabinol an effective appetite stimulant? A pilot study. J Nut Health Aging, 11(2):195. [PubMed] [Google Scholar]
  • [109].Johnson KO,Shann OM,Matu J,Holliday A,Ispoglou T,Deighton K (2020). Differences in circulating appetite-related hormone concentrations between younger and older adults: A systematic review and meta-analysis. Aging Clin Exp Res, 32(7):1233-1244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Bektas A,Schurman SH,Sen R,Ferrucci L (2018). Aging inflammation and the environment. Exp Gernotol, 105:10-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Frasca D,Blomberg BB (2020). Adipose tissue, immune aging, and cellular senescence. In: Seminars in Immunopathology, Springer; Berlin Heidelberg, 1-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Mittelbrunn M,Kroemer G (2021). Hallmarks of T cell aging. Nature Immunol, 22(6):687-698. [DOI] [PubMed] [Google Scholar]
  • [113].Franceschi C,Capri M,Monti D, Giunt S, Olivieri F, Sevini F, et al. (2007). Inflammaging and anti-inflammaging: a systemic perspective on aging and longevity emerged from studies in humans. Mech Ageing Dev, 128(1):92-105. [DOI] [PubMed] [Google Scholar]
  • [114].Fülöp T,Larbi A,Witkowski JM (2019). Human inflammaging. Gerontology, 65(5):495-504. [DOI] [PubMed] [Google Scholar]
  • [115].Tabibzadeh S (2021). Cell-centric hypotheses of aging. Front Biosci, 26:1-49. [DOI] [PubMed] [Google Scholar]
  • [116].Engelmann C,Tacke F (2022). The Potential Role of Cellular Senescence in Non-Alcoholic Fatty Liver Disease, Intl Mol Sci, 23(2):652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Wáng YXJ (2021). Gender-specific liver aging and magnetic resonance imaging. Quant Imaging Med Surg, 11(7): 2893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Baiocchi L,Glaser S,Francis H,Kennedy L,Felli E,Alpini G,Gracia-Sancho J (2021). Impact of aging on liver cells and liver disease: Focus on the biliary and vascular compartments. Hepatol Comm, 5:1125-1137 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Kelly SG,Wu K,Tassiopoulos K,Erlandson KM,Koletar SL,Palella FJ (2019). Frailty is an independent risk factor for mortality, cardiovascular disease, bone disease, and diabetes among aging adults with human immunodeficiency virus. Clin Infect Dis, 69(8):1370-1376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Jiménez-Balado J,Eich TS (2021). GABAergic dysfunction, neural network hyperactivity and memory impairments in human aging and Alzheimer’s disease. Seminars Cell Dev Biol, 116:146-159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Mercerón-Martínez D,Ibaceta-González C,Salazar C,Almaguer-Melian W,Bergado-Rosado JA,Palacios AG (2021). Alzheimer’s disease, neural plasticity, and functional recovery. J Alzheimer’s Dis, 1-14. [DOI] [PubMed] [Google Scholar]
  • [122].Verheggen IC,de Jong JJ,van Boxtel MP,Gronenschild EH,Palm WM,Postma AA,Jansen JF,Verhey FR,Backes WH (2020). Increase in blood-brain barrier leakage in healthy, older adults. Geroscience, 42(4):1183-1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Shea JM,Villeda SA (2020). Dampening the Power of the Brain—When Aging Meets Cognition. J Gerontol, 75:1607-1608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Mander BA,Winer JR,Walker MP (2017). Sleep and human aging. Neuron, 94(1):19-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Kenney WL,Munce TA (2003). Invited review: aging and human temperature regulation, J App Physiol, 95(6):2598-2603. [DOI] [PubMed] [Google Scholar]
  • [126].Von Bank H,Kirsh C,Simcox J (2021). Aging Adipose: Depot location dictates age-associated expansion and dysfunction. Ageing Res Rev, 101259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Maroulis GB (1991). Effect of aging on fertility and pregnancy. In: Semin Reprod Med, 9:165-175. [Google Scholar]
  • [128].Matorras R,Matorras F,Expósito A,Martinez L,Crisol L (2011). Decline in human fertility rates with male age: a consequence of a decrease in male fecundity with aging? Gyn Obs Invest, 71(4):229-235. [DOI] [PubMed] [Google Scholar]
  • [129].Ahmed TA,Ahmed SM,El-Gammal Z,Shouman S,Ahmed A,Mansour R,El-Badri N (2019). Oocyte aging: the role of cellular and environmental factors and impact on female fertility. Cell Biol Trans Med. 8:109-123. [DOI] [PubMed] [Google Scholar]
  • [130].Wright ME,Wise RG (2018). Can blood oxygenation level dependent functional magnetic resonance imaging be used accurately to compare older and younger populations? A mini literature review. Front Aging Neurosci, 10:371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Yeo EJ (2019). Hypoxia and aging. Exp Mol Med, 51(6):1-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Muñoz-Cánoves P,Neves J,Sousa-Victor P (2020). Understanding muscle regenerative decline with aging: new approaches to bring back youthfulness to aged stem cells. FEBS J, 287(3):406-416. [DOI] [PubMed] [Google Scholar]
  • [133].Yang YHK (2018). Aging of mesenchymal stem cells: Implication in regenerative medicine. Reg Ther, 9:120-122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].de Haan G,Lazare SS (2018). Aging of hematopoietic stem cells. Blood, J Am Soc Hematol, 131(5): 479-487. [DOI] [PubMed] [Google Scholar]
  • [135].Conboy IM,Conboy MJ,Wagers AJ,Girma ER,Weissman IL,Rando TA (2005). Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature, 433:760-764. [DOI] [PubMed] [Google Scholar]
  • [136].Carlson ME,Hsu M,Conboy IM (2008). Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells. Nature, 454:528-532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Fossel M (2002). Cell senescence in human aging and disease. Ann New York Acad Sci, 959(1):14-23. [DOI] [PubMed] [Google Scholar]
  • [138].Campisi J (2000). Cancer, aging and cellular senescence.In vivo(Athens, Greece), 14(1):183-188. [PubMed] [Google Scholar]
  • [139].Sathyan S,Ayers E,Gao T,Weiss EF,Milman S,Verghese J,Barzilai N (2020). Plasma proteomic profile of Age, health span, and all-cause mortality in older adults. Aging Cell, 19(11):e13250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Vijg J,Le Bourg E (2017). Aging and the inevitable limit to human life span, Gerontology, 63(5):432-434. [DOI] [PubMed] [Google Scholar]
  • [141].Mayer W,Niveleau A,Walter J,Fundele R,Haaf T (2000). Demethylation of the zygotic paternal genome. Nature, 403:501-502. [DOI] [PubMed] [Google Scholar]
  • [142].Greenberg MVC, (2020). Get Out and Stay Out: New Insights Into DNA Methylation Reprogramming in Mammals. Front Cell and Dev Biol, 8, 1721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Kerepesi C,Zhang B,Lee SG,Trapp A,Gladyshev VN (2021). Epigenetic clocks reveal a rejuvenation event during embryogenesis followed by aging. Sci Adv, 7(26):eabg6082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [144].Briggs R,King TJ (1952). Transplantation of living nuclei from blastula cells into enucleated frogs’ eggs. Proc Natl Acad Sci, 38(5):455-463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Campbell KH,McWhir J,Ritchie WA,Wilmut I (1996). Sheep cloned by nuclear transfer from a cultured cell line. Nature, 380(6569):64-66. [DOI] [PubMed] [Google Scholar]
  • [146].Wilmut I (2003). Dolly—her life and legacy. Cloning Stem Cells, 5(2):99-100. [DOI] [PubMed] [Google Scholar]
  • [147].Zhang B,Horvath S (2005). A general framework for weighted gene co-expression network analysis. Stat App Gen Mol Biol, 12:4. [DOI] [PubMed] [Google Scholar]
  • [148].Horvath S,Raj K (2018). DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nature, Rev Genetics, 19(6):371. [DOI] [PubMed] [Google Scholar]
  • [149].Takahashi K,Yamanaka S (2006). Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell, 126(4):663-676. [DOI] [PubMed] [Google Scholar]
  • [150].Hofmann JW,Zhao X,De Cecco M,Peterson AL,Pagliaroli L,Manivannan J,Hubbard GB,Ikeno Y,Zhang Y,Feng B,Li X (2015). Reduced expression of MYC increases longevity and enhances health-span, Cell, 160(3):477-488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Lu Y,Krishnan A,Brommer B,Tian X et al. (2020). Reversal of ageing-and injury-induced vision loss by Tet-dependent epigenetic reprogramming. Nature, 588:124-12933268865 [Google Scholar]
  • [152].Hayano M,Yang JH,Bonkowski MS, et al. (2019). DNA break-induced epigenetic drift as a cause of mammalian aging. SSRN Electronic Journal, in press. [Google Scholar]
  • [153].Rando TA,Chang HY (2012). Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock. Cell, 148(1-2):46-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Marión RM,Blasco MA (2010). Telomere rejuvenation during nuclear reprogramming. Curr Opin Genet Dev, 20:190-196. [DOI] [PubMed] [Google Scholar]
  • [155].Ksander B,Lu Y,Krishnan A,Brommer B,Tian X,Meer M,Vera D,Wang C,Zeng Q,Yu D,Horvath S (2020). Reversal of aging-induced and glaucoma-induced vision loss by in vivo epigenetic reprogramming. Invest Ophthal Visual Sci, 61(7):2364-2364. [Google Scholar]
  • [156].Ocampo A,Reddy P,Martinez-Redondo P, et al. (2016). In vivo amelioration of age-associated hallmarks by partial reprogramming. Cell, 167(7):1719-1733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Kim YJ,Seo DH,Lee SH,Lee SH,An GH, Ahn HJ,Kwon D,Seo KW,Kang KS (2018). Conditioned media from human umbilical cord blood-derived mesenchymal stem cells stimulate rejuvenation function in human skin. Biochem Biophys Rep, 16:96-102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Kim SN,Lee CJ,Nam J,Choi B,Chung E,Song SU (2021). The effects of human bone marrow-derived mesenchymal stem cell conditioned media produced with fetal bovine serum or human platelet lysate on skin rejuvenation characteristics. Intl J Stem Cells, 14(1):94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Horvath S,Mah V,Lu AT,Woo JS,Choi OW,Jasinska AJ,Riancho JA,Tung S,Coles NS,Braun J,Vinters HV (2015). The cerebellum ages slowly according to the epigenetic clock. Aging, 7(5): 294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Bunster E,Meyer RK (1993). An improved method of parabiosis. Anat Rec, 57:339-343. [Google Scholar]
  • [161].Conboy IM,Conboy MJ,Wagers AJ,Girma ER,Weissman IL,Rando TA (2005). Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature, 433:760-764. [DOI] [PubMed] [Google Scholar]
  • [162].Villeda SA,Luo J,Britschgi M,Park J-S,Ding Z,Grant JL,Couillard-Despres S,Aigner L,Rando TA,Wyss-Coray A (2011). Changes in the systemic milieu regulate adult neurogenesis. Nature, 477:90-94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Harris RB (2013). Is leptin the parabiotic “satiety” factor? Past and present interpretations. Appetite, 61:111-118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Conboy MJ,Conboy IM,Rando TA, (2013). Heterochronic parabiosis: historical perspective and methodological considerations for studies of aging and longevity. Aging Cell, 12(3):525-530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Eggel A,Wyss-Coray T (2014). A revival of parabiosis in biomedical research. Swiss Med Weekly, 144(0506). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Coleman DL (2010). A historical perspective on leptin, Nature Med, 16(10):1097-1099. [DOI] [PubMed] [Google Scholar]
  • [167].Mccay CM,Pope F,Lunsford W,Sperling G,Sambhavaphol P (1957). Parabiosis between old and young rats. Gerontology, 1(1):7-17. [DOI] [PubMed] [Google Scholar]
  • [168].Ludwig FC (1972). Mortality in syngeneic rat parabionts of different chronological age. Trans New York Acad Sci USA, 34:582-587. [DOI] [PubMed] [Google Scholar]
  • [169].Ruckh JM,Zhao JW,Shadrach JL,van Wijngaarden P,Rao TN,Wagers AJ,Franklin RJ (2012). Rejuvenation of regeneration in the aging central nervous system. Cell Stem Cell, 10(1):96-103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Brack AS,Conboy MJ,Roy S,Lee M,Kuo CJ,Keller C,Rando TA, (2007). Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science, 317:807-810. [DOI] [PubMed] [Google Scholar]
  • [171].Rohde E,Muschick P,Modersohn D,Landmann H,Löwe H (1986). The cardiovascular effect of Cordemcura on various animal models in vivo. Die Pharmazie, 41(3):194-195. [PubMed] [Google Scholar]
  • [172].Mayack SR,Shadrach JL,Kim FS,Wagers AJ (2010). Systemic signals regulate ageing and rejuvenation of blood stem cell niches. Nature, 463(7280):495-500. [DOI] [PubMed] [Google Scholar]
  • [173].Sinha M,Jang YC,Oh J,Khong D,Wu EY,Manohar R,Miller C,Regalado SG,Loffredo FS,Pancoast JR,Hirshman MF (2014). Restoring systemic GDF11 levels reverses dysfunction in mouse skeletal muscle. Science, 344(6184): 649-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Morrison SJ,Wandycz AM,Akashi K,Globerson A,Weissman IL (1996). The aging of hematopoietic stem cells. Nature Med, 2(9):1011-1016. [DOI] [PubMed] [Google Scholar]
  • [175].Rossi DJ,Bryder D,Zahn JM,Ahlenius H,Sonu R,Wagers AJ,Weissman IL (2005). Cell intrinsic alterations underlie hematopoietic stem cell aging. Proc Natl Acad Sci USA, 102(26):9194-9199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [176].Rossi DJ,Jamieson CH,Weissman IL (2008). Stems cells and the pathways to aging and cancer. Cell, 132(4):681-696. [DOI] [PubMed] [Google Scholar]
  • [177].Liang Y,Van Zant G,Szilvassy SJ (2005). Effects of aging on the homing and engraftment of murine hematopoietic stem and progenitor cells. Blood, 106(4):1479-1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Carlson ME,Hsu M,Conboy IM, (2008). Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells. Nature, 454:528-532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Conboy IM,Conboy MJ,Smythe GM,Rando TA (2003). Notch-mediated restoration of regenerative potential to aged muscle. Science, 302(5650):1575-1577. [DOI] [PubMed] [Google Scholar]
  • [180].Sahin E, DePinho RA (2010). Linking functional decline of telomeres, mitochondria and stem cells during ageing. Nature, 464(7288):520-528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [181].Jaskelioff M,Muller FL,Paik JH,Thomas E,Jiang S,Adams AC,Sahin E,Kost-Alimova M,Protopopov A,Cadiñanos J, et al. (2011). Telomerase reactivation reverses tissue degeneration in aged telomerase-deficient mice. Nature, 469:102-106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Ding Z,Wu CJ,Jaskelioff M, et al. (2012). Telomerase reactivation following telomere dysfunction yields murine prostate tumors with bone metastases. Cell, 148(5):896-907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Adler AS,Sinha S,Kawahara TL,Zhang JY,Segal E,Chang HY (2007). Motif module map reveals enforcement of aging by continual NF-kappaB activity. Genes Dev, 21:3244-3257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [184].Harrison DE,Strong R,Sharp ZD,Nelson JF,Astle CM,Flurkey K,Nadon NL,Wilkinson JE,Frenkel K,Carter CS, et al.(2009). Rapamycin fed late in life extends life-span in genetically heterogeneous mice. Nature, 460:392-395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Chen C,Liu Y,Liu Y,Zheng P, (2009). mTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells. Sci Signal, 2:ra75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Fahy GM,Brooke RT,Watson JP,Good Z,Vasanawala SS,Maecker H, et al. (2019). Reversal of epigenetic aging and immunosenescent trends in humans. Aging Cell, 18(6):e13028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Fitzgerald KN,Hodges R, Hane D, Stack E, et al. (2021). Potential reversal of epigenetic age using a diet and lifestyle intervention: a pilot randomized clinical trial. Aging, 13(7):9419-9432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [188].Choi HR,Cho KA,Kang HT,Lee JB,Kaeberlein M,Suh Y,Chung IK,Park SC (2011). Restoration of senescent human diploid fibroblasts by modulation of the extracellular matrix. Aging Cell, 10(1):148-57. [DOI] [PubMed] [Google Scholar]
  • [189].Horvath S,Singh K., Raj K,Khairnar S, et al. (2020). Reversing age: dual species measurement of epigenetic age with a single clock. bioRxiv, 1-23. [Google Scholar]
  • [190].Katsimpardi L,Litterman NK,Schein PA,Miller CM, et al. (2014). Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors. Science, 344(6184):630-634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Van Dorpe S,Bronselaer A,Nielandt J,Stalmans S, et al. (2012). The blood-brain barrier peptide database. Brain Struct Func, 217(3):687-718. [DOI] [PubMed] [Google Scholar]
  • [192].Pardridge WM, (2005). The blood-brain barrier: bottleneck in brain drug development. NeuroRx, 2(1):3-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [193].Tsay YF,Chiu CC,Tsai CB,Ho CH,Hsu PK (2007). Nitrate transporters and peptide transporters. FEBS Let, 581(12):2290-300. [DOI] [PubMed] [Google Scholar]
  • [194].Liu X,Locasale JW (2017). Metabolomics: a primer. Trends Biochem Sci, 42(4):274-284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].Rinschen MM,Ivanisevic J,Giera M,Siuzdak G (2019). Identification of bioactive metabolites using activity metabolomics. Nature Rev Mol Cell Biol, 20(6):353-367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [196].Sharma R,Ramanathan A (2020). The aging metabolome—Biomarkers to hub metabolites. Proteomics, 20(5-6):1800407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [197].Fustin JM,Ye S,Rakers C, et al. (2020). Methylation deficiency disrupts biological rhythms from bacteria to humans. Comm Biol, 3(1):1-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Cagnacci A (1996). Melatonin in relation to physiology in adult humans: Mini Review. J Pineal Res, 21(4):200-213. [DOI] [PubMed] [Google Scholar]
  • [199].Korkmaz A,Reiter RJ (2008). Epigenetic regulation: a new research area for melatonin? J Pineal Res, 44(1):41-44. [DOI] [PubMed] [Google Scholar]
  • [200].Atwood A,DeConde R,Wang SS,Mockler TC,Sabir JS,Ideker T,Kay SA (2011). Cell-autonomous circadian clock of hepatocytes drives rhythms in transcription and polyamine synthesis. Proc Natl Acad Sci, 108(45):18560-18565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [201].Tawfik A,Samra YA,Elsherbiny NM,Al-Shabrawey M (2020). Implication of hyperhomocysteinemia in blood retinal barrier (BRB) dysfunction. Biomol, 10(8):1119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [202].Navneet S,Zhao J,Wang J,Mysona B, et al. (2019). Hyperhomocysteinemia-induced death of retinal ganglion cells: the role of Müller glial cells and NRF2. Redox Biol, 24:101199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [203].Stamler JS,Osborne JA,Jaraki O, et al. (1993). Adverse vascular effects of homocysteine are modulated by endothelium-derived relaxing factor and related oxides of nitrogen, J Clin Inves, 91:308-318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].Cai B,Li X,Wang Y,Liu Y,Yang F et al. (2013). Apoptosis of bone marrow mesenchymal stem cells caused by homocysteine via activating JNK signal. PloS one, 8(5):e63561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Eren E,Yilmaz N,Aydin O (2012). High density lipoprotein and it’s dysfunction, Open Biochem J, 6:78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [206].Streck EL,Matté C,Vieira PS,Calcagnotto T,Wannmacher CM,Wajner M,Wyse AT (2003). Impairment of energy metabolism in hippocampus of rats subjected to chemically-induced hyperhomocysteinemia. Bioch Biophy Acta (BBA)-Mol Basis Dis, 1637(3):187-192. [DOI] [PubMed] [Google Scholar]
  • [207].Kumar AA, Anusree VR, Satheesh G., Vijayakumar G., Chandran M, Simon L, Lakshmi S, Pillai MR, Jaleel A, (2021). Hyperhomocysteinemia-related serum metabolome alterations not normalized by short-term folic acid treatment. Metabolomics, 17(5). 1-11. [DOI] [PubMed] [Google Scholar]
  • [208].Namekata K,Enokido Y,Ishii I,Nagai Y,Harada T,Kimura H (2004). Abnormal lipid metabolism in cystathionine β-synthase-deficient mice, an animal model for hyperhomocysteinemia. J Biol Chem, 279(51): 52961-52969. [DOI] [PubMed] [Google Scholar]
  • [209].Chen J,Li J,Wang J,Zeng D, et al. (2021). Association of serum VLDL level with hyperhomocysteinemia in hypertensive patients: A cross-sectional study. Clinical Exp Hyp, 43(1):26-33. [DOI] [PubMed] [Google Scholar]
  • [210].Waxman S,Metz J,Herbert V (1969). Defective DNA synthesis in human megaloblastic bone marrow: effects of homocysteine and methionine. J Clin Invest, 48(2):284-289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [211].Herrmann W (2006). Significance of hyperhomocysteinemia. Clin Lab, 52(7-8):367-374. [PubMed] [Google Scholar]
  • [212].Kruman II,Culmsee C,Chan SL, et al.(2000). Homocysteine elicits a DNA damage response in neurons that promotes apoptosis and hypersensitivity to excitotoxicity. J Neurosci, 20:6920-6926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [213].Nedvetsky PI,Sessa WC,Schmidt HH (2002). There's NO binding like NOS binding: protein-protein interactions in NO/cGMP signaling. Proc Natl Acad Sci, 99(26):16510-16512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Goligorsky MS,Li H,Brodsky S,Chen J (2002). Relationships between caveolae and eNOS: everything in proximity and the proximity of everything. Am [J] Phys-Renal Phys, 283(1):F1-F10. [DOI] [PubMed] [Google Scholar]
  • [215].Wan J,Deng Y,Guo J,Xiao G,Kuang D,Zhu Y,Duan Y,Wang G (2011). Hyperhomocysteinemia inhibited cardiac stem cell homing into the peri-infarcted area post myocardial infarction in rats. Exp Mol Pathol, 91(1):411-418. [DOI] [PubMed] [Google Scholar]
  • [216].Yan W,Cao Y,Zhen P,Ji D,Chai J,Xue K,Dai H,Wang W (2021). Decreased autophagy of vascular smooth muscle cells was involved in hyperhomocysteinemia-induced vascular ageing. Clin Exp Pharm Phys, 48(4):524-533. [DOI] [PubMed] [Google Scholar]
  • [217].Derouiche F,Bôle-Feysot C,Naïmi D,Coëffier M (2014). Hyperhomocysteinemia-induced oxidative stress differentially alters proteasome composition and activities in heart and aorta. Biochem Biophys Res Comm, 452(3):740-745. [DOI] [PubMed] [Google Scholar]
  • [218].Da Cunha AA,Ferreira AG,Loureiro SO,da Cunha MJ,Schmitz F,Netto CA,Wyse AT (2012). Chronic hyperhomocysteinemia increases inflammatory markers in hippocampus and serum of rats. Neurochem Res, 37(8):1660-1669. [DOI] [PubMed] [Google Scholar]
  • [219].Li JJ,Li Q,Du HP,Wang YL,You SJ, et al. (2015). Homocysteine triggers inflammatory responses in macrophages through inhibiting CSE-H2S signaling via DNA hypermethylation of CSE promoter. Intl J Mol Sci, 16(6):12560-12577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [220].Poddar R,Sivasubramanian N,DiBello PM,Robinson K,Jacobsen DW (2001). Homocysteine induces expression and secretion of monocyte chemoattractant protein-1 and interleukin-8 in human aortic endothelial cells: implications for vascular disease. Circulation, 103(22):2717-2723. [DOI] [PubMed] [Google Scholar]
  • [221].Su SJ, Huan LW, Pai LS,Liu HW,Chang KL (2005). Homocysteine at pathophysiologic concentrations activates human monocyte and induces cytokine expression and inhibits macrophage migration inhibitory factor expression. Nutrition, 21(10):994-1002. [DOI] [PubMed] [Google Scholar]
  • [222].Schroecksnadel K,Frick B,Wirleitner B,Winkler C,Schennach H,Fuchs D (2004). Moderate hyperhomocysteinemia and immune activation, Cur Pharm Biotech, 5(1):107-118. [DOI] [PubMed] [Google Scholar]
  • [223].Welch GN,Upchurch G Jr, Loscalzo J.1997. Hyperhomocyst(e)inemia and atherothrombosis. Ann N Y Acad Sci. 811:48-58 discussion 58-59. [DOI] [PubMed] [Google Scholar]
  • [224].Weiss N,Keller C,Hoffmann U,Loscalzo J (2002). Endothelial dysfunction and atherothrombosis in mild hyperhomocysteinemia. Vascular Med, 7(3):227-239. [DOI] [PubMed] [Google Scholar]
  • [225].Loscalzo J (1996). The oxidant stress of hyperhomocyst(e)inemia. J Clin Invest, 98:5-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [226].Majumder A,Singh M,George AK,Tyagi SC (2019). Restoration of skeletal muscle homeostasis by hydrogen sulfide during hyperhomocysteinemia-mediated oxidative/er stress condition. Canadian J Phys Pharma, 97(6):441-456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [227].Harker LA,Slichter SJ,Scott CR,Ross R (1974). Homocystinemia. Vascular injury and arterial thrombosis. New Engl J Med, 291:537-543. [DOI] [PubMed] [Google Scholar]
  • [228].Fryer RH,Wilson BD,Gubler DB,Fitzgerald LA,Rodgers GM (1993). Homocysteine, a risk factor for premature vascular disease and thrombosis, induces tissue factor activity in endothelial cells. Arteriosclerosis Thrombosis, 13:1327-1333. [DOI] [PubMed] [Google Scholar]
  • [229].Rodgers GM,Kane WH (1986). Activation of endogenous factor V by a homocysteine-induced vascular endothelial cell activator. J Clin Invest, 77:1909-1916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [230].Rodgers GM,Conn MT (1990). Homocysteine, an atherogenic stimulus, reduces protein C activation by arterial and venous endothelial cells. Blood, 75:895-901. [PubMed] [Google Scholar]
  • [231].Lentz SR,Sadler JE, (1991). Inhibition of thrombomodulin surface expression and protein C activation by the thrombogenic agent homocysteine. J Clin Invest, 88:1906-1914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [232].Nishinaga M,Ozawa T,Shimada K (1993). Homocysteine, a thrombogenic agent, suppresses anticoagulant heparan sulfate expression in cultured porcine aortic endothelial cells. J Clin Invest, 92:1381-1386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [233].Hajjar KA (1993). Homocysteine-induced modulation of tissue plasminogen activator binding to its endothelial cell membrane receptor. J Clin Invest, 91:2873-2879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [234].Lipton SA,Kim WK,Choi YB,Kumar S, et al. (1997). Neurotoxicity associated with dual actions of homocysteine at the N-methyl-D-aspartate receptor. Proc Natl Acad Sci, 94(11): 5923-5928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [235].Tyagi N,Vacek TP,Fleming JT,Vacek JC,Tyagi SC (2011). Hyperhomocysteinemia decreases bone blood flow. Vascular Health Risk Man, 7:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [236].Saito M,Marumo K (2018). The effects of homocysteine on the skeleton, Curr Osteo Rep, 16(5):554-560. [DOI] [PubMed] [Google Scholar]
  • [237].Baines M,Kredan MB,Davison A,Higgins G,West C,Fraser WD,Ranganath LR (2007). The association between cysteine, bone turnover, and low bone mass. Calc Tissue Intl, 81(6):450-454. [DOI] [PubMed] [Google Scholar]
  • [238].Morreels CL Jr, Fletcher BD,Weilbaecher RG,Dorst JP, (1968). The roentgenographic features of homocystinuria. Radiol, 90:1150-1158. [DOI] [PubMed] [Google Scholar]
  • [239].van Meurs JB,Dhonukshe-Rutten RA,Pluijm SM, et al.(2004). Homocysteine levels and the risk of osteoporotic fracture. N Engl J Med, 350:2033-2041. [DOI] [PubMed] [Google Scholar]
  • [240].Buysschaert M,Dramais AS,Wallemacq PE, Herman, MP (2000). Hyperhomocysteinemia in type 2 diabetes: relationship to macroangiopathy, nephropathy, and insulin resistance. Diabetes Care, 23(12): 1816-1822. [DOI] [PubMed] [Google Scholar]
  • [241].Wang CS,Wong TC,Duong TV, et al. (2019). Hyperhomocysteinemia associated with low muscle mass, muscle function in elderly hemodialysis patients: An analysis of multiple dialysis centers. BioMed Res Intl, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [242].Majumder A, Singh M, Behera J, Theilen NT, George AK, Tyagi N, Metreveli N, Tyagi SC, (2018). Hydrogen sulfide alleviates hyperhomocysteinemia-mediated skeletal muscle atrophy via mitigation of oxidative and endoplasmic reticulum stress injury. Am [J] Phys-Cell Phys, 315(5). C609-C622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [243].Veeranki S,Lominadze D,Tyagi SC (2015). Hyperhomocysteinemia inhibits satellite cell regenerative capacity through p38 alpha/beta MAPK signaling. Am J Phys-Heart Circ Phys, 309(2):H325-H334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [244].Zhang N,Chen S,Chen Y,Guo X,Sun G,Qian H,Sun Y (2016). Daytime sleepiness is associated with hyperhomocysteinemia in rural area of China: A cross-sectional study. Eur J Int Med, 35:73-77. [DOI] [PubMed] [Google Scholar]
  • [245].Kruman II, Kumaravel TS,Lohani A, et al.(2002). Folic acid deficiency and homocysteine impair DNA repair in hippocampal neurons and sensitize them to amyloid toxicity in experimental models of Alzheimer's disease. J Neurosci, 22:1752-1762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [246].Lipton SA,Kim WK,Choi YB, et al.(1997). Neurotoxicity associated with dual actions of homocysteine at the N-methyl-D-aspartate receptor. Proc Natl Acad Sci USA, 94:5923-5928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [247].Ho PI,Collins SC,Dhitavat S, et al.(2001). Homocysteine potentiates beta-amyloid neurotoxicity: role of oxidative stress. J Neuroch, 78:249-253. [DOI] [PubMed] [Google Scholar]
  • [248].Vafai SB,Stock JB, (2002). Protein phosphatase 2A methylation: a link between elevated plasma homocysteine and Alzheimer's disease. FEBS Lett. 518:(1-3): 1-4. [DOI] [PubMed] [Google Scholar]
  • [249].den Heijer T,Vermeer SE,Clarke R,Oudkerk M,Koudstaal PJ,Hofman A,Breteler MMB (2002). Homocysteine and brain atrophy on MRI of non-demented elderly. Brain, 126(1):170-175. [DOI] [PubMed] [Google Scholar]
  • [250].Miller JW,Green R,Ramos MI, et al.(2003). Homocysteine and cognitive function in the Sacramento Area Latino Study on Aging. American J Clin Nutr, 78:441-447. [DOI] [PubMed] [Google Scholar]
  • [251].Baloh RW,Ying SH,Jacobson KM, (2003). A longitudinal study of gait and balance dysfunction in normal older people. Arch Neurol, 60:835-839. [DOI] [PubMed] [Google Scholar]
  • [252].Kuo HK,Sorond FA,Chen JH,Hashmi A,Milberg WP,Lipsitz LA (2005). The role of homocysteine in multisystem problems: a systematic review. The Journals of Gerontology Series A: Biol Sci Med Sci, 60(9): 1190-1201. [DOI] [PubMed] [Google Scholar]
  • [253].Wouters MG,Boers GH,Blom HJ,Trijbels FJ, et al. (1993). Hyperhomocysteinemia: a risk factor in women with unexplained recurrent early pregnancy loss. Fertil Steril, 60(5):820-825. [PubMed] [Google Scholar]
  • [254].Hoogeveen EK,Kostense PJ,Eysink PE, et al. (2000). Hyperhomocysteinemia is associated with the presence of retinopathy in type 2 diabetes mellitus: the Hoorn study. Arch Int Med, 160(19):2984-2990. [DOI] [PubMed] [Google Scholar]
  • [255].Bagi Z,Cseko C,Tóth E,Koller A (2003). Oxidative stress-induced dysregulation of arteriolar wall shear stress and blood pressure in hyperhomocysteinemia is prevented by chronic vitamin C treatment. Am J Phys- Heart Circ Phys, 285(6):H2277-H2283. [DOI] [PubMed] [Google Scholar]
  • [256].Tsai JC,Wang H,Perrella MA, et al.(1996). Induction of cyclin A gene expression by homocysteine in vascular smooth muscle cells. J Clin Invest, 97:146-153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [257].McCully KS (1969). Vascular pathology of homocysteinemia: implications for the pathogenesis of arteriosclerosis. Am J Path, 56:111-28. [PMC free article] [PubMed] [Google Scholar]
  • [258].Fassbender K,Mielke O,Bertsch T,Nafe B,Froschen S,Hennerici M (1999). Homocysteine in cerebral macroangiography and microangiopathy. Lancet, 353:1586-1587. [DOI] [PubMed] [Google Scholar]
  • [259].Stehouwer CD,Weijenberg MP,van den Berg M,Jakobs C,Feskens EJ,Kromhout D (1998). Serum homocysteine and risk of coronary heart disease and cerebrovascular disease in elderly men: a 10-year follow-up. Arterial Thromb Vas Biol, 18:1895-1901. [DOI] [PubMed] [Google Scholar]
  • [260].Boushey CJ,Beresford SA,Omenn GS,Motulsky AG (1995). A quantitative assessment of plasma homocysteine as a risk factor for vascular disease. Probable benefits of increasing folic acid intakes. JAMA, 274:1049-1057. [DOI] [PubMed] [Google Scholar]
  • [261].Stampfer MJ,Malinow MR,Willett WC, et al.(1992). A prospective study of plasma homocyst(e)ine and risk of myocardial infarction in US physicians. JAMA, 268:877-881. [PubMed] [Google Scholar]
  • [262].Sun T,Ghosh AK,Eren M,Miyata T,Vaughan DE (2019). PAI-1 contributes to homocysteine-induced cellular senescence. Cell Signal, 64:109394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [263].Zhang D,Sun X,Liu J,Xie X,Cui W,Zhu Y (2015). Homocysteine accelerates senescence of endothelial cells via DNA hypomethylation of human telomerase reverse transcriptase. Arteriosclerosis Thromb Vas Biol, 35(1):71-78. [DOI] [PubMed] [Google Scholar]
  • [264].Herrmann W,Knapp JP (2002). Hyperhomocysteinemia: a new risk factor for degenerative diseases. Clin Lab, 48(9-10):471-481. [PubMed] [Google Scholar]
  • [265].Wu LL,Wu JT (2002). Hyperhomocysteinemia is a risk factor for cancer and a new potential tumor marker. Clin Chi Acta, 322(1-2):21-28. [DOI] [PubMed] [Google Scholar]
  • [266].Keshteli AH,Baracos VE,Madsen L (2015). Hyperhomocysteinemia as a potential contributor of colorectal cancer development in inflammatory bowel diseases: a review. World J Gastroenterol, 21(4):1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [267].Hoogeveen EK, Kostense PJ, Jakobs C, et al. (2000). Hyperhomocysteinemia increases risk of death, especially in type 2 diabetes: 5-year follow-up of the Hoorn Study. Circulation, 101(13):1506-1511. [DOI] [PubMed] [Google Scholar]
  • [268].Maldonado C,Soni CV,Todnem ND, et al. (2010). Hyperhomocysteinemia and sudden cardiac death: potential arrhythmogenic mechanisms. Cur Vas Pharma, 8(1):64-74. [DOI] [PubMed] [Google Scholar]
  • [269].Donnelly ET,McClure N,Lewis SE (1999). The effect of ascorbate and α-tocopherol supplementation in vitro on DNA integrity and hydrogen peroxide-induced DNA damage in human spermatozoa. Mutagenesis, 14(5):505-512. [DOI] [PubMed] [Google Scholar]
  • [270].Seddon AR,Liau Y,Pace PE,Miller AL, et al. (2021). Genome-wide impact of hydrogen peroxide on maintenance DNA methylation in replicating cells. Epigenetics Chromatin, 14(1):1-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [271].Niu Y,DesMarais TL,Tong Z,Yao Y,Costa M (2015). Oxidative stress alters global histone modification and DNA methylation, Free Rad Biol Med, 82:22-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [272].Driessens N,Versteyhe S,Ghaddhab C, et al. (2009). Hydrogen peroxide induces DNA single-and double-strand breaks in thyroid cells and is therefore a potential mutagen for this organ. Endocr Relat Cancer, 16(3):845. [DOI] [PubMed] [Google Scholar]
  • [273].Chen QM,Prowse KR,Tu VC,Purdom S,Linskens MH (2001). Uncoupling the senescent phenotype from telomere shortening in hydrogen peroxide-treated fibroblasts. Exp Cell Res, 265(2):294-303. [DOI] [PubMed] [Google Scholar]
  • [274].Makpol S,Zainuddin A,Rahim NA,Yusof YAM,Ngah WZW (2010). Alpha-tocopherol modulates hydrogen peroxide-induced DNA damage and telomere shortening of human skin fibroblasts derived from differently aged individuals. Planta Med, 76(09):869-875. [DOI] [PubMed] [Google Scholar]
  • [275].Maeda T,Guan JZ,Koyanagi M,Makino N (2013). Telomerase activity and telomere length distribution in vascular endothelial cells in a short-term culture under the presence of hydrogen peroxide. Geriatr Gerontol Intl, 13(3):774-782. [DOI] [PubMed] [Google Scholar]
  • [276].Boesten DM,de Vos-Houben JM,Timmermans L,den Hartog GJ,Bast A,Hageman GJ (2013). Accelerated aging during chronic oxidative stress: a role for PARP-1. Oxid Med Cell Longev, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [277].Kordowitzki P (2021). Oxidative Stress Induces Telomere Dysfunction and Shortening in Human Oocytes of Advanced Age Donors. Cells, 10(8):1866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [278].Clementi ME,Maulucci G,Bianchetti G,Pizzoferrato M,Sampaolese B,Tringali G (2021). Cytoprotective Effects of Punicalagin on Hydrogen-Peroxide-Mediated Oxidative Stress and Mitochondrial Dysfunction in Retinal Pigment Epithelium Cells. Antioxidants, 10(2):192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [279].Masschelin PM,Cox AR,Chernis N,Hartig SM (2020). The impact of oxidative stress on adipose tissue energy balance. Front Physiol, 10:1638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [280].Xie W (2008). Determination of NAD+ and NADH level in a single cell under hydrogen peroxide stress by capillary electrophoresis. Iowa State University. [DOI] [PubMed] [Google Scholar]
  • [281].Tretter L,Adam-Vizi V (2000). Inhibition of Krebs cycle enzymes by hydrogen peroxide: a key role of α-ketoglutarate dehydrogenase in limiting NADH production under oxidative stress. J Neurosci, 20(24):8972-8979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [282].Colussi C,Albertini MC,Coppola S,Rovidati S,Galli F,Ghibelli L (2000). H2O2-induced block of glycolysis as an active ADP-ribosylation reaction protecting cells from apoptosis. FASEB J, 14(14):2266-2276. [DOI] [PubMed] [Google Scholar]
  • [283].Chen L,Xu B,Liu L,Luo Y,Yin J, et al. (2010). Hydrogen peroxide inhibits mTOR signaling by activation of AMPK α leading to apoptosis of neuronal cells. Lab Invest, 90(5):762-773 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [284].Zhang J,Johnston G,Stebler B,Keller ET (2001). Hydrogen Peroxide Activates NFκB and the Interleukin-6 Promoter Through NFκB-Inducing Kinase. Antioxidants Redox Signal, 3(3):493-504. [DOI] [PubMed] [Google Scholar]
  • [285].Pierre N,Barbé C,Gilson H,Deldicque L,Raymackers JM,Francaux M (2014). Activation of ER stress by hydrogen peroxide in C2C12 myotubes. Biochem Biophys Res Commun, 450(1):459-463. [DOI] [PubMed] [Google Scholar]
  • [286].Pallepati P,Averill-Bates DA (2011). Activation of ER stress and apoptosis by hydrogen peroxide in HeLa cells: protective role of mild heat preconditioning at 40 C. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research, 1813(12):1987-1999. [DOI] [PubMed] [Google Scholar]
  • [287].Thannickal VJ,Fanburg BL (2000). Reactive oxygen species in cell signaling. American J Physiology- Lung Cell Mol Physiol, 279(6):L1005-L1028. [DOI] [PubMed] [Google Scholar]
  • [288].Miyoshi N,Oubrahim H,Chock PB,Stadtman ER (2006). Age-dependent cell death and the role of ATP in hydrogen peroxide-induced apoptosis and necrosis. Proc Natl Acad Sci, 103(6):1727-1731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [289].Gilgun-Sherki Y,Melamed E,Offen D (2001). Oxidative stress induced-neurodegenerative diseases: the need for antioxidants that penetrate the blood brain barrier. Neuropharmacol, 40(8):959-975. [DOI] [PubMed] [Google Scholar]
  • [290].Klaunig JE,Kamendulis LM (2004). The role of oxidative stress in carcinogenesis. Annu. Rev. Pharmacol. ToxicoL, 44:239-267. [DOI] [PubMed] [Google Scholar]
  • [291].Klaunig JE,Kamendulis LM,Hocevar BA (2010). Oxidative stress and oxidative damage in carcinogenesis. Tox Path, 38(1):96-109. [DOI] [PubMed] [Google Scholar]
  • [292].Schäfer M,Werner S (2008). Oxidative stress in normal and impaired wound repair. Pharmacol Res, 58(2):165-171. [DOI] [PubMed] [Google Scholar]
  • [293].Nishiyama Y,Yamamoto H,Allakhverdiev SI,Inaba M,Yokota A,Murata N (2001). Oxidative stress inhibits the repair of photodamage to the photosynthetic machinery. EMBO J, 20(20):5587-5594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [294].Aragno M,Mastrocola R,Catalano MG,Brignardello E,Danni O,Boccuzzi G (2004). Oxidative stress impairs skeletal muscle repair in diabetic rats. Diabetes, 53(4):1082-1088. [DOI] [PubMed] [Google Scholar]
  • [295].Abeti R,Duchen MR (2012). Activation of PARP by oxidative stress induced by β-amyloid: implications for Alzheimer’s disease. Neurochem Res, 37(11):2589-2596. [DOI] [PubMed] [Google Scholar]
  • [296].Buelow B,Song Y,Scharenberg AM (2008). The Poly (ADP-ribose) polymerase PARP-1 is required for oxidative stress-induced TRPM2 activation in lymphocytes. J Biol Chem, 283(36):24571-24583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [297].Long Y,Liu X,Tan XZ,Jiang CX, et al. (2020). ROS-induced NLRP3 inflammasome priming and activation mediate PCB 118-induced pyroptosis in endothelial cells. Ecotoxicol Env Safety, 189:109937. [DOI] [PubMed] [Google Scholar]
  • [298].Wang Y,Gao J,Wu F,Lai C, et al. (2021). Biological and epigenetic alterations of mitochondria involved in cellular replicative and hydrogen peroxide-induced premature senescence of human embryonic lung fibroblasts. Ecotoxicol Env Safety, 216:112204. [DOI] [PubMed] [Google Scholar]
  • [299].Brandl A,Meyer M,Bechmann V,Nerlich M,Angele P (2011). Oxidative stress induces senescence in human mesenchymal stem cells. Exp Cell Res, 317(11):1541-1547. [DOI] [PubMed] [Google Scholar]
  • [300].Dayem AA,Choi HY,Kim JH,Cho SG (2010). Role of oxidative stress in stem, cancer, and cancer stem cells. Cancers, 2(2):859-884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [301].Wang J,Liu Y,Shen H,Li H,Wang Z,Chen G (2020). Nox2 and Nox4 participate in ROS-induced neuronal apoptosis and brain injury during ischemia-reperfusion in rats. In Subarachnoid Hemorrhage, Springer, Cham, 47-54. [DOI] [PubMed] [Google Scholar]
  • [302].Matsura T,Kai M,Fujii Y,Ito H,Yamada K (1999). Hydrogen peroxide-induced apoptosis in HL-60 cells requires caspase-3 activation, Free Rad Res, 30(1):73-83. [DOI] [PubMed] [Google Scholar]
  • [303].Lochhead JJ,McCaffrey G,Quigley CE,Finch J,DeMarco M,Nametz N,Davis TP (2010). Oxidative stress increases blood-brain barrier permeability and induces alterations in occludin during hypoxia-reoxygenation, J Cerebral Blood Flow Metab, 30(9):1625-1636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [304].Haorah J,Ramirez SH,Schall K,Smith D,Pandya R,Persidsky Y (2007). Oxidative stress activates protein tyrosine kinase and matrix metalloproteinases leading to blood-brain barrier dysfunction, J Neurochem, 101(2):566-576. [DOI] [PubMed] [Google Scholar]
  • [305].Basu S,Michaëlsson K,Olofsson H,Johansson S,Melhus H (2001). Association between oxidative stress and bone mineral density. Biochem Biophys Res Commun, 288(1):275-279. [DOI] [PubMed] [Google Scholar]
  • [306].Siwik DA,Pagano PJ,Colucci WS (2001). Oxidative stress regulates collagen synthesis and matrix metalloproteinase activity in cardiac fibroblasts. Am [J] Phys-Cell Phys, 280(1):C53-C60. [DOI] [PubMed] [Google Scholar]
  • [307].Yao H,Arunachalam G,Hwang JW,Chung S,Sundar IK,Kinnula VL,Crapo JD,Rahman I (2010). Extracellular superoxide dismutase protects against pulmonary emphysema by attenuating oxidative fragmentation of ECM. Proc Natl Acad Sci USA, 107(35):15571-15576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [308].Maritim AC,Sanders A,Watkins JB III (2003). Diabetes, oxidative stress, and antioxidants: a review. J Biochem Mol Toxic, 17(1):24-38. [DOI] [PubMed] [Google Scholar]
  • [309].Hsieh YS,Chen PN,Yu CH,Chen CH,Tsai TT,Kuo DY (2015). Involvement of oxidative stress in the regulation of NPY/CART-mediated appetite control in amphetamine-treated rats. Neurotoxicology, 48:131-141. [DOI] [PubMed] [Google Scholar]
  • [310].Zhang L,Ebenezer PJ,Dasuri K,Fernandez-Kim SO,Francis J,Mariappan N,Gao Z,Ye J,Bruce-Keller AJ,Keller JN (2011). Aging is associated with hypoxia and oxidative stress in adipose tissue: implications for adipose function, Am J Phys-Endo Metab, 301(4): E599-E607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [311].Bhattacharyya A,Chattopadhyay R,Mitra S,Crowe SE (2014). Oxidative stress: an essential factor in the pathogenesis of gastrointestinal mucosal diseases. Phys Rev, 94(2):329-354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [312].Cannizzo ES,Clement CC,Sahu R,Follo C,Santambrogio L (2011). Oxidative stress, inflamm-aging and immunosenescence. J Proteomics, 74(11):2313-2323. [DOI] [PubMed] [Google Scholar]
  • [313].Li S,Tan HY,Wang N,Zhang ZJ,Lao L,Wong CW,Feng Y (2015). The role of oxidative stress and antioxidants in liver diseases. Intl J Mol Sci, 16(11):26087-26124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [314].Parola M,Robino G (2001). Oxidative stress-related molecules and liver fibrosis. J Hepatol, 35(2): 297-306. [DOI] [PubMed] [Google Scholar]
  • [315].Ahn B,Ranjit R,Premkumar P,Pharaoh G,Piekarz KM, et al. (2019). Mitochondrial oxidative stress impairs contractile function but paradoxically increases muscle mass via fibre branching. J Cachexia, Sarcopenia Muscle, 10(2):411-428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [316].Patki G, Solanki N,Atrooz F, Allam F, Salim S, (2013). Depression, anxiety-like behavior and memory impairment are associated with increased oxidative stress and inflammation in a rat model of social stress. Brain Res, 1539, 73-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [317].Berr C,Balansard B,Arnaud J,Roussel AM,Alpérovitch A,EVA Study Group (2000). Cognitive decline is associated with systemic oxidative stress: the EVA study. J Am Geriatrics Soc, 48(10):1285-1291. [DOI] [PubMed] [Google Scholar]
  • [318].Aitken RJ,Baker MA (2006). Oxidative stress, sperm survival and fertility control. Mol Cell Endocrinol, 250(1-2):66-69. [DOI] [PubMed] [Google Scholar]
  • [319].Chen J,Liu Y,Zhao Z,Qiu J (2021). Oxidative stress in the skin: Impact and related protection. Intl J Cos Sci, 43(5):495-509. [DOI] [PubMed] [Google Scholar]
  • [320].Hwang I,Tang D,Paik J (2021). Oxidative stress sensing and response in neural stem cell fate. Free Rad Biol Med, 169:74-83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [321].Tian Z,Dai X (2021). Oxidative Stress-Mediated Stem Cell Aging 3. Oxidative Stress: Human Dis Med, 55-75. [Google Scholar]
  • [322].Kurz DJ,Decary S,Hong Y,Trivier E,Akhmedov A,Erusalimsky JD (2004). Chronic oxidative stress compromises telomere integrity and accelerates the onset of senescence in human endothelial cells. J cell Sci, 117(11): 2417-2426. [DOI] [PubMed] [Google Scholar]
  • [323].Pole A,Dimri M,Dimri GP (2016). Oxidative stress, cellular senescence and ageing. AIMS Mol Sci, 3(3). [Google Scholar]
  • [324].El Assar M,Angulo J,Rodríguez-Mañas L (2013). Oxidative stress and vascular inflammation in aging. Free Rad Biol Med, 65:380-401. [DOI] [PubMed] [Google Scholar]
  • [325].Percy C,Pat B,Poronnik P,Gobe G (2005). Role of oxidative stress in age-associated chronic kidney pathologies. Advances Chronic Kidney Dis, 12(1):78-83. [DOI] [PubMed] [Google Scholar]
  • [326].Liguori I,Russo G,Curcio F,Bulli G, et al. (2018). Oxidative stress, aging, and diseases. Clin Int Aging, 13:757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [327].Quan Y,Xin Y,Tian G,Zhou J,Liu X (2020). Mitochondrial ROS-modulated mtDNA: a potential target for cardiac aging. Oxidative Med Cell Longevity, 2020:1-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [328].Khansari N,Shakiba Y,Mahmoudi M (2009). Chronic inflammation and oxidative stress as a major cause of age-related diseases and cancer. Recent Pat Inflamm Allergy Drug Discov, 3(1):73-80. [DOI] [PubMed] [Google Scholar]
  • [329].Sohal RS,Weindruch R (1996). Oxidative stress, caloric restriction, and aging. Science, 273(5271): 59-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [330].Ashraf A,Clark M,So PW (2018). The aging of iron man, Front Aging Neurosci, 10:65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [331].Altamura S,Muckenthaler MU (2009). Iron toxicity in diseases of aging: Alzheimer's disease, Parkinson's disease and atherosclerosis. J Alzheimer's Dis, 16(4):879-895. [DOI] [PubMed] [Google Scholar]
  • [332].Boldyrev AA,Aldini G,Derave W (2013). Physiology and pathophysiology of carnosine. Physiol Rev, 93:1803-1845. [DOI] [PubMed] [Google Scholar]
  • [333].Kalyankar GD,Meister A (1959). Enzymatic synthesis of carnosine and related β-alanyl and γ-aminobutyryl peptides. J Biol Chem, 234(12):3210-3218. [PubMed] [Google Scholar]
  • [334].Winnick T,Winnick RE (1959). Pathways and the physiological site of anserine formation, Nature, 183(4673):1466-1468. [DOI] [PubMed] [Google Scholar]
  • [335].Drozak J,Veiga-da-Cunha M,Vertommen D,Stroobant V,Van Schaftingen E (2010). Molecular identification of carnosine synthase as ATP-grasp domain-containing protein 1 (ATPGD1). J Biol Chem, 285(13):9346-9356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [336].Horinishi H,Grillo M,Margolis FL (1978). Purification and characterization of carnosine synthetase from mouse olfactory bulbs. J Neurochem, 31(4):909-919. [DOI] [PubMed] [Google Scholar]
  • [337].Nagai K,Niijima A,Yamano T,Otani H,Okumra N,Tsuruoka N,Nakai M,Kiso Y (2003). Possible role of L-carnosine in the regulation of blood glucose through controlling autonomic nerves. Exp Biol Med, 228(10):1138-1145. [DOI] [PubMed] [Google Scholar]
  • [338].Yamashita T,Shimada S,Guo W,Sato K,Kohmura E,Hayakawa T,Takagi T,Tohyama M (1997). Cloning and functional expression of a brain peptide/histidine transporter. J Biol Chem, 272(15): 10205-10211. [DOI] [PubMed] [Google Scholar]
  • [339].Daniel H (2004). Molecular and integrative physiology of intestinal peptide transport. Ann Rev Phys, 66:361-384. [DOI] [PubMed] [Google Scholar]
  • [340].Daniel H,Kottra G (2004). The proton oligopeptide cotransporter family SLC15 in physiology and pharmacology. Pflügers Arch, 447(5):610-618. [DOI] [PubMed] [Google Scholar]
  • [341].Hanson HT, Smith E.L, (1949). Carnosinase: An enzyme of swine kidney. J Biol Chem, 179(2). 789-801. [PubMed] [Google Scholar]
  • [342].Lenney JF (1990). Separation and characterization of two carnosine-splitting cytosolic dipeptidases from hog kidney (carnosinase and non-specific dipeptidase). Biol Chem Hoppe-Seyler, 371:433-440. [DOI] [PubMed] [Google Scholar]
  • [343].Teufel M,Saudek V,Ledig JP,Bernhardt A,Boularand S,Carreau A,Cairns NJ,Carter C,Cowley DJ,Duverger D,Ganzhorn AJ (2003). Sequence identification and characterization of human carnosinase and a closely related non-specific dipeptidase. J Biol Chem, 278(8): 6521-6531. [DOI] [PubMed] [Google Scholar]
  • [344].Fonteh AN,Harrington RJ,Tsai A,Liao P,Harrington MG, (2007). Free amino acid and dipeptide changes in the body fluids from Alzheimer’s disease subjects. Amino acids, 32(2):213-224. [DOI] [PubMed] [Google Scholar]
  • [345].Johnson P,Hammer JL (1992). Histidine dipeptide levels in ageing and hypertensive rat skeletal and cardiac muscles. Comp Biochem Physiol B Biochem Mol Biol, 103(4): 981-984. [DOI] [PubMed] [Google Scholar]
  • [346].Stuerenburg HJ,Kunze K (1999). Concentrations of free carnosine (a putative membrane-protective antioxidant) in human muscle biopsies and rat muscles. Arch Gerontol Geriatr, 29(2):107-113. [DOI] [PubMed] [Google Scholar]
  • [347].Tallon MJ,Harris RC,Maffulli N,Tarnopolsky MA (2007). Carnosine, taurine and enzyme activities of human skeletal muscle fibres from elderly subjects with osteoarthritis and young moderately active subjects. BioGerontology, 8(2):129-137. [DOI] [PubMed] [Google Scholar]
  • [348].Derave W,Jones G,Hespel P,Harris RC (2008). Creatine supplementation augments skeletal muscle carnosine content in senescence-accelerated mice (SAMP8). Rejuvenation Res, 11(3):641-647. [DOI] [PubMed] [Google Scholar]
  • [349].Gardner ML,Illingworth KM, et al. (1991). Intestinal absorption of the intact peptide carnosine in man, and comparison with intestinal permeability to lactulose. J Phys, 439(1): 411-422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [350].Bauchart C,Savary-Auzeloux I,Patureau Mirand P,Thomas E,Morzel M,Rémond D (2007). Carnosine concentration of ingested meat affects carnosine net release into the portal vein of minipigs. J Nutr, 137(3):589-593. [DOI] [PubMed] [Google Scholar]
  • [351].Wu T,Tao Y,Tsang F,Abe K, et al. (2015). The effect of L-carnosine on the circadian resetting of clock genes in the heart of rats. Mol Biol Rep, 42(1): 87-94. [DOI] [PubMed] [Google Scholar]
  • [352].Oppermann H,Alvanos A,Seidel C,Meixensberger J,Gaunitz F, (2019). Carnosine influences transcription via epigenetic regulation as demonstrated by enhanced histone acetylation of the pyruvate dehydrogenase kinase 4 promoter in glioblastoma cells. Amino acids, 51(1):61-71. [DOI] [PubMed] [Google Scholar]
  • [353].Calabrese V,Davinelli S,Luca M,Zella D,Calabrese EJ,Scapagnini G, (2015). Inflammaging, oxidative stress and carnosine: role of hormetic vitagenes. Imidazole dipeptides: chemistry, analysis, function and effects. Cambridge: Royal Soc Chem, 238-256. [Google Scholar]
  • [354].Torreggiani MTA (1998). A pulse radiolysis study of carnosine in aqueous solutioN, Intl J Rad Biol, 74(3):333-340. [DOI] [PubMed] [Google Scholar]
  • [355].Klebanov GI,Teselkin YO,Babenkova IV,Popov I, et al. (1997). Evidence for a direct interaction of superoxide anion radical with carnosine. IUBMB Life, 43(1):99-106. [DOI] [PubMed] [Google Scholar]
  • [356].McFarland GA,Holliday R (1999). Further evidence for the rejuvenating effects of the di-peptide L-Carnosine on cultured diploid fibroblasts. Exp Gerontol, 34: 35-45. [DOI] [PubMed] [Google Scholar]
  • [357].Baek SH,Noh AR,Kim KA, et al. (2014). Modulation of mitochondrial function and Autophagy mediates carnosine neuroprotection against ischemic brain damage. Stroke, 45(8): 2438-2443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [358].Cararo JH,Streck EL,Schuck PF,da C Ferreira G (2015). Carnosine and related peptides: therapeutic potential in disorders. Aging Dis, 6(5):369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [359].Hipkiss AR,Baye E,de Courten B (2016). Carnosine and the processes of ageing. Maturitas, 93: 28-33. [DOI] [PubMed] [Google Scholar]
  • [360].Ooi TC,Chan KM,Sharif R (2017). Antioxidant, Anti-inflammatory, and genomic stability enhancement effects of Zinc L-carnosine: A potential cancer chemopreventive agent?. Nutr Cancer, 69(2):201-210. [DOI] [PubMed] [Google Scholar]
  • [361].Yeargans GS,Seidler NW (2003). Carnosine promotes the heat denaturation of glycated proteiN, Biochem Biophys Res Commun, 300(1):75-80. [DOI] [PubMed] [Google Scholar]
  • [362].Aldini G,Orioli M,Rossoni G,Savi F,Braidotti P,Vistoli G,Yeum KJ,Negrisoli G,Carini M (2011). The carbonyl scavenger carnosine ameliorates dyslipidaemia and renal function in Zucker obese rats. J Cell Mol Med, 15(6):1339-1354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [363].Johnson HA,Devaney KO (1982). Thermal instability in the endoplasmic reticulum of the rat hepatocyte. Virchows Archiv B, 40(1):357-364. [DOI] [PubMed] [Google Scholar]
  • [364].Babizhayev MA (1989). Antioxidant activity of L-carnosine, a natural histidine-containing dipeptide in crystalline lens. Biochimica et Biophysica Acta (BBA)-Lipids Lipid Metab, 1004(3):363-371. [DOI] [PubMed] [Google Scholar]
  • [365].Yan H,Guo Y,Zhang J,Ding Z,Ha W,Harding JJ, (2008). Effect of carnosine, aminoguanidine, and aspirin drops on the prevention of cataracts in diabetic rats. Mol Vision, 14: 2282-2291. [PMC free article] [PubMed] [Google Scholar]
  • [366].Attanasio F,Cataldo S,Fisichella S,Nicoletti S,Nicoletti VG,Pignataro B,Savarino A,Rizzarelli E (2009). Protective effects of l- and d-carnosine on alpha-crystallin amyloid fibril formation: implications for cataract disease. Biochem, 48: 6522-6531 [DOI] [PubMed] [Google Scholar]
  • [367].Babizhayev MA, Yermakova VN,Sakina NL,Evstigneeva RP,Rozhkova EA,Zheltukhina GA (1996). Nα-Acetylcarnosine is a prodrug of L-carnosine in ophthalmic application as antioxidant. Clinica Chimica Acta, 254(1):1-21. [DOI] [PubMed] [Google Scholar]
  • [368].Babizhayev MA,Deyev AI. Yermakova VN, et al. (2002). Efficacy of N-acetylcarnosine in the treatment of cataracts. Drugs in R&D, 3(2): 87-103. [DOI] [PubMed] [Google Scholar]
  • [369].Bonner AB,Swann ME,Marway JS,Heap LC,Preed VR, (1995). Lysosomal and nonlysosomal protease activities of the brain in response to ethanol feeding. Alcohol, 12(6): 505-509. [DOI] [PubMed] [Google Scholar]
  • [370].Bharadwaj LA,Davies GF,Xavier IJ,Ovsenek N (2002). l-carnosine and verapamil inhibit hypoxia-induced expression of hypoxia inducible factor (HIF-1 α) in H9c2 cardiomyoblasts. Pharma Res, 45(3): 175-181. [DOI] [PubMed] [Google Scholar]
  • [371].Hipkiss AR (2005). Glycation, ageing and carnosine: are carnivorous diets beneficial? Mech ageing Dev, 126(10):1034-1039. [DOI] [PubMed] [Google Scholar]
  • [372].Kamei J,Ohsawa M,Miyata S,Tanaka SI (2008). Preventive effect of L-carnosine on changes in the thermal nociceptive threshold in streptozotocin-induced diabetic mice. Eur J Pharmacol, 600(1-3): 83-86. [DOI] [PubMed] [Google Scholar]
  • [373].Riedl E,Pfister F,Braunagel M,Brinkkötter P, et al. (2011). Carnosine prevents apoptosis of glomerular cells and podocyte loss in STZ diabetic rats. Cell Phys Biochem, 28(2): 279-288. [DOI] [PubMed] [Google Scholar]
  • [374].Preston JE, Hipkiss AR, Himsworth DT, Romero IA, Abbott JN, (1998). Toxic effects of β-amyloid (25-35) on immortalised rat brain endothelial cell: protection by carnosine, homocarnosine and β-alanine. Neurosci Lett, 242(2). 105-108. [DOI] [PubMed] [Google Scholar]
  • [375].Corona C,Frazzini V,Silvestri E,Lattanzio R,La Sorda R,Piantelli M,Canzoniero LM,Ciavardelli D,Rizzarelli E,Sensi SL (2011). Effects of dietary supplementation of Carnosine on mitochondrial dysfunction, amyloid pathology, and cognitive deficits in 3xTg-AD mice. PloS one, 6: e17971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [376].Herculano B,Tamura M,Ohba A,Shimatani M,Kutsuna N,Hisatsune T (2013). β-alanyl-L-histidine rescues cognitive deficits caused by feeding a high fat diet in a transgenic mouse model of Alzheimer's disease. J Alzheimer’s Dis, 33(4):983-997. [DOI] [PubMed] [Google Scholar]
  • [377].Kim KS,Choi SY,Kwon HY,Won MH,Kang TC,Kang JH (2002). The ceruloplasmin and hydrogen peroxide system induces α-synuclein aggregation in vitro. Biochimie, 84(7): 625-631. [DOI] [PubMed] [Google Scholar]
  • [378].Kang JH,Kim KS (2003). Enhanced oligomerization of the alpha-synuclein mutant by the Cu, Zn-superoxide dismutase and hydrogen peroxide system. Mol Cells, 15(1): 87-93. [PubMed] [Google Scholar]
  • [379].Tsai SJ,Kuo WW,Liu WH,Yin MC (2010). Antioxidative and anti-inflammatory protection from carnosine in the striatum of MPTP-treated mice. J Agricult Food Chem, 58(21):11510-11516. [DOI] [PubMed] [Google Scholar]
  • [380].Boldyrev A,Fedorova T,Stepanova M, et al. (2008). Carnisone increases efficiency of DOPA therapy of Parkinson's disease: a pilot study. Rejuvenation Res, 11(4):821-827. [DOI] [PubMed] [Google Scholar]
  • [381].Hipkiss AR,Brownson C (2000). Carnosine reacts with protein carbonyl groups: another possible role for the anti-ageing peptide? Biogerontolgy, 1(3):217-223. [DOI] [PubMed] [Google Scholar]
  • [382].Hipkiss AR,Brownson C,Bertani MF,Ruiz E,Ferro A (2002). Reaction of carnosine with aged proteins: another protective process? An New York Acad Sci, 959(1):285-294. [DOI] [PubMed] [Google Scholar]
  • [383].Hipkiss AR (2009). On the enigma of carnosine’s anti-ageing actions. Exp Gerontol, 44(4):237-242. [DOI] [PubMed] [Google Scholar]
  • [384].Shao L,Li QH,Tan ZM (2004). L-Carnosine reduces telomere damage and shortening rate in cultured normal fibroblasts. Biochem Biophys Res Commun, 324:931-936. [DOI] [PubMed] [Google Scholar]
  • [385].Zhang MJ,Wang SP,Mao LL,Leak RK,Shi YJ,Zhang WT, et al.(2014). Omega-3 Fatty Acids Protect the Brain against Ischemic Injury by Activating Nrf2 and Upregulating Heme Oxygenase 1. J Neurosci, 34(5):1903-1915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [386].Baye E,Ukropcova B,Ukropec J,Hipkiss A,Aldini G,De Courten B (2016). Physiological and therapeutic effects of carnosine on cardiometabolic risk and disease. Amino acids, 48(5):1131-1149. [DOI] [PubMed] [Google Scholar]
  • [387].Boldyrev AA,Gallant SC,Sukhich GT (1999). Carnosine: the protective, anti-aging peptide. Mol Asp Med, 19:581-587. [DOI] [PubMed] [Google Scholar]
  • [388].Yuneva MO,Bulygina ER,Gallant SC,Kramarenko GG,Stvolinsky SL,Semyonova ML,Boldyrev AA (1999). Effect of carnosine on age-induced changes in senescence-accelerated mice. [J] Anti-Aging Med, 2(4): 337-342. [Google Scholar]
  • [389].Gallant S,Semyonova M,Yuneva M (2000). Carnosine as a potential anti-senescence drug. Biochemistry C/c of Biokhimiia, 65(7):866-868. [PubMed] [Google Scholar]
  • [390].Nagai K,Suda T (1986). Antineoplastic effects of carnosine and beta-alanine--physiological considerations of its antineoplastic effects. Nihon seirigaku zasshi. J Phys Soc Jap, 48(11): 741-747. [PubMed] [Google Scholar]
  • [391].Holliday R,McFarland GA (1996). Inhibition of the growth of transformed and neoplastic cells by the dipeptide carnosine. Br J Cancer, 73(8):966-971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [392].Gaunitz F,Hipkiss AR (2012). Carnosine and cancer: a perspective. Amino Acids, 43:135-142. [DOI] [PubMed] [Google Scholar]
  • [393].Iovine B,Iannella ML,Nocella F,Pricolo MR,Bevilacqua MA (2012). Carnosine inhibits KRAS-mediated HCT116 proliferation by affecting ATP and ROS productioN, Cancer Lett, 315(2):122-128. [DOI] [PubMed] [Google Scholar]
  • [394].Bachrach U (2010). The early history of polyamine research. Plant Phys Biochem, 48(7):490-495. [DOI] [PubMed] [Google Scholar]
  • [395].Bitrián M,Zarza X,Altabella T,Tiburcio AF,Alcázar R (2012). Polyamines under abiotic stress: metabolic crossroads and hormonal crosstalks in plants. Metabolites, 2(3):516-528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [396].Xu TT,Li H,Dai Z,Lau GK,Li BY,Zhu WL, et al. (2020). Spermidine and spermine delay brain aging by inducing Autophagy in SAMP8 mice. Aging, 12(7):6401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [397].Coleman CS,Pegg AE (1997). Proteasomal degradation of spermidine/spermine N 1-acetyltransferase requires the carboxyl-terminal glutamic acid residues. J Biol Chem, 272(18):12164-12169. [DOI] [PubMed] [Google Scholar]
  • [398].Atwood A,DeConde R,Wang SS, et al. (2011). Cell-autonomous circadian clock of hepatocytes drives rhythms in transcription and polyamine synthesis. Proc Natl Acad Sci USA, 108(45): 18560-18565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [399].Zwighaft Z,Aviram R,Shalev M, et al. (2015). Circadian clock control by polyamine levels through a mechanism that declines with age. Cell Metab, 22(5): 874-885. [DOI] [PubMed] [Google Scholar]
  • [400].Pietrocola F,Lachkar S,Enot DP, et al. (2015). Spermidine induces autophagy by inhibiting the acetyltransferase EP300. Cell Death Diff, 22(3): 509-516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [401].Madeo F,Pietrocola F,Eisenberg T,Kroemer G (2014). Caloric restriction mimetics: towards a molecular definition. Nature, Rev Drug Dis, 13(10): 727-740. [DOI] [PubMed] [Google Scholar]
  • [402].Soda K,Kano Y,Chiba F,Koizumi K,Miyaki Y (2013). Increased polyamine intake inhibits age-associated alteration in global DNA methylation and 1, 2-dimethylhydrazine-induced tumorigenesis. PloS one, 8(5):e64357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [403].Soda K (2015). Biological effects of polyamines on the prevention of aging-associated diseases and on life-span extension. Food Sci Tech Res, 21(2):145-157. [Google Scholar]
  • [404].Soda K (2020). Spermine and gene methylation: A mechanism of life-span extension induced by polyamine-rich diet. Amino acids, 52(2):213-224. [DOI] [PubMed] [Google Scholar]
  • [405].Eisenberg T,Knauer H,Schauer A, et al. (2009). Induction of autophagy by spermidine promotes longevity. Nature Cell Biol, 11(11):1305-1314. [DOI] [PubMed] [Google Scholar]
  • [406].Mandal S,Mandal A,Johansson HE,Orjalo AV,Park MH (2013). Depletion of cellular polyamines, spermidine and spermine, causes a total arrest in translation and growth in mammalian cells. Proc Natl Acad Sci USA, 110(6):2169-2174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [407].Porter CW,Bergeron RJ (1983). Spermidine requirement for cell proliferation in eukaryotic cells: structural specificity and quantitation, Science, 219(4588):1083-1085. [DOI] [PubMed] [Google Scholar]
  • [408].Ramot Y,Tiede S,Bíró T, et al. (2011). Spermidine promotes human hair growth and is a novel modulator of human epithelial stem cell functions. PloS one, 6(7):e22564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [409].Oredsson SM (2003). Polyamine dependence of normal cell-cycle progression. Biochemical Society Transactions, 31(2):366-370. [DOI] [PubMed] [Google Scholar]
  • [410].Chattopadhyay MK,Chen W,Poy G,Cam M,Stiles D,Tabor H, (2009). Microarray studies on the genes responsive to the addition of spermidine or spermine to a Saccharomyces cerevisiae spermidine synthase mutant. Yeast, 26(10):531-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [411].Pegg AE,Borchardt RT,Coward JK, (1981). Effects of inhibitors of spermidine and spermine synthesis on polyamine concentrations and growth of transformed mouse fibroblasts. Biochem J, 194(1):79-89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [412].Landau G, Bercovich Z, Park MH, Kahana C, (2010). The role of polyamines in supporting growth of mammalian cells is mediated through their requirement for translation initiation and elongation. J Biol Chem, 285(17). 12474-12481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [413].Rubinsztein DC,Mariño G,Kroemer G (2011). Autophagy and aging. Cell, 146(5):682-695. [DOI] [PubMed] [Google Scholar]
  • [414].Ren J,Zhang Y (2018). Targeting autophagy in aging and aging-related cardiovascular diseases. Trends Pharma Sci, 39(12):1064-1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [415].Salminen A,Kaarniranta K (2009). Regulation of the aging process by autophagy. Trends Mol Med, 15(5):217-224. [DOI] [PubMed] [Google Scholar]
  • [416].Hansen M,Rubinsztein DC,Walker DW (2018). Autophagy as a promoter of longevity: insights from model organisms. Nature Rev Mol Cell Biol, 19(9):579-593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [417].Green DR (2019). Polyamines and aging: A CLEAR connection? Mol Cell, 76(1): 5-7. [DOI] [PubMed] [Google Scholar]
  • [418].Morselli E,Mariño G,Bennetzen MV, et al. (2011). Spermidine and resveratrol induce Autophagy by distinct pathways converging on the acetylproteome. J Cell Biol, 192(4):615-629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [419].Zhou S,Gu J,Liu R,Wei S, et al. (2018). Spermine alleviates acute liver injury by inhibiting liver-resident macrophage pro-inflammatory response through ATG5-dependent autophagy. Front Immunol, 9: 948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [420].Eisenberg T,Abdellatif M,Zimmermann A,Schroeder S,Pendl T,Harger A,Stekovic S,Schipke J,Magnes C,Schmidt A,Ruckenstuhl C (2017). Dietary spermidine for lowering high blood pressure. Autophagy, 13(4):767-769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [421].LaRocca TJ,Gioscia-Ryan RA,Hearon CM Jr,Seals DR (2013). The autophagy. enhancer spermidine reverses arterial aging. Mech Ageing Dev, 134(7-8):314-320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [422].Eisenberg T,Abdellatif M,Schroeder S,Primessnig U,Stekovic S,Pendl T,Harger A,Schipke J,Zimmermann A,Schmidt A,Tong M (2016). Cardioprotection and life-span extension by the natural polyamine spermidine. Nature Med, 22(12):1428-1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [423].Wirth M,Schwarz C,Benson G, et al. (2019). Effects of spermidine supplementation on cognition and biomarkers in older adults with subjective cognitive decline (SmartAge)—study protocol for a randomized controlled trial. Alzheimer’s Res Therapy, 11(1):1-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [424].Freitag K,Sterczyk N,Schulz J,Houtman J, et al. (2020). The autophagy activator Spermidine ameliorates Alzheimer's disease pathology and neuroinflammation in mice. bioRxiv, 1-44 [Google Scholar]
  • [425].Eisenberg T,Michael E,Kroemer G,Tavernarakis N (2014). Spermidine protects against α-synuclein neurotoxicity. Cell Cycle, 13(24):3903-3908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [426].Jeong JW,Cha HJ,Han MH, et al. (2018). Spermidine protects against oxidative stress in inflammation models using macrophages and zebrafish. Biomol Therapeutics, 26(2):146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [427].Choi YH,Park HY (2012). Anti-inflammatory effects of spermidine in lipopolysaccharide-stimulated BV2 microglial cells. J Biomed Sci, 19(1): 1-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [428].Paul S,Kang SC (2013). Natural polyamine inhibits mouse skin inflammation and macrophage activation, Inflammation Res, 62(7): 681-688. [DOI] [PubMed] [Google Scholar]
  • [429].Yuan H,Wu SX,Zhou YF,Peng F (2021). Spermidine Inhibits Joints Inflammation and Macrophage Activation in Mice with Collagen-Induced Arthritis. J Inflammation Res, 14: 2713-2721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [430].Baek AR,Hong J,Song KS, et al. (2020). Spermidine attenuates bleomycin-induced lung fibrosis by inducing autophagy, inhibiting endoplasmic reticulum stress (ERS)-induced cell death in mice. Exp Mol Med, 52(12):2034-2045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [431].Minois N (2014). Molecular Basis of the ‘Anti-Aging'Effect of Spermidine and Other Natural Polyamines-A Mini-Review. Gerontology, 60(4):319-326. [DOI] [PubMed] [Google Scholar]
  • [432].Rider JE,Hacker A,Mackintosh CA, et al. (2007). Spermine and spermidine mediate protection against oxidative damage caused by hydrogen peroxide. Amino acids, 33(2): 231-240. [DOI] [PubMed] [Google Scholar]
  • [433].Douki T,Bretonniere Y,Cadet J (2000). Protection against radiation-induced degradation of DNA bases by polyamines. Radiat Res, 153(1):29-35. [DOI] [PubMed] [Google Scholar]
  • [434].Pedreño E,López-Contreras AJ,Cremades A,Peñafiel R (2005). Protecting or promoting effects of spermine on DNA strand breakage induced by iron or copper ions as a function of metal concentration, J Inorganic Biochem, 99(10):2074-2080. [DOI] [PubMed] [Google Scholar]
  • [435].Dykstra WG,Herbst EJ (1965). Spermidine in regenerating liver: relation to rapid synthesis of ribonucleic acid. Science, 149(3682): 428-429. [DOI] [PubMed] [Google Scholar]
  • [436].Noro T,Namekata K,Kimura A, et al.2015). Spermidine promotes retinal ganglion cell survival and optic nerve regeneration in adult mice following optic nerve injury. Cell Death Dis, 6(4):e1720-e1720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [437].Jing YH,Yan JL,Wang QJ, et al. (2018). Spermidine ameliorates the neuronal aging by improving the mitochondrial function in vitro. Exp Gernotol, 108:77-86. [DOI] [PubMed] [Google Scholar]
  • [438].Yamamoto T,Hinoi E,Fujita H,Iezaki T, et al. (2012). The natural polyamines spermidine and spermine prevent bone loss through preferential disruption of osteoclastic activation in ovariectomized mice. Br J Pharmacol, 166(3):1084-1096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [439].Zhang X,Zhang L,Chen Z, et al. (2021). Exogenous spermine attenuates diabetic kidney injury in rats by inhibiting AMPK/mTOR signaling pathway. Intl J Mol Med, 47(3): 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [440].Ito D,Ito H,Ideta T,Kanbe A,Ninomiya S,Shimizu M (2021). Systemic and topical administration of spermidine accelerates skin wound healing. Cell Comm Signal, 19(1): 1-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [441].Yue F,Li W,Zou J, et al. (2017). Spermidine prolongs life-span and prevents liver fibrosis and hepatocellular carcinoma by activating MAP1S-mediated autophagy. Cancer Res, 77(11): 2938-2951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [442].Madeo F,Carmona-Gutierrez D,Kepp O, Kroeme, G (2018). Spermidine delays aging in humans. Aging (Albany NY), 10(8):2209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [443].Kiechl S,Pechlaner R,Willeit P, et al. (2018). Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr, 108(2):371-380. [DOI] [PubMed] [Google Scholar]
  • [444].Ray RM,Zimmerman BJ,McCormack SA,Patel TB,Johnson LR (1999). Polyamine depletion arrests cell cycle and induces inhibitors p21Waf1/Cip1, p27Kip1, and p53 in IEC-6 cells. Am [J] Phys-Cell Phys, 276(3):C684-C691. [DOI] [PubMed] [Google Scholar]
  • [445].Pucciarelli S,Moreschini B,Micozzi D, et al. (2012). Spermidine and spermine are enriched in whole blood of nona/centenarians. Rejuvenation Res. 15(6): 590-595. [DOI] [PubMed] [Google Scholar]
  • [446].Tabibzadeh S (2021). From genoprotection to rejuvenation, Front Biosci, 26: 97-162. [DOI] [PubMed] [Google Scholar]
  • [447].Levine DC,Hong H,Weidemann BJ, et al. (2020). NAD+ controls circadian reprogramming through PER2 nuclear translocation to counter aging. Mol Cell, 78(5):835-849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [448].Schreiber V,Dantzer F,Ame JC,De Murcia G (2006). Poly (ADP-ribose): novel functions for an old molecule. Nature Rev Mol Cell Biol, 7(7):517-28. [DOI] [PubMed] [Google Scholar]
  • [449].Asher G,Reinke H,Altmeyer M,Gutierrez-Arcelus M,Hottiger MO,Schibler U (2010). Poly (ADP-ribose) polymerase 1 participates in the phase entrainment of circadian clocks to feeding. Cell, 142(6):943-53. [DOI] [PubMed] [Google Scholar]
  • [450].Gibson BA,Kraus WL (2012). New insights into the molecular and cellular functions of poly (ADP-ribose) and PARPs. Nature Rev Mol Cell Biol, 13(7):411-24. [DOI] [PubMed] [Google Scholar]
  • [451].Zhao Q,Wang J,Levichkin IV,Stasinopoulos S,Ryan MT,Hoogenraad NJ, (2002). A mitochondrial specific stress response in mammalian cells. EMBO J. 21(17):4411-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [452].Yoneda T,Benedetti C,Urano F,Clark SG,Harding HP,Ron D (2004). Compartment-specific perturbation of protein handling activates genes encoding mitochondrial chaperones. J Cell Sci, 117(18):4055-66. [DOI] [PubMed] [Google Scholar]
  • [453].Durieux J,Wolff S,Dillin A (2011). The cell-non-autonomous nature, of electron transport chain-mediated longevity. Cell, 144(1):79-91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [454].Honda Y,Honda S (1999). The daf-2 gene network for longevity regulates oxidative stress resistance and Mn-superoxide dismutase gene expression in Caenorhabditis elegans. FASEB J, 13(11):1385-93. [PubMed] [Google Scholar]
  • [455].Mouchiroud L,Houtkooper RH,Moullan N,Katsyuba E,Ryu D,Cantó C,Mottis A,Jo YS,Viswanathan M,Schoonjans K,Guarente L (2013). The NAD+/sirtuin pathway modulates longevity through activation of mitochondrial UPR and FOXO signaling. Cell, 154(2):430-441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [456].Massudi H,Grant R,Braidy N,Guest J,Farnsworth B,Guillemin GJ (2012). Age-associated changes in oxidative stress and NAD+ metabolism in human tissue. PloS one, 7:1-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [457].Rabini RA,Petruzzi E,Staffolani R, et al. (1997). Diabetes mellitus and subjects' ageing: a study on the ATP content and ATP-related enzyme activities in human erythrocytes. Eur J Clin Invest, 27(4): 327-332. [DOI] [PubMed] [Google Scholar]
  • [458].Zhu XH,Lu M,Lee BY,Ugurbil K,Chen W (2015). In vivo NAD assay reveals the intracellular NAD contents and redox state in healthy human brain and their age dependences. Proc Natl Acad Sci USA, 112(9): 2876-2881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [459].Imai SI,Guarente L (2014). NAD+ and sirtuins in aging and disease. Trends Cell Biol, 24(8):464-471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [460].Rajman L,Chwalek K,Sinclair DA (2018). Therapeutic potential of NAD-boosting molecules: the In vivo evidence. Cell Metab, 27(3): 529-547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [461].Clement J,Wong M,Poljak A,Sachdev P,Braidy N (2019). The plasma NAD+ metabolome is dysregulated in “normal” aging. Rejuvenation Res, 22(2): 121-130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [462].Gaur U,Tu J,Li D,Gao Y, et al. (2017). Molecular evolutionary patterns of NAD+/Sirtuin aging signaling pathway across taxa. PloS one, 12:e0182306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [463].Mills KF,Yoshida S,Stein LR,Grozio A, et al. (2016). Long-term administration of nicotinamide mononucleotide mitigates age-associated physiological decline in mice. Cell Metab, 24(6):795-806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [464].Yoshino J,Baur JA,Imai SI (2018). NAD+ intermediates: the biology and therapeutic potential of NMN and NR. Cell Metab, 27(3):513-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [465].Stein LR,Imai SI (2014). Specific ablation of Nampt in adult neural stem cells recapitulates their functional defects during aging. EMBO J, 33(12):1321-1340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [466].Yoon MJ,Yoshida M,Johnson S,Takikawa A,Usui I,Tobe K,Nakagawa T,Yoshino J,Imai SI (2015). SIRT1-mediated eNAMPT secretion from adipose tissue regulates hypothalamic NAD+ and function in mice. Cell Metab, 21(5):706-717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [467].Zhang J,Cai Q,Jiang M,Liu Y, et al. (2017). Mesencephalic astrocyte-derived neurotrophic factor alleviated 6-OHDA-induced cell damage via ROS-AMPK/mTOR mediated autophagic inhibition, Exp Gernotol, 89:45-56. [DOI] [PubMed] [Google Scholar]
  • [468].Frederick DW,Davis JG,Dávila A, et al. (2015). Increasing NAD synthesis in muscle via nicotinamide phosphoribosyltransferase is not sufficient to promote oxidative metabolism. J Biol Chem, 290(3):1546-1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [469].Dellinger RW,Santos SR,Morris M,Evans M,Alminana D,Guarente L,Marcotulli E (2017). Repeat dose NRPT (nicotinamide riboside and pterostilbene) increases NAD+ levels in humans safely and sustainably: a randomized, double-blind, placebo-controlled study. NPJ Aging Mech Dis, 3(1):1-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [470].Yamamoto T,Byun J,Zhai P,Ikeda Y,Oka S,Sadoshima J (2014). Nicotinamide mononucleotide, an intermediate of NAD+ synthesis, protects the heart from ischemia and reperfusion. PloS one, 9(6):e98972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [471].Park JH,Long A,Owens K,Kristian T (2016). Nicotinamide mononucleotide inhibits post-ischemic NAD+ degradation and dramatically ameliorates brain damage following global cerebral ischemia. Neurobiol Dis, 95:102-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [472].Williams PA,Harder JM,Foxworth NE,Cochran KE,Philip VM,Porciatti V,Smithies O,John SW (2017). Vitamin B3 modulates mitochondrial vulnerability and prevents glaucoma in aged mice. Science, 355(6326):756-60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [473].Mitchell SJ,Bernier M,Aon MA,Cortassa S,Kim EY,Fang EF,Palacios HH,Ali A,Navas-Enamorado I,Di Francesco A,Kaiser TA (2018). Nicotinamide improves aspects of health-span, but not life-span, in mice. Cell Metab, 27(3):667-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [474].Fang EF,Lautrup S,Hou Y,Demarest TG,Croteau DL,Mattson MP,Bohr VA (2017). NAD+ in aging: molecular mechanisms and translational implications. Trends Mol Med, 23(10):899-916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [475].Johnson S,Imai SI (2018). NAD+ biosynthesis, aging, and disease. F1000Research, 7-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [476].Cantó C,Houtkooper RH,Pirinen E, et al. (2012). The NAD+ precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity. Cell Metab, 15(6):838-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [477].Poljsak B,Milisav I (2016). NAD+ as the link between oxidative stress, inflammation, caloric restriction, exercise, DNA repair, longevity, and health span, Rejuvenation Res, 19(5):406-13. [DOI] [PubMed] [Google Scholar]
  • [478].Trammell SA,Weidemann BJ,Chadda A,Yorek MS,Holmes A,Coppey LJ,Obrosov A,Kardon RH,Yorek MA,Brenner C (2016). Nicotinamide riboside opposes type 2 diabetes and neuropathy in mice. Scientific Rep, 6:26933. 1-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [479].Trammell SA,Schmidt MS,Weidemann BJ,Redpath P,Jaksch F,Dellinger RW,Li Z,Abel ED,Migaud ME,Brenner C (2016). Nicotinamide riboside is uniquely and orally bioavailable in mice and humans. Nature Commun, 7:12948. 1-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [480].Long AN,Owens K,Schlappal AE,Kristian T,Fishman PS,Schuh RA (2015). Effect of nicotinamide mononucleotide on brain mitochondrial respiratory deficits in an Alzheimer's disease-relevant murine model. BMC Neurology, 15(1):1-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [481].Wang X,Hu X,Yang Y,Takata T,Sakurai T (2016). Nicotinamide mononucleotide protects against ß-amyloid oligomer-induced cognitive impairment and neuronal death. Brain Res, 1643:1-9. [DOI] [PubMed] [Google Scholar]
  • [482].Yao Z,Yang W,Gao Z,Jia P (2017). Nicotinamide mononucleotide inhibits JNK activation to reverse Alzheimer disease. Neurosci Lett, 647:133-40. [DOI] [PubMed] [Google Scholar]
  • [483].Kiss T,Balasubramanian P,Valcarcel-Ares MN,Tarantini S,Yabluchanskiy A,Csipo T,Lipecz A,Reglodi D,Zhang XA,Bari F,Farkas E (2019). Nicotinamide mononucleotide (NMN) treatment attenuates oxidative stress and rescues angiogenic capacity in aged cerebromicrovascular endothelial cells: a potential mechanism for the prevention of vascular cognitive impairment. Geroscience, 41(5):619-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [484].Gong B,Pan Y,Vempati P,Zhao W,Knable L,Ho L,Wang J,Sastre M,Ono K,Sauve AA,Pasinetti GM (2013). Nicotinamide riboside restores cognition through an upregulation of proliferator-activated receptor-? coactivator 1a regulated ß-secretase 1 degradation and mitochondrial gene expression in Alzheimer's mouse models. Neurobiol Aging, 34(6):1581-1588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [485].Schöndorf DC,Ivanyuk D,Baden P,Sanchez-Martinez A,De Cicco S,Yu C,Giunta I,Schwarz LK,Di Napoli G,Panagiotakopoulou V,Nestel S (2018). The NAD+ precursor nicotinamide riboside rescues mitochondrial defects and neuronal loss in iPSC and fly models of Parkinson's disease. Cell reports, 23(10):2976-2988. [DOI] [PubMed] [Google Scholar]
  • [486].Sorrentino V,Romani M,Mouchiroud L,Beck JS,Zhang H,D'Amico D,Moullan N,Potenza F,Schmid AW,Rietsch S,Counts SE (2017). Enhancing mitochondrial proteostasis reduces amyloid-ß proteotoxicity. Nature, 552(7684):187-193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [487].Hou Y,Lautrup S,Cordonnier S,Wang Y,Croteau DL,Zavala E,Zhang Y,Moritoh K,O'Connell JF,Baptiste BA,Stevnsner TV (2018). NAD+ supplementation normalizes key Alzheimer's features and DNA damage responses in a new AD mouse model with introduced DNA repair deficiency. Proc Natl Acad Sci USA, 115(8):E1876-1885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [488].Gallo CM,Smith DL,Smith JS (2004). Nicotinamide clearance by Pnc1 directly regulates Sir2-mediated silencing and longevity. Mol Cell Biol, 24(3):1301-1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [489].van der Horst A,Schavemaker JM,Pellis-van Berkel W,Burgering BM (2007). The Caenorhabditis elegans nicotinamidase PNC-1 enhances survival. Mech Ageing Dev, 128(4):346-349. [DOI] [PubMed] [Google Scholar]
  • [490].Schmeisser K,Mansfeld J,Kuhlow D,Weimer S,Priebe S,Heiland I,Birringer M,Groth M,Segref A,Kanfi Y,Price NL (2013). Role of sirtuins in life-span regulation is linked to methylation of nicotinamide. Nature Chem Biol, 9(11):693-700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [491].Yoshino J,Mills KF,Yoon MJ,Imai SI (2011). Nicotinamide mononucleotide, a key NAD+ intermediate, treats the pathophysiology of diet-and age-induced diabetes in mice. Cell Metab, 14(4):528-536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [492].Misiak M,Vergara Greeno R,Baptiste BA,Sykora P,Liu D,Cordonnier S,Fang EF,Croteau DL,Mattson MP,Bohr VA (2017). DNA polymerase ß decrement triggers death of olfactory bulb cells and impairs olfaction in a mouse model of Alzheimer's disease. Aging Cell, 16(1):162-172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [493].Aman Y,Qiu Y,Tao J,Fang EF (2018). Therapeutic potential of boosting NAD+ in aging and diseases. Trans Med Aging, 2:30-7. [Google Scholar]
  • [494].Belenky P,Racette FG,Bogan KL,McClure JM,Smith JS,Brenner C, (2007). Nicotinamide riboside promotes Sir2 silencing and extends life-span via Nrk and Urh1/Pnp1/Meu1 pathways to NAD+. Cell, 129(3):473-484. [DOI] [PubMed] [Google Scholar]
  • [495].Lu SP,Kato M,Lin SJ (2009). Assimilation of endogenous nicotinamide riboside is essential for calorie restriction-mediated life span extension in Saccharomyces cerevisiae. J Biol Chem, 284(25):17110-17119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [496].Tsang F,James C,Kato M,Myers V,Ilyas I,Tsang M,Lin SJ (2015). Reduced Ssy1-Ptr3-Ssy5 (SPS) signaling extends replicative life span by enhancing NAD+ homeostasis in Saccharomyces cerevisiae. J Biol Chem, 290(20):12753-12764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [497].Luchetti F,Canonico B,Betti M,Arcangeletti M, et al. (2010). Melatonin signaling and cell protection functioN, FASEB J, 24(10): 3603-3624. [DOI] [PubMed] [Google Scholar]
  • [498].Sandyk R (1996). Melatonin supplements for aging. International journal of neuroscience, 87(3-4): 219-224. [DOI] [PubMed] [Google Scholar]
  • [499].Grivas TB,Savvidou OD (2007). Melatonin the" light of night" in human biology and adolescent idiopathic scoliosis. Scoliosis, 2(1):1-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [500].Karasek M (2004). Melatonin, human aging, and diseases. Exp Gerontol, 39(11-12):1723-1729. [DOI] [PubMed] [Google Scholar]
  • [501].Karasek M (2007). Does melatonin play a role in aging processes? J Physiol Pharmacol, 58(6):105-113. [PubMed] [Google Scholar]
  • [502].Magri F,Sarra S,Cinchetti W,Guazzoni V, et al. (2004). Qualitative and quantitative changes of melatonin levels in physiological and pathological aging and in centenarians. J Pineal Res, 36(4):256-261. [DOI] [PubMed] [Google Scholar]
  • [503].Bonnefont-Rousselot D,Collin F (2010). Melatonin: action as antioxidant and potential applications in human disease and aging. Toxicol, 278(1):55-67. [DOI] [PubMed] [Google Scholar]
  • [504].Acuña-Castroviejo D,Carretero M,Doerrier C, et al. (2012). Melatonin protects lung mitochondria from aging. Age, 34(3):681-692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [505].Srinivasan V,Pandi-Perumal SR,Cardinali DP,Poeggeler B,Hardeland R (2006). Melatonin in Alzheimer's disease and other neurodegenerative disorders. Behav Brain Funct, 2(1):1-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [506].López A,García JA,Escames G, et al. (2009). Melatonin protects the mitochondria from oxidative damage reducing oxygen consumption, membrane potential, and superoxide anion production, J Pineal Res, 46:188-198. [DOI] [PubMed] [Google Scholar]
  • [507].Kleszczynski K,Fischer TW (2012). Melatonin and human skin aging. Dermato-endocrinol, 4(3):245-252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [508].Hardeland R (2013). Melatonin and the theories of aging: a critical appraisal of melatonin's role in antiaging mechanisms. J Pineal Res, 55(4): 325-356. [DOI] [PubMed] [Google Scholar]
  • [509].Kang JW,Hong JM,Lee SM (2016). Melatonin enhances mitophagy and mitochondrial biogenesis in rats with carbon tetrachloride-induced liver fibrosis. J Pineal Res, 60(4): 383-393. [DOI] [PubMed] [Google Scholar]
  • [510].Yoon YM,Kim HJ,Lee JH,Lee SH (2019). Melatonin enhances mitophagy by upregulating expression of heat shock 70 kDa protein 1L in human mesenchymal stem cells under oxidative stress. Intl J Mol Sci, 20(18):4545. 1-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [511].Gutierrez-Cuesta J,Tajes M,Jiménez A,Coto-Montes A,Camins A,Pallàs M (2008). Evaluation of potential pro-survival pathways regulated by melatonin in a murine senescence model. J Pineal Res, 45(4): 497-505. [DOI] [PubMed] [Google Scholar]
  • [512].Ortega-Arellano HF,Jimenez-Del-Rio M,Velez-Pardo C (2021). Melatonin Increases Life Span, Restores the Locomotor Activity, and Reduces Lipid Peroxidation (LPO) in Transgenic Knockdown Parkin Drosophila melanogaster Exposed to Paraquat or Paraquat/Iron. Neurotox Res, 1-13. [DOI] [PubMed] [Google Scholar]
  • [513].Rodríguez MI,Escames G,López LC, et al. (2008). Improved mitochondrial function and increased life span after chronic melatonin treatment in senescent prone mice. Exp Gerontol, 43(8):749-756. [DOI] [PubMed] [Google Scholar]
  • [514].Sanchez-Barcelo EJ,Mediavilla MD,Tan DX,Reiter RJ (2010). Clinical uses of melatonin: evaluation of human trials. Curr Med Chem, 17(19):2070-2095. [DOI] [PubMed] [Google Scholar]
  • [515].Harrison AP,Pierzynowski SG (2008). Biological effects of 2-oxoglutarate with particular emphasis on the regulation of protein, mineral and lipid absorption/metabolism, muscle performance, kidney function, bone formation and cancerogenesis, all viewed from a healthy ageing perspective state of the art-review article. J Physiol Pharmacol, 59(Suppl 1):91-106. [PubMed] [Google Scholar]
  • [516].Wu N,Yang M,Gaur U,Xu H,Yao Y,Li D (2016). Alpha-ketoglutarate: physiological functions and applications. Biomol Therapeutics, 24(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [517].Riedel E,Nündel M,Hampl H (1996). α-Ketoglutarate application in hemodialysis patients improves amino acid metabolism. Nephron, 74(2):261-265. [DOI] [PubMed] [Google Scholar]
  • [518].Dakshayani KB,Subramanian P (2006). α-ketoglutarate modulates the circadian patterns of lipid peroxidation and antioxidant status during N-nitrosodiethylamine-induced hepatocarcinogenesis in rats. [J] Med Food. 9(1):90-97. [DOI] [PubMed] [Google Scholar]
  • [519].Bayliak MM,Lylyk MP,Shmihel HV,Sorochynska OM, et al. (2017). Dietary alpha-ketoglutarate promotes higher protein and lower triacylglyceride levels and induces oxidative stress in larvae and young adults but not in middle-aged Drosophila melanogaster. Comp Biochem Physiol A Mol Integr Physiol, 204: 28-39. [DOI] [PubMed] [Google Scholar]
  • [520].Yao K,Yin Y,Li X,Xi P,Wang J,Lei J,Hou Y,Wu G (2012). Alpha-ketoglutarate inhibits glutamine degradation and enhances protein synthesis in intestinal porcine epithelial cells. Amino acids, 42(6): 2491-2500. [DOI] [PubMed] [Google Scholar]
  • [521].Bayliak M, Burdyliuk N, Lushchak V, (2017). Growth on alpha-ketoglutarate increases oxidative stress resistance in the yeast Saccharomyces cerevisiae. Intl J Microbiol, (2017). 1-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [522].Zdzisińska B,Żurek A,Kandefer-Szerszeń M (2017). Alpha-ketoglutarate as a molecule with pleiotropic activity: well-known and novel possibilities of therapeutic use. Archivum immunologiae et therapiae experimentalis, 65(1):21-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [523].Niemiec T,Sikorska J,Harrison A,Szmidt M, et al. (2011). Alpha-ketoglutarate stabilizes redox homeostasis and improves arterial elasticity in aged mice. J Phys Pharma, 62(1):37. [PubMed] [Google Scholar]
  • [524].McLain AL,Szweda PA,Szweda LI (2011). α-Ketoglutarate dehydrogenase: a mitochondrial redox sensor. Free radical Res, 45(1):29-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [525].Bayliak MM,Lylyk MP, Sorochynska OM (2017). Dietary alpha-ketoglutarate partially prevents decline in locomotor activity and cold tolerance in Drosophila melanogaster. Biologia, 72(4):458-467. [Google Scholar]
  • [526].Liu S,He L,Yao K (2018). The antioxidative function of alpha-ketoglutarate and its applications. BioMed Res Intl, 2018:1-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [527].Bayliak MM,Hrynkiv OV,Knyhynytska RV,Lushchak VI (2018). Alpha-ketoglutarate enhances freeze-thaw tolerance and prevents carbohydrate-induced cell death of the yeast Saccharomyces cerevisiae. Arch Micobiol, 200(1):33-46. [DOI] [PubMed] [Google Scholar]
  • [528].Colomb V,Dabbas M,Goulet O,Talbotec C,Corriol O,Ricour C (2002). Prepubertal growth in children with long-term parenteral nutrition. Horm Res Paediatr, 58(Suppl. 1): 2-6. [DOI] [PubMed] [Google Scholar]
  • [529].Cynober L (2004). Ornithine α-ketoglutarate as a potent precursor of arginine and nitric oxide: a new job for an old friend. J Nutrition, 134(10): 2858S-2862S. [DOI] [PubMed] [Google Scholar]
  • [530].Carey BW,Finley LW,Cross JR,Allis CD,Thompson CB (2015). Intracellular α-ketoglutarate maintains the pluripotency of embryonic stem cells. Nature, 518(7539):413-416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [531].TeSlaa T,Chaikovsky AC,Lipchina I, et al. (2016). α-Ketoglutarate accelerates the initial differentiation of primed human pluripotent stem cells. Cell Metab, 24(3):485-493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [532].Wang Y,Deng P,Liu Y,Wu Y, et al. (2020). Alpha-ketoglutarate ameliorates osteoporosis via regulating histone methylations. Nature Commun, 11(1): 1-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [533].Zhang Z,He C,Zhang L,Zhu T, et al. (2019). Alpha-ketoglutarate affects murine embryo development through metabolic and epigenetic modulations. Reproduction, 158(2):123-133. [DOI] [PubMed] [Google Scholar]
  • [534].Satpute R,Lomash V,Kaushal M,Bhattacharya R (2013). Neuroprotective effects of alpha-ketoglutarate and ethyl pyruvate against motor dysfunction and oxidative changes caused by repeated 1-methyl-4-phenyl-1, 2, 3, 6 tetrahydropyridine exposure in mice. Human Exp Toxicol, 32(7):747-758. [DOI] [PubMed] [Google Scholar]
  • [535].Li Y,Li X,Gao Y,Huang C,Lin D (2021). NMR-Based Metabolomic Analysis for the Effects of α-Ketoglutarate Supplementation on C2C12 Myoblasts in Different Energy States. Molecules, 26(7):1841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [536].An D,Zeng Q,Zhang P,Ma Z, et al. (2021). Alpha-ketoglutarate ameliorates pressure overload-induced chronic cardiac dysfunction in mice. Redox Biol, 46:102088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [537].Harrison AP,Bruggemann D,Bartels EM,Andrea K,Pierzynowski S (2009). Healthy ageing: the beneficial effect of dietary supplementation with alpha-ketoglutarate on arterial elasticity in elderly mice. J Pre-Clin Clin Res, 3(1). [Google Scholar]
  • [538].Tian Q, Liu X, Du M, (2020). Alpha-ketoglutarate for adipose tissue rejuvenation. Aging (Albany NY), 12(14). p.13845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [539].Son ED,Choi GH,Kim H,Lee B,Chang IS,Hwang JS (2007). Alpha-ketoglutarate stimulates procollagen production in cultured human dermal fibroblasts, and decreases UVB-induced wrinkle formation following topical application on the dorsal skin of hairless mice. Biol Pharmaceutical Bull, 30(8):1395-1399. [DOI] [PubMed] [Google Scholar]
  • [540].Żurek A,Mizerska-Kowalska M,Sławińska-Brych A, et al. (2019). Alpha ketoglutarate exerts a pro-osteogenic effect in osteoblast cell lines through activation of JNK and mTOR/S6K1/S6 signaling pathways. Toxicol Applied Pharmacol. 374: 53-64. [DOI] [PubMed] [Google Scholar]
  • [541].Dobrowolski P,Tomaszewska E,Bienko M,Radzki RP,Pierzynowski SG (2013). The effect of dietary administration of 2-oxoglutaric acid on the cartilage and bone of growing rats. Br J Nutr, 110(4):651-658. [DOI] [PubMed] [Google Scholar]
  • [542].Dobrowolski P,Tomaszewska E,Radzki RP, et al. (2013). Can 2-oxoglutarate prevent changes in bone evoked by omeprazole? Nutr, 29(3):556-561. [DOI] [PubMed] [Google Scholar]
  • [543].Harrison AP,Tygesen MP,Sawa-Wojtanowicz B,Husted S,Tatara MR (2004). α-Ketoglutarate treatment early in postnatal life improves bone density in lambs at slaughter. Bone, 35(1):204-209. [DOI] [PubMed] [Google Scholar]
  • [544].Śliwa E (2010). 2-Oxoglutaric acid administration diminishes fundectomy-induced osteopenia in pigs. J Animal Phys Animal Nutr, 94(5): e86-e95. [DOI] [PubMed] [Google Scholar]
  • [545].Radzki RP,Bieńko M,Filip R,Pierzynowski SG (2016). The protective and therapeutic effect of exclusive and combined treatment with Alpha-ketoglutarate sodium salt and ipriflavone on bone loss in orchidectomized rats. J Nut Health Aging, 20(6):628-636. [DOI] [PubMed] [Google Scholar]
  • [546].Wang Y, Deng P., Liu Y, Wu Y, Chen Y, Guo Y, Zhang S, Zheng X., Zhou L, Liu W, Li Q., (2020). Alpha-ketoglutarate ameliorates osteoporosis via regulating histone methylations. Nature Commun, 11(1). 1-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [547].He L,Li H,Huang N,Zhou X, et al. (2017). Alpha-ketoglutarate suppresses the NF-kB-mediated inflammatory pathway and enhances the PXR-regulated detoxification pathway. Oncotarge, 8(61):102974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [548].Tian Q,Bravo Iniguez A,Sun Q, et al. (2021). Dietary Alpha-Ketoglutarate Promotes Epithelial Metabolic Transition and Protects against DSS-Induced Colitis. Mol Nutr Food Res, 65(7):936. [DOI] [PubMed] [Google Scholar]
  • [549].Hou Y,Wang L,Ding B, et al. (2011). Alpha-ketoglutarate and intestinal function, Front Biosci, 16: 1186-1196. [DOI] [PubMed] [Google Scholar]
  • [550].Tian Q,Zhao J,Yang Q, et al. (2020). Dietary alpha-ketoglutarate promotes beige adipogenesis and prevents obesity in middle-aged mice. Aging Cell, 19(1):e13059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [551].Okabe K,Nawaz A,Nishida Y, et al. (2020). NAD+ metabolism regulates preadipocyte differentiation by enhancing α-ketoglutarate-mediated histone H3K9 demethylation at the PPARγ promoter. Front Cell Dev Biol, 8:1409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [552].Zhang Z,He C,Gao Y,Zhang L, et al. (2021). α-ketoglutarate delays age-related fertility decline in mammals. Aging Cell, 20(2):e13291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [553].Radzki RP,Bieńko M,Pierzynowski SG (2009). Effect of dietary alpha-ketoglutarate on blood lipid profile during hypercholesterolaemia in rats. Scand J Clin Lab Invest, 69(2): 175-180. [DOI] [PubMed] [Google Scholar]
  • [554].Chin RM,Fu X,Pai MY, et al. (2014). The metabolite α-ketoglutarate extends lifespan by inhibiting ATP synthase and TOR. Nature, 510(7505):397-401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [555].Shahmirzadi AA,Edgar D,Liao CY, et al. (2020). Alpha-ketoglutarate, an endogenous metabolite, extends lifespan and compresses morbidity in aging mice. Cell Metab, 32(3):447-456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [556].Su Y,Wang T,Wu N,Li D,Fan X,Xu Z,Mishra SK,Yang M (2019). Alpha-ketoglutarate extends Drosophila lifespan by inhibiting mTOR and activating AMPK. Aging,11(12):4183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [557].He L,Zhou X,Huang N, et al. (2017). Administration of alpha-ketoglutarate improves epithelial restitution under stress injury in early-weaning piglets. Oncotarget, 8(54): 91965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [558].Bayliak MM,Lushchak VI (2020). Pleiotropic effects of alpha-ketoglutarate as a potential anti-ageing agent. Ageing Res Rev, 101237. [DOI] [PubMed] [Google Scholar]
  • [559].Ostrakhovitch EA,Tabibzadeh S (2015). Homocysteine in chronic kidney disease. Adv Clin Chem, 72: 77-106. [DOI] [PubMed] [Google Scholar]
  • [560].Fowler B (2005). May. Homocysteine: overview of biochemistry, molecular biology, and role in disease processes. Seminars Vas Med, 5: 77-86. [DOI] [PubMed] [Google Scholar]
  • [561].Tinelli C,Di Pino A,Ficulle E,Marcelli S,Feligioni M (2019). Hyperhomocysteinemia as a risk factor and potential nutraceutical target for certain pathologies. Front Nutr, 6, 49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [562].Skovby F, (1989). Inborn errors of metabolism causing homocysteinemia and related vascular involvement. Pathophysiol Haemostasis Thromb, 19(Suppl. 1). 4-9. [DOI] [PubMed] [Google Scholar]
  • [563].Kuo HK, Sorond FA, Chen JH, Hashmi A, Milberg W.P, Lipsitz LA, (2005). The role of homocysteine in multisystem problems: a systematic review. The Journals of Gerontology Series A: Biol Sci Med Sci, 60(9). 1190-1201. [DOI] [PubMed] [Google Scholar]
  • [564].Kang SS,Wong PW,Malinow MR (1992). Hyperhomocyst (e) inemia as a risk factor for occlusive vascular disease. An Rev Nut, 12(1):279-298. [DOI] [PubMed] [Google Scholar]
  • [565].Refsum H, Smith AD, Ueland PM, et al. (2004). Facts and recommendations about total homocysteine determinations: an expert opinioN, Clin Chem, 50(1): 3-32. [DOI] [PubMed] [Google Scholar]
  • [566].Kado DM,Bucur A,Selhub J,Rowe JW,Seeman T (2002). Homocysteine levels and decline in physical function: MacArthur Studies of Successful Aging. Am J Med, 113(7):537-542. [DOI] [PubMed] [Google Scholar]
  • [567].Majumder A,Singh M,George AK,Tyagi SC (2019). Restoration of skeletal muscle homeostasis by hydrogen sulfide during hyperhomocysteinemia-mediated oxidative/er stress condition, Canadian J Phys Pharma, 97(6):441-456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [568].Jakubowski H,Zhang L,Bardeguez A,Aviv A (2000). Homocysteine thiolactone and protein homocysteinylation in human endothelial cells: implications for atherosclerosis. Circ Res, 87(1):45-51. [DOI] [PubMed] [Google Scholar]
  • [569].Bełtowski J (2005). Protein homocysteinylation: a new mechanism of atherogenesis? Modyfikacja białek przez tiolakton homocysteiny-nowy mechanizm powstawania miażdżycy? Postepy Hig Med Dosw, 59:392-404. [PubMed] [Google Scholar]
  • [570].Eren E,Yilmaz N,Aydin O (2012). High density lipoprotein and it’s dysfunction, The Open Biochem J, 6:78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [571].Fournier I,Ploye F,Cottet-Emard JM,Brun J,Claustrat B (2002). Folate deficiency alters melatonin secretion in rats. J Nutr, 132(9):2781-2784. [DOI] [PubMed] [Google Scholar]
  • [572].Challet E,Dumont S,Mehdi MK,Allemann C, et al. (2013). Aging-like circadian disturbances in folate-deficient mice. Neurobiol Aging, 34(6):1589-1598. [DOI] [PubMed] [Google Scholar]
  • [573].Romerowicz-Misielak M,Kozioł K,Nowak S,Koziorowski M (2021). Altered circadian dynamics of Per2 after cystathionine-β-synthase and/or cystathionine-γ-lyase pharmacological inhibition in serum-shocked NIH-3T3 cells. Archives of Biochemistry and Biophysics, 697:108713. [DOI] [PubMed] [Google Scholar]
  • [574].Carson NA,Neill DW (1962). Metabolic abnormalities detected in a survey of mentally backward individuals in Northern Ireland. Arch Dis Child. 37:505-513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [575].Testai FD,Gorelick PB (2010). Inherited metabolic disorders and stroke part 2: homocystinuria, organic acidurias, and urea cycle disorders. Arch Neurol. 67(2):148-153. [DOI] [PubMed] [Google Scholar]
  • [576].Booth GL,Wang EE (2000). Preventive health care, (2000) update: screening and management of hyperhomocysteinemia for the prevention of coronary artery disease events. CMAJ, 163(1): 21-29. [PMC free article] [PubMed] [Google Scholar]
  • [577].Selhub J,Jacques PF,Wilson PW,Rush D,Rosenberg IH (1993). Vitamin status and intake as primary determinants of homocysteinemia in an elderly population, JAMA. 270:2693-2698 [DOI] [PubMed] [Google Scholar]
  • [578].Smith AD,Smith SM,De Jager CA, et al. (2010). Homocysteine-lowering by B vitamins slows the rate of accelerated brain atrophy in mild cognitive impairment: a randomized controlled trial. PloS one, 5(9):e12244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [579].Siniscalchi A,Gallelli L,Mercuri NB,Ferreri Ibbadu G,De Sarro G (2006). Role of lifestyle factors on plasma homocysteine levels in Parkison's disease patients treated with levodopa. Nut Neurosci, 9(1-2):11-16. [DOI] [PubMed] [Google Scholar]
  • [580].Harman D (1956). Aging: a theory based on free radical and radiation chemistry. J Gerontol, 11(3): 298-300. [DOI] [PubMed] [Google Scholar]
  • [581].Harman D (1968). Free radical theory of aging: effect of free radical reaction inhibitors on the mortality rate of male LAF1 mice. J Gerontol, 23(4):476-82. [DOI] [PubMed] [Google Scholar]
  • [582].Johnson F,Giulivi C (2005). Superoxide dismutases and their impact upon human health. Mol Aspects Med, 26(4-5):340-352. [DOI] [PubMed] [Google Scholar]
  • [583].Pearl R (1928). The rate of living, London: University Press, 356-359. [Google Scholar]
  • [584].Chance B,Sies H,Boveris A (1979). Hydroperoxide metabolism in mammalian organs. Phys Rev, 59(3):527-605. [DOI] [PubMed] [Google Scholar]
  • [585].Halliwell B (1989). Free radicals, reactive oxygen species and human disease: a critical evaluation with special reference to atherosclerosis. Br J Exp Pathol, 70(6):737. [PMC free article] [PubMed] [Google Scholar]
  • [586].Packer L,Fuehr K (1977). Low oxygen concentration extends the lifespan of cultured human diploid cells. Nature, 267(5610):423-425. [DOI] [PubMed] [Google Scholar]
  • [587].Yu BP (1994). Cellular defenses against damage from reactive oxygen species. Phys Rev, 74(1):139-62. [DOI] [PubMed] [Google Scholar]
  • [588].Beckman KB,Ames BN (1998). The free radical theory of aging matures. Phys Rev, 78, 547-581. [DOI] [PubMed] [Google Scholar]
  • [589].Sohal RS,Sohal BH (1991). Hydrogen peroxide release by mitochondria increases during aging. Mech Ageing Dev, 57(2):187-202. [DOI] [PubMed] [Google Scholar]
  • [590].Cutler RG (1991). Antioxidants and aging. Am J Clin Nutr, 53(1 Suppl): 373S-379S. [DOI] [PubMed] [Google Scholar]
  • [591].Haining JL, Legan JS (1973). Catalase turnover in rat liver and kidney as a function of age. Exp Gerontol, 8(2): 85-91. [DOI] [PubMed] [Google Scholar]
  • [592].Morgan MJ,Liu ZG (2011). Crosstalk of reactive oxygen species and NF-kappaB signaling. Cell Res, 21(1): 103-115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [593].Zhu S,Li W,Zhou H,Wei W, et al. (2010). Reprogramming of human primary somatic cells by OCT4 and chemical compounds. Cell Stem Cell, 7(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [594].Li Y,Zhang Q,Yin X,Yang W, et al.2011). Generation of iPSCs from mouse fibroblasts with a single gene, Oct4, and small molecules. Cell Res, 21(1):196-204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [595].Yuan X,Wan H,Zhao X,Zhu S,Zhou Q,Ding S (2011). Brief report: combined chemical treatment enables Oct4-induced reprogramming from mouse embryonic fibroblasts. Stem Cells, 29(3):549-553 [DOI] [PubMed] [Google Scholar]
  • [596].Ma T,Xie M,Laurent T,Ding S (2013). Progress in the reprogramming of somatic cells. Circulation Res, 112(3):562-574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [597].Hou P,Li Y,Zhang X,Liu C, et al. (2013). Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds. Science, 341(6146):651-654. [DOI] [PubMed] [Google Scholar]
  • [598].Tran KA,Dillingham CM,Sridharan R (2019). The role of α-ketoglutarate-dependent proteins in pluripotency acquisition and maintenance. J Biol Chem, 294(14):5408-5419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [599].Du Y,Liu Z,Cao X,Chen X, et al. (2017). Nucleosome eviction along with H3K9ac deposition enhances Sox2 binding during human neuroectodermal commitment. Cell Death Diff, 24(6):1121-1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [600].Leitch HG, McEwen KR, et al. (2013). Naive pluripotency is associated with global DNA hypomethylatioN, Nature, Struct Mol Biol, 20(3):311-316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [601].Plath K,Lowry WE (2011). Progress in understanding reprogramming to the induced pluripotent state. Nature, Rev Genetics, 12(4): 253-265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [602].Mikkelsen TS,Hanna J,Zhang X, et al. (2008). Dissecting direct reprogramming through integrative genomic analysis. Nature, 454(7200):49-55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [603].Gao Y,Chen J,Li K,Wu T, et al. (2013). Replacement of Oct4 by Tet1 during iPSC induction reveals an important role of DNA methylation and hydroxymethylation in reprogramming. Cell Stem Cell, 12(4):453-469. [DOI] [PubMed] [Google Scholar]
  • [604].Rada-Iglesias A,Wysocka J (2011). Epigenomics of human embryonic stem cells and induced pluripotent stem cells: insights into pluripotency and implications for disease. Genome Med, 3(6):1-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [605].Sridharan R,Gonzales-Cope M,Chronis C, et al. (2013). Proteomic and genomic approaches reveal critical functions of H3K9 methylation and heterochromatin protein-1γ in reprogramming to pluripotency. Nature, Cell Biol, 15(7):872-882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [606].Chen J,Liu H,Liu J,Qi J, et al. (2013). H3K9 methylation is a barrier during somatic cell reprogramming into iPSCs. Nature Genetics, 45(1):34-42. [DOI] [PubMed] [Google Scholar]
  • [607].Boland MJ,Nazor KL,Loring JF (2014). Epigenetic regulation of pluripotency and differentiation, Circulation Res, 115(2):311-324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [608].Fu E,Shen J,Dong Z,Zhang W,Zhang Y,Chen F,Cheng Z,Zhao X, Shua L, Lu X (2019). Histone demethylase Kdm2a regulates germ cell genes and endogenous retroviruses in embryonic stem cells. Epigenomics, 11(7):751-766. [DOI] [PubMed] [Google Scholar]
  • [609].He S,Sun H,Lin L,Zhang Y, et al. (2017). Passive DNA demethylation preferentially up-regulates pluripotency-related genes and facilitates the generation of induced pluripotent stem cells. J Biol Chem, 292(45):18542-18555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [610].Pereira CF,Piccolo FM,Tsubouchi T, et al. (2010). ESCs require PRC2 to direct the successful reprogramming of differentiated cells toward pluripotency. Cell Stem Cell, 6(6):547-556. [DOI] [PubMed] [Google Scholar]
  • [611].Rao RA,Dhele N,Cheemadan S,Ketkar A, et al. (2015). Ezh2 mediated H3K27me3 activity facilitates somatic transition during human pluripotent reprogramming. Sci Rep, 5(1):1-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [612].Zhang YL,Zhao LW,Zhang J, et al. (2018). DCAF 13 promotes pluripotency by negatively regulating SUV 39H1 stability during early embryonic development. EMBO J, 37(18):e98981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [613].Dawlaty MM,Ganz K,Powell BE, et al. (2011). Tet1 is dispensable for maintaining pluripotency and its loss is compatible with embryonic and postnatal development. Cell Stem Cell, 9(2):166-175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [614].Fu X,Wu X,Djekidel MN,Zhang Y (2019). Myc and Dnmt1 impede the pluripotent to totipotent state transition in embryonic stem cells. Nature Cell Biol, 21(7):835-844 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [615].Liang G,Zhang Y (2013). Embryonic stem cell and induced pluripotent stem cell: an epigenetic perspective. Cell Res, 23(1):49-69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [616].Wang T,Chen K,Zeng X, et al. (2011). The histone demethylases Jhdm1a/1b enhance somatic cell reprogramming in a vitamin-C-dependent manner. Cell Stem Cell, 9(6):575-587. [DOI] [PubMed] [Google Scholar]
  • [617].Hu X,Zhang L,Mao SQ,Li Z, et al. (2014). Tet and TDG mediate DNA demethylation essential for mesenchymal-to-epithelial transition in somatic cell reprogramming. Cell Stem Cell, 14(4):512-522. [DOI] [PubMed] [Google Scholar]
  • [618].Doege CA,Inoue K,Yamashita T, et al. (2012). Early-stage epigenetic modification during somatic cell reprogramming by Parp1 and Tet2. Nature, 488(7413):652-655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [619].Schwarz SD,Grundbacher E,Hrovat AM, et al. (2020). Inducible TDG knockout models to study epigenetic regulation. F1000Research, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [620].Rada-Iglesias A,Wysocka J (2011). Epigenomics of human embryonic stem cells and induced pluripotent stem cells: insights into pluripotency and implications for disease. Genome Med, 3(6):1-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [621].Gonzales-Cope M,Sidoli S,Bhanu NV,Won KJ,Garcia BA, (2016). Histone H4 acetylation and the epigenetic reader Brd4 are critical regulators of pluripotency in embryonic stem cells. BMC Genomics, 17(1):1-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [622].Hezroni H, Sailaja BS, Meshorer E, (2011). Pluripotency-related, valproic acid (VPA)-induced genome-wide histone H3 lysine 9 (H3K9) acetylation patterns in embryonic stem cells. J Biol Chem, 286(41). 35977-35988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [623].Hezroni H,Tzchori I,Davidi A, et al. (2011). H3K9 histone acetylation predicts pluripotency and reprogramming capacity of ES cells. Nucleus, 2(4):300-309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [624].Lv L,Lu X,Fen T,ur Rehman S, et al. (2020). Valproic acid enhances in vitro developmental competence of porcine handmade cloned embryos. Livestock Science, 233:103957. [Google Scholar]
  • [625].Tafrova JI,Tafrov ST (2014). Human histone acetyltransferase 1 (Hat1) acetylates lysine 5 of histone H2A in vivo. Mol Cell Biochem Valproic acid enhances in vitro developmental competence of porcine handmade cloned embryos. Livestock Science 392(1):259-272. [DOI] [PubMed] [Google Scholar]
  • [626].Cosentino MS,Oses C,Echegaray CV, et al. (2019). Kat6b modulates Oct4 and Nanog binding to chromatin in embryonic stem cells and is required for efficient neural differentiation, J Mol Biol Valproic acid enhances in vitro developmental competence of porcine handmade cloned embryos. Livestock Science, 431(6):1148-1159. [DOI] [PubMed] [Google Scholar]
  • [627].Wiesel-Motiuk N,Assaraf YG (2020). The key roles of the lysine acetyltransferases KAT6A and KAT6B in physiology and pathology. Drug Resistance Updates, 100729. [DOI] [PubMed] [Google Scholar]
  • [628].Mu X,Yan S,Fu C,Wei A (2015). The histone acetyltransferase MOF promotes induces generation of pluripotent stem cells. Cellular Reprogram, 17(4):259-267. [DOI] [PubMed] [Google Scholar]
  • [629].Li X,Li L,Pandey R,Byun JS, et al. (2012). The histone acetyltransferase MOF is a key regulator of the embryonic stem cell core transcriptional network. Cell Stem Cell, 11(2):163-178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [630].He R,Dantas A,Riabowol K (2021). Histone acetyltransferases and stem cell identity. Cancers, 13(10):2407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [631].Jamaladdin S,Kelly RD,O’Regan L, et al. (2014). Histone deacetylase (HDAC) 1 and 2 are essential for accurate cell division and the pluripotency of embryonic stem cells. Proc Natl Acad Sci USA, 111(27): 9840-9845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [632].Kidder BL,Palmer S (2012). HDAC1 regulates pluripotency and lineage specific transcriptional networks in embryonic and trophoblast stem cells. Nucl Acids Res, 40(7):2925-2939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [633].Williams EO,Taylor AK,Bell EL, et al. (2016). Sirtuin 1 promotes deacetylation of Oct4 and maintenance of naive pluripotency. Cell Reports, 17(3):809-820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [634].Xu P,Wang TT,Liu XZ,Wang NY, et al. (2019). Sirt6 regulates efficiency of mouse somatic reprogramming and maintenance of pluripotency. Stem cell Res Therapy, 10(1):1-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [635].Etchegaray JP Chavez L,Huang Y, et al. (2015). The histone deacetylase SIRT6 controls embryonic stem cell fate via TET-mediated production of 5-hydroxymethylcytosine. Nature Cell Biol, 17(5):545-557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [636].Chang AR,Ferrer CM,Mostoslavsky R (2020). SIRT6, a mammalian deacylase with multitasking abilities. Phys Rev, 100(1):145-169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [637].Sharma A,Diecke S,Zhang WY, et al. (2013). The role of SIRT6 protein in aging and reprogramming of human induced pluripotent stem cells. J Biol Chem, 288(25):18439-18447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [638].Freund A,Laberge RM,Demaria M,Campisi J (2012). Lamin B1 loss is a senescence-associated biomarker. Mol Biol Cell, 23(11):2066-2075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [639].Wang Y,Tian C,Zheng JC (2013). FoxO3a contributes to the reprogramming process and the differentiation of induced pluripotent stem cells. Stem Cell Dev, 22(22):2954-2963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [640].Jang J,Wang Y,Kim HS,Lalli MA,Kosik KS (2014). Nrf2, a regulator of the proteasome, controls self-renewal and pluripotency in human embryonic stem cells. Stem Cells, 32(10):2616-2625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [641].Papapetrou EP,Tomishima MJ,Chambers SM, et al. (2009). Stoichiometric and temporal requirements of Oct4, Sox2, Klf4, and c-Myc expression for efficient human iPSC induction and differentiatioN, Proc Natl Acad Sci USA, 106(31):12759-12764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [642].Okita K,Yamanaka S (2011). Induced pluripotent stem cells: opportunities and challenges. Philosophical Transactions of the Royal Society B: Biol Sci, 366(1575): 2198-2207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [643].Araki R,Hoki Y,Uda M, et al. (2011). Crucial role of c-Myc in the generation of induced pluripotent stem cells. Stem Cells, 29(9):1362-1370. [DOI] [PubMed] [Google Scholar]
  • [644].Carey BW,Finley LW,Cross JR,Allis CD,Thompson CB (2015). Intracellular α-ketoglutarate maintains the pluripotency of embryonic stem cells. Nature, 518(7539):413-416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [645].Dutta D (2017). Histone chaperone in regulation of cellular metabolism dictating stem cell fate?. Stem cell investigation, 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [646].Roberti A,Fernández AF,Fraga MF (2021). Nicotinamide N-methyltransferase: At the crossroads between cellular metabolism and epigenetic regulation, Mol Metab, 101165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [647].Blundell G,Jones BG,Rose FA,Tudball N (1996). Homocysteine mediated endothelial cell toxicity and its amelioration, Atherosclerosis, 122(2):163-172. [DOI] [PubMed] [Google Scholar]
  • [648].Makhro AV,Mashkina AP,Solenaya OA, et al. (2008). Carnosine protects cells from oxidative stress induced by hyperhomocysteinemia. Neurochem J, 2(3):202-208. [Google Scholar]
  • [649].Boldyrev AA (2009). Molecular mechanisms of homocysteine toxicity. Biochem, 74(6):589-598. [DOI] [PubMed] [Google Scholar]
  • [650].Nielsen FH (2009). Boron deprivation decreases liver S-adenosylmethionine and spermidine and increases plasma homocysteine and cysteine in rats. J Trace Elements Med Biol, 23(3):204-213. [DOI] [PubMed] [Google Scholar]
  • [651].Karolczak K,Watala C (2021). Melatonin as a Reducer of Neuro-and vasculotoxic Oxidative Stress Induced by Homocysteine. Antioxidants, 10(8):1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [652].Lavie L,Lavie P (2004). Daily rhythms in plasma levels of homocysteine. J Circadian Rhythms, 2(1):1-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [653].Baydas G,Gursu MF,Cikim G, et al. (2002). Effects of pinealectomy on the levels and the circadian rhythm of plasma homocysteine in rats. J Pineal Res, 33(3):151-155. [DOI] [PubMed] [Google Scholar]
  • [654].Maron BA,Loscalzo J (2009). The treatment of hyperhomocysteinemia. Ann Rev Med, 60:39-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [655].Baydas G,Özer M,Yasar A,Tuzcu M,Koz ST (2005). Melatonin improves learning and memory performances impaired by hyperhomocysteinemia in rats. Brain Res, 1046(1-2):187-194. [DOI] [PubMed] [Google Scholar]
  • [656].Soenen S,Rayner CK,Jones KL,Horowitz M (2016). The ageing gastrointestinal tract. Current Clin Nut Metabolic Care, 19(1):12-18. [DOI] [PubMed] [Google Scholar]

Articles from Aging and Disease are provided here courtesy of JKL International LLC

RESOURCES