Büşra Nur Dal1, İlknur Altuntaş1, Oytun Erbaş1

1ERBAS Institute of Experimental Medicine, Illinois, USA & Gebze, Türkiye

Keywords: Aging, anti-aging, cellular aging, DNA damage, oxidative stress, protein modifications

Abstract

Numerous theories exist concerning aging and its causes. Understanding the factors contributing to aging allows for the implementation of necessary precautions, and while complete prevention of aging may not be achievable, significant strides can be made. Recent advancements in genetic science have brought aging research to a prominent position. Studies on aging will play a crucial role in the future discourse on longevity, emphasizing the importance of the current state of genetic research in this field. The collective findings in this review contribute to a comprehensive understanding of aging, laying the groundwork for future discussions on longevity and the intricate interplay of genetics in the aging process.

Introduction

Aging and longevity are determined by a complex interplay of genetic, non-genetic, and environmental factors.[1] Throughout history, humankind has perennially questioned, ‘Why do we age?’ or ‘Why am I aging?’ and has endeavored to find answers. Presently, research is underway to investigate the causes of aging, with various theories proposed to explain the underlying reasons for the aging process.[2] Aging is highly variable, dependent on the rates at which changes occur throughout an organism’s lifespan.[3] It is recognized that aging is a biological condition arising from stress, manifesting in both physiological and pathological processes.[4] Studies elucidating the pace and overall impact of cellular aging define it as a state of cellular quiescence, irreversibility, and reduced replicative capacity relative to aging.[5]

BIOGERONTOLOGY

Biogerontology is referred to as the science of biological aging, examining the effects of aging on living organisms. The field dealing with the treatment of diseases related to old age is known as ‘Geriatrics.’ It is assumed that aging arises from the Gerontogenic effect. Biological aging is not controlled by a specific mechanism or regulator. Longevity-correlation analyses conducted on the lifespan of monozygotic and dizygotic twins have indicated that genes have approximately a 25% effect on lifespan. Non-genetic factors, such as environmental factors, healthy nutrition, physical activity, etc., have been observed to contribute to lifespan by more than 75%.[6,7]

The field of ‘Gerontology’ studies the biochemical processes and alterations in gene activity that affect genes influencing longevity. The emergence of aging is assumed to involve two types of gerontogenic interactions: the discovery of late-acting mutations that manifest their effects later during fertilization and birth, and the period of growth, development, and maturation.[8] In economically developed countries, the average lifespan is between 80 and 85 years. The recorded maximum human lifespan to date is 122 years, five months, and 14 days.[9]

Genes involved in repair and maintenance pathways, crucial for longevity, can have gerontological conditions affecting the aging phenotype. These genetic pathways are classified as general and specific pathways.[10]

GENETIC FACTORS CONTRIBUTING TO AGING

Telomere Shortening

Genes responsible for telomere extension do not interact during cell division, resulting in telomere shortening after each division. Once telomeres reach a certain length, cell division ceases, and cellular aging begins. While telomeres are active in germ cells, they remain passive in somatic cells. Germ cells, being passed on to the next generation, maintain their active state.[11]

Olovnikov[12] first discovered in 1973 that telomere shortening could control lifespan. In 1990, insights into telomere structure were elucidated through the work of scientists Harley et al.[13]

Aging in human cells occurs in two stages:

M1 Phase: Cell division halts, and aging initiates once telomeres reach a specific length. The cell cannot progress from the G0 or G1 phase to the S phase, leading to a halt in division. If the cell becomes unable to divide, it begins to age. Active expression of oncogenes, including Werner syndrome and various genetic diseases, occurs due to changes in telomere length and function.

M2 Phase: To transition from M1 to M2 phase, the p53 and Rb-like proteins of the cell are disrupted by viral oncogenes during M1. These proteins, not found in the G1 phase, skip to the G2 phase and proceed to the S phase, allowing cell division to continue. In somatic cells, telomerase enzyme activity decreases, leading to telomere shortening. Cells die at M2 if telomeres become excessively short. If telomere length remains at a certain level at M2, cells surpass M2 and continue dividing, a phenomenon regulated or reactivated by telomerase enzyme regulation.[14]

Deoxyribonucleic acid (DNA) tumor viruses such as simian virus 40, human papillomavirus, and Adenovirus, along with chemical carcinogens and radiation, can prevent cellular aging through the transformation process. Tumor viruses bind to tumor cells and deactivate the cell suppressor tumors, p53, and p110Rb proteins. Evidence suggests that the viral transformation of p53 and p110Rb molecules leads to an extension of lifespan by inhibiting cellular inactivation. Immortal cells lacking p53 and p100Rb molecules are observed in cases of mutations and chromosomal abnormalities.[15]

Telomeres are structures containing non-coding DNA with the sequence 5’-TTAGGG-3’.[16] Transcription occurring in the telomerase reverse transcriptase (TERT) promoter ensures the maintenance of telomerase levels in various cell types. Lack of TERT expression in human fibroblasts results in telomere shortening, leading to aging after cell division and replication.[17] Telomerase’s primary function is to confer immortality to cells by preserving the integration of chromosome ends. DNA polymerase synthesizes telomeres through reverse transcription, preventing the fusion of chromosomes. Telomerase has significant implications in diseases such as cancer metabolism and aging.[18]

The initial evidence suggesting that telomere length contributes to aging was observed in primary fibroblasts, where telomeres shortened with increasing donor age, leading to replicative senescence when telomeres reached a critically short length.[19]

Telomerase was first discovered in the ciliated protozoan Tetrahymena thermophila, which divides its macronuclear genome into 20,000 small chromosomes.[20] Telomeres, formed by the assembly of double-stranded TTAGGG repeats through specific proteins, protect the genomic structure by preventing chromosomal fusion. Telomere length varies within each cell, and human leukocytes typically have an average of 92 telomeres of varying lengths.[21] Humans are born with telomeres ranging from 5 to 15 kb, influenced by environmental factors. Telomere shortening, varying between 20-50 bp, occurs due to oxidative stress and various factors damaging DNA.[22]

DNA Damage

DNA damage occurs spontaneously in cells that make up the human body every day, alongside changes in DNA methylation and histone modifications. These damages lead to cell cycle arrest due to the halting of DNA and ribonucleic acid (RNA) polymerases, initiating a response indicating the need for DNA repair. Conditions such as apoptosis and senescence contribute to aging. DNA repair induces epigenomic changes that assist in the repair process.[23]

While DNA damage was initially thought to cause genetic instability, recent in vivo and in vitro studies suggest that DNA inflammation induces type 1 interferons and other inflammatory mechanisms within the cell. Accumulation of DNA damage in cells due to prolonged stimuli leads to chronic inflammation, tissue degeneration, and functional impairment with aging, and indirectly activates pro-inflammatory signals of DNA damage response.[24,25]

An example linking persistent DNA damage to premature aging is observed in Fanconi anemia, an autosomal genetic disorder causing advanced bone marrow failure through hematopoietic stem and progenitor cells.[26]

Mammalian cells contain two genomes: nuclear and mitochondrial. Nuclear DNA includes approximately 20,000-25,000 intergenic genomes, while mitochondrial DNA has a circular plasmid containing 37 genes that code for 16,569 bases. Inherited mtDNA mutations have been implicated in various human diseases.[27,28]

Various factors influencing stress can trigger cellular stress. Nuclear DNA damage, often occurring with double-strand breaks, is fundamental to senescence. Senescence, discovered through telomere loss in human fibroblasts, limits cell numbers and induces cell cycle arrest through DNA damage.[29]

Oxidative stress facilitates DNA damage, contributing to aging. Factors inducing DNA damage, including oxidative stress, lead to damage to DNA bases or single-strand breaks. Increased oxidative stress accelerates telomere shortening at the ends of chromosomes.[30,31]

Although limited information is available on the transmembrane protein PLA2R1’s role in cancer and aging, studies have shown that it induces cellular aging by increasing reactive oxygen species (ROS) production and the amount of DNA damage, ultimately leading to cell death.[32]

Studies indicate that Rapamycin extends the lifespan of various organisms, including mice, Caenorhabditis elegans, Drosophila, and Hydra. [33] The translation of p53 results in an increase in DNA damage.[34]

CELLULAR DEATH AND OXIDATIVE STRESS IN AGING

Aging leads to a decrease in cell numbers due to programmed cell death occurring in various cell types.[35]

The oxidative stress theory of aging is based on the accumulation of oxidative damage in macromolecules through ROS, leading to functional losses associated with aging.[36] This theory has gained acceptance in long-lived species, including Saccharomyces cerevisiae, transgenic mice, C. elegans, birds, and the naked mole-rat (Heterocephalus glaber). Malondialdehyde and 4-hydroxynonenal are used to determine oxidative stress, both being toxic and mutagenic aldehydes.[37] Proposed by Harman[38] in 1956, the theory suggests that elevated ROS levels facilitate oxidative stress, causing structural damage to DNA macromolecules and resulting in damage at the cellular and tissue levels.[39,40]

Oxidative stress increases mitochondrial membrane permeability, leading to the release of factors that restrict cell survival, causing tissue damage through apoptosis and necrosis due to ROS.[41]

Reactive oxygen species serve as radicals to generate molecular oxygen, arising from enzymatic and non-enzymatic mechanisms. Various antioxidants remove ROS from within the cell.[42] Reactive oxygen species have short lifespans and rapidly react with biomolecules to alter their activities. Low levels of ROS within the cell allow for normal cellular function through redox signaling.[43] An increase in ROS formation with a decrease in neutralization leads to an elevation in oxidative stress at the cellular and tissue levels.[44,45]

Oxidative stress induces damage by causing lipid peroxidation in mitochondrial membranes and other membranes, as well as irreversible modifications in nucleic acids.[44] Activation of nicotinamide adenine dinucleotide phosphate oxidase leads to oxidative stress, increasing the levels of ROS.[46] Studies have shown that caffeine, an alkaloid, has effects on aging and oxidative stress. For instance, in human vascular endothelial cells, caffeine has been observed to prevent aging induced by oxidative stress. The effects of low concentrations of caffeine on cellular and tissue-level changes associated with aging caused by oxidative stress remain uncertain.[47]

The rise in oxidative stress with a decrease in mitochondrial antioxidant levels disrupts cell signaling, leading to the loss of cell and tissue homeostasis and promoting aging.[48] The continuous accumulation of oxidative damage in proteins, cellular lipids, or DNA accelerates the rate of aging.[49] Oxidative damage and chronic inflammation, linked to aging at the cellular and tissue levels, form a foundation that encourages functional decline in cardiovascular and skeletal muscle systems.[50] Oxidative stress increases significantly with intense exercise and ischemia, compounding the effects of aging.[51]

As mitochondrial oxidative damage increases with age, it alters mitochondrial dynamics by balancing fission, fusion, and autophagy processes.[52] Oxidative stress not only affects cell survival, apoptosis, and cancer migration but also possesses bactericidal properties.[53] Studies indicate that dysfunctional mitochondria affected by an increased level of oxidative stress play a role in the pathogenesis of neurodegenerative disorders such as Alzheimer’s, Parkinson’s, and Huntington’s diseases.[54] Cumulative oxidative stress emerges as a primary factor contributing to aging and age-related neurodegenerative disorders in cellular, molecular, and behavioral studies.[55]

Given the high oxygen consumption in the retina, it exhibits resistance to oxidative stress.[56] Mitochondrial and motor impairments specific to Parkinson’s disease arise in the midbrain’s substantia nigra region due to the death of dopaminergic neurons triggered by oxidative stress.[57]

During aging, mitochondria without any function produce more ROS, leading to increased damage to telomeres due to oxidative breakdown. Telomeric DNA damage caused by oxidative stress has been strongly evidenced in human and animal models to accelerate rapid telomere shortening.[58] Intestinal aging induces structural changes and increases oxidative stress, significantly impacting the health of the elderly.[59] As endogenous antioxidant systems become less effective with age, elderly individuals become more sensitive to oxidative stress.[60] Nitro-oxidative stress plays a crucial role in endothelial cell dysfunction and inflammation.[61]

Exposure to an excessive amount of ROS results in oxidative stress-mediated cellular damage, causing oxidation of biomolecules, including DNA, proteins, and lipids.[62] The generation of oxidative stress and inflammatory agents will lead to increased endoplasmic reticulum stress and mitochondrial dysfunction.[63]

CELLULAR AGING

Cellular aging involves irreversible losses resulting from the loss of the replicative capacity of primary cells initially due to DNA damage occurring in defective telomeres. Telomere dysfunction arises from telomere shortening during DNA replication.[64] Cellular aging represents the state of the cell stress response, characterized by morphological and biochemical changes. It is a complex process where cells remain active, but the cell cycle stops.[65,66] Cellular aging was first defined using non-dividing diploid fibroblast cell lines by Hayflick and Moorhead[67] in 1961. It is a condition where physiological functions, such as cellular differentiation, decrease over time. The number of aging cells increases with age in various tissues and organs. Cellular aging is one of the most significant features of aging.[68,69]

Several conditions contribute to cellular aging, including DNA damage, inflammation, oncogenes, mitogens, reactive metabolites, proteotoxic stress, and damage occurring in molecular patterns.[70] Cellular aging is a state where repeated symmetric cell copies, critically short telomeres, and the permanent cessation of the cell cycle due to the DNA damage response occur. Most aging cells show epigenetic and chromatin structural changes.[71]

Studies have demonstrated that excessive mitochondrial DNA mutations lead to physiological mitochondrial dysfunction and premature aging.[72,73] Cellular aging can facilitate wound healing. For example, inflammation-induced cellular aging reduces the critical role of fibroblast migration and proliferation in the formation of new tissue when new tissue formation occurs.[74] Cellular aging is a controlled process involving both positive and negative conditions such as embryonic development, wound healing, tumor suppression, and aging.[75]

Flow cytometry imaging is a device used to observe in vivo aging cells, enabling the evaluation of numerous aging markers at the single-cell level and their detection at the protein level.[76] Aging cells contribute to the prevention of tumor formation by causing cell cycle arrest.[77] Hypotheses suggest that aging cells, accumulating in various tissues over time, age, and space, contribute to impairments associated with many chronic diseases in humans and animals.[78] Cancer cells are susceptible to many stress factors that contribute to aging, such as oncogenic signaling, replicative stress, hypoxia, ROS, and nutrient deprivation. Some anti-cancer treatments also trigger the aging of cancer cells. Different types of leukocytes mediate the removal of aging cells by the immune system. Most of these leukocytes are present in the innate immune system.[79] DNA damage, telomere shortening, oncogene activation, metabolic signals, mechanical stress, and mitochondrial dysfunction trigger the formation of aging cells.[80]

PROTEIN MODIFICATIONS

Amino acids, particularly tyrosine, are found around the protein shell, and these proteins are known to contribute to biological aging and age-related diseases.[81] Cellular changes occur significantly with aging. Caveolin‐1 has been suggested as a marker in fibroblasts and endothelial cells.[82] Many cellular and signaling pathways, including mammalian targets of rapamycin (mTOR), Sirtuin 1, and AMP-activated protein kinase, contribute to aging. The mTOR contributes to the extension of human life and the slowing of the aging process. Autophagy and inflammation have a significant impact during the aging process.[83] The accumulation of damaged proteins has been implicated in age-related diseases such as aging, type 2 diabetes, cancer, neurodegenerative disorders, cardiovascular diseases, and visible impairments.[84]

Accumulation of damage in cellular proteins, lipids, and cell organelles occurs during the aging process, leading to disorders at the cellular, organellar, and organ levels, contributing to age-related diseases and cell and organism death.[85] Amino acid limitation reduces age-related DNA damage and extends lifespan by blocking the Tor1/Sch9 cascade. All amino acids contribute to cellular sensitization.[86,87]

ENVIRONMENTAL FACTORS

The effects of exercise on longevity have been observed in genetically determined long-lived individuals such as centenarians.[88] A significantly less recognized factor is air pollution. Rapid industrialization and urbanization have turned environmental pollution into a public health issue. In 2019, the WHO determined that 99% of the world’s population lives in places where air pollution levels exceed WHO limits, identifying air pollution as the single largest environmental health factor for humans. Air pollution has been identified as a cause of degenerative formations in skin aging, pigmentary problems in the skin, and the onset of skin disorders.[89]

PROGERIA SYNDROME

The expression of progerin leads to nuclear morphological abnormalities, misregulated gene expression, chromatin changes, mitochondrial dysfunction, defects in DNA repair, and rapid telomere shortening, promoting cellular decline and causing premature aging. The most characteristic feature in the cytology of fibroblasts of HutchinsonGilford progeria syndrome patients is the occurrence of nuclear morphological abnormalities.[90]

Genomic instability occurs due to deficiencies in DNA repair in premature aging syndromes, creating a state of instability that facilitates the early and high occurrence of cancer cases. This instability also contributes to the acceleration of aging and cancer processes.[91] Hutchinson-Gilford progeria syndrome is a condition in children characterized by gradual aging at the cellular and organismal levels. Jonathan Hutchinson discovered this disease in 1886. Progeria occurs due to a mutation in lamin A, encoded by the LMNA gene.[92] Hutchinson-Gilford progeria syndrome is observed in one in 4-8 million newborns, and aging symptoms appear 18-24 months after birth. These symptoms include growth retardation, thin and wrinkled skin, abnormal pigmentation, subcutaneous fat loss, joint stiffening, and weakened bone structure.[93]

LONGEVITY GENES

A significant portion of people wish to live longer, especially when their health is in good condition, and some aspire to extend their lives limitlessly. Two different pathways are identified for yeast Ras2 activation: protein kinase A (PKA) and mitogenactivated protein kinase pathways. There is evidence supporting the role of the PKA pathway in promoting longevity.[94] Naked mole-rats have a longer lifespan, while hypomorphic ribosomal yeast mutants have a shorter lifespan. Accuracy in yeast mitochondrial ribosomes is associated with longevity.[95]

Genes associated with increased lifespan have been discovered in yeast, worms, flies, and mice, resulting in respective enhancements in longevity.[96,97] Numerous theories have been proposed regarding the evolution of aging, including the programmed death theory, the mutation accumulation theory of aging, the antagonistic pleiotropy theory of aging, and the evolutionary maintenance theory.[97] The autophagic decline is observed in many organisms with aging.[98,99]

ANTI-AGING

Various treatments, including medications, exercise programs, and hormone therapies, are spreading worldwide as medical interventions to mitigate the effects of aging. The global market for anti-aging health products is steadily increasing.[100]

Conditions such as telomere loss, genomic instability, epigenetic changes, proteostasis loss, irregular nutrient sensing, and mitochondrial dysfunction are notable molecular and cellular occurrences in the aging process.[101]

Mutations in mitochondrial DNA in elderly individuals lead to genomic instability. Telomeres are located at the ends of chromosomes and facilitate cellular aging once they reach a critical length known as the Hayflick limit. Aging is a state of decreased bodily functions.[102] The skin is affected by aging, resulting in thinning, loss of elasticity, and dermal-epidermal flattening.[103,104] Harrison found that rapamycin has been observed to increase survival rates by 14% in females and 9% in males.[105]

In conclusion, the intricate nature of aging involves a dynamic interplay between genetic, non-genetic, and environmental factors. Biogerontology explores the effects of aging on living organisms, emphasizing the multifaceted influences that contribute to longevity. Telomere shortening, DNA damage, and oxidative stress emerge as pivotal factors in cellular aging, influencing processes such as apoptosis and senescence. The role of longevity genes, identified in various organisms, sheds light on potential pathways for extending lifespan. Progeria syndrome exemplifies how genetic mutations can accelerate aging, while environmental factors, including air pollution, also play a role in age-related skin disorders. The somatic mutation theory highlights the impact of DNA damage on aging, emphasizing the need for cellular responses and repair mechanisms. Overall, the comprehensive understanding of these molecular and cellular processes contributes to ongoing research in antiaging interventions and underscores the complex dynamics that govern the aging phenomenon.

Cite this article as: Dal BN, Altuntaş İ, Erbaş O. Genetic Aspects of Aging and Anti-Aging Strategies. JEB Med Sci 2023;4(3):156-164.

Conflict of Interest

The authors declared no conflicts of interest with respect to the authorship and/or publication of this article.

Financial Disclosure

The authors received no financial support for the research and/or authorship of this article.

References

  1. Mirzaei H, Di Biase S, Longo VD. Dietary Interventions, Cardiovascular Aging, and Disease: Animal Models and Human Studies. Circ Res. 2016 May 13;118:1612-25.
  2. Guarente L, Kenyon C. Genetic pathways that regulate ageing in model organisms. Nature. 2000 Nov 9;408:255-62.
  3. Kirkwood TB. Understanding the odd science of aging. Cell. 2005 Feb 25;120:437-47.
  4. Kuilman T, Michaloglou C, Mooi WJ, Peeper DS. The essence of senescence. Genes Dev. 2010 Nov 15;24:2463-79.
  5. Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, et al. Cellular Senescence: Defining a Path Forward. Cell. 2019 Oct 31;179:813-27.
  6. Rattan SIS. Biogerontology: research status, challenges and opportunities. Acta Biomed. 2018 Jun 7;89:291-301.
  7. Brooks-Wilson AR. Genetics of healthy aging and longevity. Hum Genet. 2013 Dec;132:1323-38.
  8. Rattan SIS. Biogerontology: research status, challenges and opportunities. Acta Biomed. 2018 Jun 7;89:291-301.
  9. Naumova E, Ivanova M, Pawelec G. Immunogenetics of ageing. Int J Immunogenet. 2011 Oct;38:373-81.
  10. Rodop BB, Erbaş O. Mitochondrial dysfunction in aging and disease: The role of nutrition. D J Med Sci 2023;9:43-55.
  11. Gao X, Yu X, Zhang C, Wang Y, Sun Y, Sun H, et al. Telomeres and Mitochondrial Metabolism: Implications for Cellular Senescence and Age-related Diseases. Stem Cell Rev Rep. 2022 Oct;18:2315-27.
  12. Olovnikov AM. A theory of marginotomy. The incomplete copying of template margin in enzymic synthesis of polynucleotides and biological significance of the phenomenon. J Theor Biol. 1973 Sep 14;41:181-90.
  13. Harley CB, Futcher AB, Greider CW. Telomeres Shorten During Ageing of Human Fibroblasts. Nature 1990;345:458-60.
  14. Polat Y, Erbaş O. Heart Rate Dynamics and Its Role in Aging and Lifespan. JEB Med Sci 2023;4:140-48.
  15. Bryan TM, Reddel RR. Telomere Dynamics and Telomerase Activity in In vivo Immortalised Human Cells. Europ J Canc 1997;33:767-73.
  16. Diehl MC, Idowu MO, Kimmelshue KN, York TP, Jackson-Cook CK, Turner KC, et al. Elevated TRF2 in advanced breast cancers with short telomeres. Breast Cancer Res Treat. 2011 Jun;127:623-30.
  17. Roake CM, Artandi SE. Regulation of human telomerase in homeostasis and disease. Nat Rev Mol Cell Biol. 2020 Jul;21:384-97.
  18. Judasz E, Lisiak N, Kopczyński P, Taube M, Rubiś B. The Role of Telomerase in Breast Cancer's Response to Therapy. Int J Mol Sci. 2022 Oct 25;23:12844.
  19. Bernardes de Jesus B, Blasco MA. Telomerase at the intersection of cancer and aging. Trends Genet. 2013 Sep;29:513-20.
  20. Miracco EJ, Jiang J, Cash DD, Feigon J. Progress in structural studies of telomerase. Curr Opin Struct Biol. 2014 Feb;24:115-24.
  21. Daşdelen D, Kayaaltı A, Altuntaş İ, Erbaş O. N-Methyl D-Aspartic Acid Receptors: An Overview. JEB Med Sci 2022;3:118-24.
  22. Sagris M, Theofilis P, Antonopoulos AS, Tsioufis K, Tousoulis D. Telomere Length: A Cardiovascular Biomarker and a Novel Therapeutic Target. Int J Mol Sci. 2022 Dec 16;23:16010.
  23. Soto-Palma C, Niedernhofer LJ, Faulk CD, Dong X. Epigenetics, DNA damage, and aging. J Clin Invest. 2022 Aug 15;132:e158446.
  24. Zhao Y, Simon M, Seluanov A, Gorbunova V. DNA damage and repair in age-related inflammation. Nat Rev Immunol. 2023 Feb;23:75-89.
  25. Arvanitaki ES, Stratigi K, Garinis GA. DNA damage, inflammation and aging: Insights from mice. Front Aging. 2022 Sep 7;3:973781.
  26. Ou HL, Schumacher B. DNA damage responses and p53 in the aging process. Blood. 2018 Feb 1;131:488-495.
  27. Patel J, Baptiste BA, Kim E, Hussain M, Croteau DL, Bohr VA. DNA damage and mitochondria in cancer and aging. Carcinogenesis. 2020 Dec 31;41:1625-34.
  28. Babbar M, Basu S, Yang B, Croteau DL, Bohr VA. Mitophagy and DNA damage signaling in human aging. Mech Ageing Dev. 2020 Mar;186:111207.
  29. Di Micco R, Krizhanovsky V, Baker D, d'Adda di Fagagna F. Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nat Rev Mol Cell Biol. 2021 Feb;22:75-95.
  30. Schumacher B, Pothof J, Vijg J, Hoeijmakers JHJ. The central role of DNA damage in the ageing process. Nature. 2021 Apr;592:695-703.
  31. Campisi J. Aging, cellular senescence, and cancer. Annu Rev Physiol. 2013;75:685-705.
  32. Huna A, Griveau A, Vindrieux D, Jaber S, Flaman JM, Goehrig D, et al. PLA2R1 promotes DNA damage and inhibits spontaneous tumor formation during aging. Cell Death Dis. 2021 Feb 16;12:190.
  33. Blagosklonny MV. DNA- and telomere-damage does not limit lifespan: evidence from rapamycin. Aging (Albany NY). 2021 Feb 12;13:3167-75.
  34. Steffens Reinhardt L, Groen K, Newton C, Avery-Kiejda KA. The role of truncated p53 isoforms in the DNA damage response. Biochim Biophys Acta Rev Cancer. 2023 May;1878:188882.
  35. Tower J. Programmed cell death in aging. Ageing Res Rev. 2015 Sep;23:90-100.
  36. Liguori I, Russo G, Curcio F, Bulli G, Aran L, Della-Morte D, et al. Oxidative stress, aging, and diseases. Clin Interv Aging. 2018 Apr 26;13:757-72.
  37. Kudryavtseva AV, Krasnov GS, Dmitriev AA, Alekseev BY, Kardymon OL, Sadritdinova AF, et al. Mitochondrial dysfunction and oxidative stress in aging and cancer. Oncotarget. 2016 Jul 19;7:44879-905.
  38. HARMAN D. Aging: a theory based on free radical and radiation chemistry. J Gerontol. 1956 Jul;11:298-300.
  39. Aykan K, Altuntaş İ, Erbaş O. DNA repair mechanisms: DNA repair defects and related diseases. D J Med Sci 2022;8:130-40.
  40. Moldogazieva NT, Mokhosoev IM, Mel'nikova TI, Porozov YB, Terentiev AA. Oxidative Stress and Advanced Lipoxidation and Glycation End Products (ALEs and AGEs) in Aging and Age-Related Diseases. Oxid Med Cell Longev. 2019 Aug 14;2019:3085756.
  41. Vatner SF, Zhang J, Oydanich M, Berkman T, Naftalovich R, Vatner DE. Healthful aging mediated by inhibition of oxidative stress. Ageing Res Rev. 2020 Dec;64:101194.
  42. Cencioni C, Spallotta F, Martelli F, Valente S, Mai A, Zeiher AM, et al. Oxidative stress and epigenetic regulation in ageing and age-related diseases. Int J Mol Sci. 2013 Aug 28;14:17643-63.
  43. Cahill-Smith S, Li JM. Oxidative stress, redox signalling and endothelial dysfunction in ageing-related neurodegenerative diseases: a role of NADPH oxidase 2. Br J Clin Pharmacol. 2014 Sep;78:441-53.
  44. Mecocci P, Boccardi V, Cecchetti R, Bastiani P, Scamosci M, Ruggiero C, et al. A Long Journey into Aging, Brain Aging, and Alzheimer's Disease Following the Oxidative Stress Tracks. J Alzheimers Dis. 2018;62:1319-35.
  45. Hajam YA, Rani R, Ganie SY, Sheikh TA, Javaid D, Qadri SS, et al. Oxidative Stress in Human Pathology and Aging: Molecular Mechanisms and Perspectives. Cells. 2022 Feb 5;11:552.
  46. Pala HG, Erbas O, Pala EE, Artunc Ulkumen B, Akman L, Akman T, et al. The effects of sunitinib on endometriosis. J Obstet Gynaecol. 2015 Feb;35:183-7.
  47. Li YF, Ouyang SH, Tu LF, Wang X, Yuan WL, Wang GE, et al. Caffeine Protects Skin from Oxidative Stress-Induced Senescence through the Activation of Autophagy. Theranostics. 2018 Nov 10;8:5713-30.
  48. Coryell PR, Diekman BO, Loeser RF. Mechanisms and therapeutic implications of cellular senescence in osteoarthritis. Nat Rev Rheumatol. 2021 Jan;17:47-57.
  49. Li L, Chen B, Zhu R, Li R, Tian Y, Liu C, et al. Fructus Ligustri Lucidi preserves bone quality through the regulation of gut microbiota diversity, oxidative stress, TMAO and Sirt6 levels in aging mice. Aging (Albany NY). 2019 Nov 12;11:9348-68.
  50. El Assar M, Álvarez-Bustos A, Sosa P, Angulo J, Rodríguez-Mañas L. Effect of Physical Activity/Exercise on Oxidative Stress and Inflammation in Muscle and Vascular Aging. Int J Mol Sci. 2022 Aug 5;23:8713.
  51. Bartnik BL, Juurlink BH, Devon RM. Macrophages: their myelinotrophic or neurotoxic actions depend upon tissue oxidative stress. Mult Scler. 2000 Feb;6:37-42.
  52. Lee S, Jeong SY, Lim WC, Kim S, Park YY, Sun X, et al. Mitochondrial fission and fusion mediators, hFis1 and OPA1, modulate cellular senescence. J Biol Chem. 2007 Aug 3;282:22977-83.
  53. Rahimifard M, Baeeri M, Mousavi T, Azarnezhad A, Haghi-Aminjan H, Abdollahi M. Combination therapy of cisplatin and resveratrol to induce cellular aging in gastric cancer cells: Focusing on oxidative stress, and cell cycle arrest. Front Pharmacol. 2023 Jan 4;13:1068863.
  54. Taylor E, Kim Y, Zhang K, Chau L, Nguyen BC, Rayalam S, et al. Antiaging Mechanism of Natural Compounds: Effects on Autophagy and Oxidative Stress. Molecules. 2022 Jul 8;27:4396.
  55. Singh P, Barman B, Thakur MK. Oxidative stress-mediated memory impairment during aging and its therapeutic intervention by natural bioactive compounds. Front Aging Neurosci. 2022 Jul 25;14:944697.
  56. Zhao B, Zhu L, Ye M, Lou X, Mou Q, Hu Y, et al. Oxidative stress and epigenetics in ocular vascular aging: an updated review. Mol Med. 2023 Feb 27;29:28.
  57. Hernández-Cruz EY, Eugenio-Pérez D, Ramírez-Magaña KJ, Pedraza-Chaverri J. Effects of Vegetal Extracts and Metabolites against Oxidative Stress and Associated Diseases: Studies in Caenorhabditis elegans. ACS Omega. 2023 Feb 27;8:8936-59.
  58. Dokuyucu R, Kokacya H, Inanir S, Copoglu US, Erbas O. Antipsychotic-like effect of minocycline in a rat model. Int J Clin Exp Med. 2014 Oct 15;7:3354-61.
  59. Jiang N, Zhao Z. Intestinal aging is alleviated by uridine via regulating inflammation and oxidative stress in vivo and in vitro. Cell Cycle. 2022 Jul;21:1519-31.
  60. Gola F, Gaiaschi L, Roda E, De Luca F, Ferulli F, Vicini R, et al. Voghera Sweet Pepper: A Potential Ally against Oxidative Stress and Aging. Int J Mol Sci. 2023 Feb 14;24:3782.
  61. Bacanoiu MV, Danoiu M, Rusu L, Marin MI. New Directions to Approach Oxidative Stress Related to Physical Activity and Nutraceuticals in Normal Aging and Neurodegenerative Aging. Antioxidants (Basel). 2023 Apr 26;12:1008.
  62. Kristiani L, Kim Y. The Interplay between Oxidative Stress and the Nuclear Lamina Contributes to Laminopathies and Age-Related Diseases. Cells. 2023 Apr 25;12:1234.
  63. Dong W, Chen W, Zou H, Shen Z, Yu D, Chen W, et al. Ginsenoside Rb1 Prevents Oxidative Stress-Induced Apoptosis and Mitochondrial Dysfunction in Muscle Stem Cells via NF-κB Pathway. Oxid Med Cell Longev. 2022 Nov 24;2022:9159101.
  64. Miwa S, Kashyap S, Chini E, von Zglinicki T. Mitochondrial dysfunction in cell senescence and aging. J Clin Invest. 2022 Jul 1;132:e158447.
  65. Uyar B, Palmer D, Kowald A, Murua Escobar H, Barrantes I, Möller S, et al. Single-cell analyses of aging, inflammation and senescence. Ageing Res Rev. 2020 Dec;64:101156.
  66. Csekes E, Račková L. Skin Aging, Cellular Senescence and Natural Polyphenols. Int J Mol Sci. 2021 Nov 23;22:12641.
  67. Hayflick L, Moorhead PS. The Serial Cultivation of Human Telomerase Activity Diploid Cell Strains. Exp Cell Res 1961;25:585-621.
  68. Guo J, Huang X, Dou L, Yan M, Shen T, Tang W, et al. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Ther. 2022 Dec 16;7:391.
  69. Boyajian JL, Ghebretatios M, Schaly S, Islam P, Prakash S. Microbiome and Human Aging: Probiotic and Prebiotic Potentials in Longevity, Skin Health and Cellular Senescence. Nutrients. 2021 Dec 18;13:4550.
  70. Pignolo RJ, Passos JF, Khosla S, Tchkonia T, Kirkland JL. Reducing Senescent Cell Burden in Aging and Disease. Trends Mol Med. 2020 Jul;26:630-38.
  71. Liu J, Wang L, Wang Z, Liu JP. Roles of Telomere Biology in Cell Senescence, Replicative and Chronological Ageing. Cells. 2019 Jan 15;8:54.
  72. Di Micco R, Krizhanovsky V, Baker D, d'Adda di Fagagna F. Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nat Rev Mol Cell Biol. 2021 Feb;22:75-95.
  73. Miwa S, Kashyap S, Chini E, von Zglinicki T. Mitochondrial dysfunction in cell senescence and aging. J Clin Invest. 2022 Jul 1;132:e158447.
  74. Huang W, Hickson LJ, Eirin A, Kirkland JL, Lerman LO. Cellular senescence: the good, the bad and the unknown. Nat Rev Nephrol. 2022 Oct;18:611-27.
  75. Liu RM. Aging, Cellular Senescence, and Alzheimer's Disease. Int J Mol Sci. 2022 Feb 11;23:1989.
  76. Cohn RL, Gasek NS, Kuchel GA, Xu M. The heterogeneity of cellular senescence: insights at the single-cell level. Trends Cell Biol. 2023 Jan;33:9-17.
  77. Schmitt CA, Wang B, Demaria M. Senescence and cancer- role and therapeutic opportunities. Nat Rev Clin Oncol. 2022 Oct;19:619-36.
  78. Saul D, Kosinsky RL, Atkinson EJ, Doolittle ML, Zhang X, LeBrasseur NK, et al. A new gene set identifies senescent cells and predicts senescence-associated pathways across tissues. Nat Commun. 2022 Aug 16;13:4827.
  79. Marin I, Boix O, Garcia-Garijo A, Sirois I, Caballe A, Zarzuela E, et al. Cellular Senescence Is Immunogenic and Promotes Antitumor Immunity. Cancer Discov. 2023 Feb 6;13:410-31.
  80. Erdogan MA, Erdogan A, Erbas O. The Anti-Seizure Effect of Liraglutide on Ptz-Induced Convulsions Through its Anti-Oxidant and Anti-Inflammatory Properties. Neurochem Res. 2023 Jan;48:188-95.
  81. Fichtner M, Schuster S, Stark H. Influence of spatial structure on protein damage susceptibility: a bioinformatics approach. Sci Rep. 2021 Mar 2;11:4938.
  82. Kruglikov IL, Zhang Z, Scherer PE. Caveolin-1 in skin aging - From innocent bystander to major contributor. Ageing Res Rev. 2019 Nov;55:100959.
  83. Hassani B, Goshtasbi G, Nooraddini S, Firouzabadi N. Pharmacological Approaches to Decelerate Aging: A Promising Path. Oxid Med Cell Longev. 2022 Jul 11;2022:4201533.
  84. Aragonès G, Rowan S, Francisco SG, Whitcomb EA, Yang W, Perini-Villanueva G, et al. The Glyoxalase System in Age-Related Diseases: Nutritional Intervention as Anti-Ageing Strategy. Cells. 2021 Jul 22;10:1852.
  85. Rottenberg H, Hoek JB. The Mitochondrial Permeability Transition: Nexus of Aging, Disease and Longevity. Cells. 2021 Jan 6;10:79.
  86. Brandhorst S, Longo VD. Protein Quantity and Source, Fasting-Mimicking Diets, and Longevity. Adv Nutr. 2019 Nov 1;10:S340-S350.
  87. Al-Wahiby S, Wong HP, Slijepcevic P. Shortened telomeres in murine scid cells expressing mutant hRAD54 coincide with reduction in recombination at telomeres. Mutat Res. 2005 Oct 15;578:134-42.
  88. Aksoy D, Solmaz V, Çavuşoğlu T, Meral A, Ateş U, Erbaş O. Neuroprotective Effects of Eexenatide in a Rotenone-Induced Rat Model of Parkinson's Disease. Am J Med Sci. 2017 Sep;354:319-24.
  89. Martic I, Jansen-Dürr P, Cavinato M. Effects of Air Pollution on Cellular Senescence and Skin Aging. Cells. 2022 Jul 17;11:2220.
  90. Gonzalo S, Kreienkamp R, Askjaer P. Hutchinson-Gilford Progeria Syndrome: A premature aging disease caused by LMNA gene mutations. Ageing Res Rev. 2017 Jan;33:18-29.
  91. Paccosi E, Balajee AS, Proietti-De-Santis L. A matter of delicate balance: Loss and gain of Cockayne syndrome proteins in premature aging and cancer. Front Aging. 2022 Jul 21;3:960662.
  92. Lamis A, Siddiqui SW, Ashok T, Patni N, Fatima M, Aneef AN. Hutchinson-Gilford Progeria Syndrome: A Literature Review. Cureus. 2022 Aug 31;14:e28629.
  93. Erbaş O, Altuntaş İ, Çağlar Ö, Özyilmaz E, Sari E, Üzümcü İ, et al. Experimental Model of Cardiotoxicity [Internet]. Risk Factors for Cardiovascular Disease. IntechOpen; 2022. Available from: http://dx.doi. org/10.5772/intechopen.101401
  94. Taormina G, Ferrante F, Vieni S, Grassi N, Russo A, Mirisola MG. Longevity: Lesson from Model Organisms. Genes (Basel). 2019 Jul 9;10:518.
  95. Bjedov I, Rallis C. The Target of Rapamycin Signalling Pathway in Ageing and Lifespan Regulation. Genes (Basel). 2020 Sep 3;11:1043.
  96. Moskalev AA, Aliper AM, Smit-McBride Z, Buzdin A, Zhavoronkov A. Genetics and epigenetics of aging and longevity. Cell Cycle. 2014;13:1063-77.
  97. Yilmaz M, Aktug H, Oltulu F, Erbas O. Neuroprotective effects of folic acid on experimental diabetic peripheral neuropathy. Toxicol Ind Health. 2016 May;32:832-40.
  98. Hansen M, Rubinsztein DC, Walker DW. Autophagy as a promoter of longevity: insights from model organisms. Nat Rev Mol Cell Biol. 2018 Sep;19:579-93.
  99. Le Couteur DG, Barzilai N. New horizons in life extension, healthspan extension and exceptional longevity. Age Ageing. 2022 Aug 2;51:afac156.
  100. Nadeeshani H, Li J, Ying T, Zhang B, Lu J. Nicotinamide mononucleotide (NMN) as an anti-aging health product-Promises and safety concerns. J Adv Res. 2021 Aug 11;37:267-78.
  101. Du Y, Gao Y, Zeng B, Fan X, Yang D, Yang M. Effects of anti-aging interventions on intestinal microbiota. Gut Microbes. 2021 Jan-Dec;13:1994835.
  102. da Costa JP, Vitorino R, Silva GM, Vogel C, Duarte AC, Rocha-Santos T. A synopsis on aging-Theories, mechanisms and future prospects. Ageing Res Rev. 2016 Aug;29:90-112.
  103. Xue H, Thaivalappil A, Cao K. The Potentials of Methylene Blue as an Anti-Aging Drug. Cells. 2021 Dec 1;10:3379.
  104. Li Z, Zhang Z, Ren Y, Wang Y, Fang J, Yue H, et al. Aging and age-related diseases: from mechanisms to therapeutic strategies. Biogerontology. 2021 Apr;22:165-87.
  105. Fu W, Wu G. Targeting mTOR for Anti-Aging and Anti-Cancer Therapy. Molecules. 2023 Apr 1;28:3157.