Why do we age? The main signs of aging

Authors

DOI:

https://doi.org/10.14739/2409-2932.2024.1.289224

Keywords:

aging, natural selection, genomic instability, telomere wear

Abstract

Aging is a gradual and irreversible pathophysiological process. This is manifested in the decline of tissue and cell functions and a significant variety of pathologies associated with aging, including neurodegenerative diseases, cardiovascular diseases, metabolic disorders, increased risk of diseases of the skeleton and the immune system. Although modern medical advances have made a certain contribution to human health and greatly extended life expectancy, along with the aging of society, various chronic diseases are gradually emerging, which are the most important causes of disability and death of the elderly.

Aim. The aim of the work is to analyze modern ideas in the field of causes and manifestations of aging processes for the possibility of developing a strategy to slow down its course. At the molecular and cellular level, aging is a complex biological process involving the gradual deterioration of various cellular and molecular processes in the body over time.

The length of a person’s life is closely related to the decrease in the possibilities of repair and regeneration of tissues and organs. In response to stress at the molecular, cellular, and systemic levels, genetic, epigenetic, and environmental regulatory factors cause a decrease in the body’s physiological capabilities. They use complex molecular mechanisms that together contribute to aging. Molecular mechanisms (such as telomere shortening, accumulation of DNA damage, metabolic changes, and excessive free radical generation) strongly link various factors to the rate of aging. Collectively, these mechanisms inhibit cell proliferation, alter metabolism and gene expression, and induce high levels of free radicals, maintaining a senescent cell phenotype. Although the number of early senescent cells is low, they can limit the regenerative capacity of tissue stem cells and lead to the accumulation of cellular damage, thereby contributing to age-related diseases.

Conclusions. Current advances in high-throughput genomics, transcriptomics, proteomics, and metabolomics enable the characterization and quantification of thousands of epigenetic markers, transcripts, proteins, metabolites, and can reveal general changes that occur with age in complex organisms at the molecular level. Thus, the integration of these molecular markers and related molecular mechanisms into a comprehensive assessment of biological age to prevent age-related functional decline and morbidity is becoming an increasingly pressing issue in medical science and should be implemented in practice as soon as possible.

Author Biographies

Ugis Kletnieks, Latvian Longevity Association, Jurmala

Doctor of MBA, Member of the Board of the Smart Materials and Technologies Competence Center

Audrius Butkevichius, International Nobel Information Centre, Official Representation in Ukraine, Dnipro

MD, Moderator of Projects on Military Medicine

V. V. Hladyshev, Zaporizhzhia State Medical and Pharmaceutical University, Ukraine

PhD, DSc, Professor, Head of the Department of Medicines Technology

S. S. Sokolovskyi, National Technical University “Dnipro Polytechnic”, Ukraine

graduate student of the Department of State Administration and Local Self-Government

References

Bonkowski MS, Sinclair DA. Slowing ageing by design: the rise of NAD+ and sirtuin-activating compounds. Nat Rev Mol Cell Biol. 2016;17(11):679-90. doi: https://doi.org/10.1038/nrm.2016.93

López-Otín C, Kroemer G. Hallmarks of Health. Cell. 2021 Jan 7;184(1):33-63. doi: https://doi.org/10.1016/j.cell.2020.11.034. Epub 2020 Dec 18. Erratum in: Cell. 2021;184(7):1929-39.

López-Otín C, Pietrocola F, Roiz-Valle D, Galluzzi L, Kroemer G. Meta-hallmarks of aging and cancer. Cell Metab. 2023;35(1):12-35. doi: https://doi.org/10.1016/j.cmet.2022.11.001

López V, Fernández AF, Fraga MF. The role of 5-hydroxymethylcytosine in development, aging and age-related diseases. Ageing Res Rev. 2017;37:28-38. doi: https://doi.org/10.1016/j.arr.2017.05.002

Kennedy BK, Berger SL, Brunet A, Campisi J, Cuervo AM, Epel ES, et al. Geroscience: linking aging to chronic disease. Cell. 2014;159(4):709-13. doi: https://doi.org/10.1016/j.cell.2014.10.039

Schumacher B, Pothof J, Vijg J, Hoeijmakers JHJ. The central role of DNA damage in the ageing process. Nature. 2021;592(7856):695-703. doi: https://doi.org/10.1038/s41586-021-03307-7

Simon M, Yang J, Gigas J, Earley EJ, Hillpot E, Zhang L, et al. A rare human centenarian variant of SIRT6 enhances genome stability and interaction with Lamin A. EMBO J. 2022;41(21):e110393. doi: https://doi.org/10.15252/embj.2021110393

Vijg J, Dong X. Pathogenic Mechanisms of Somatic Mutation and Genome Mosaicism in Aging. Cell. 2020;182(1):12-23. doi: https://doi.org/10.1016/j.cell.2020.06.024

Williams AB, Schumacher B. p53 in the DNA-Damage-Repair Process. Cold Spring Harb Perspect Med. 2016;6(5):a026070. doi: https://doi.org/10.1101/cshperspect.a026070

Regulski MJ. Cellular Senescence: What, Why, and How. Wounds. 2017;29(6):168-74.

Martincorena I, Fowler JC, Wabik A, Lawson AR, Abascal F, Hall MW, et al. Somatic mutant clones colonize the human esophagus with age. Science. 2018;362(6417):911-7. doi: https://doi.org/10.1126/science.aau3879

Cagan A, Baez-Ortega A, Brzozowska N, Abascal F, Coorens TH, Sanders MA, et al. Somatic mutation rates scale with lifespan across mammals. Nature. 2022;604(7906):517-24. doi: https://doi.org/10.1038/s41586-022-04618-z

Hennekam RCM. Pathophysiology of premature aging characteristics in Mendelian progeroid disorders. Eur J Med Genet. 2020;63(11):104028. doi: https://doi.org/10.1016/j.ejmg.2020.104028

Son JM, Lee C. Mitochondria: multifaceted regulators of aging. BMB Rep. 2019;52(1):13-23. doi: https://doi.org/10.5483/BMBRep.2019.52.1.300

Chiang JL, Shukla P, Pagidas K, Ahmed NS, Karri S, Gunn DD, et al. Mitochondria in Ovarian Aging and Reproductive Longevity. Ageing Res Rev. 2020;63:101168. doi: https://doi.org/10.1016/j.arr.2020.101168

Quan Y, Xin Y, Tian G, Zhou J, Liu X. Mitochondrial ROS-Modulated mtDNA: A Potential Target for Cardiac Aging. Oxid Med Cell Longev. 2020;2020:9423593. doi: https://doi.org/10.1155/2020/9423593

Guo J, Huang X, Dou L, Yan M, Shen T, Tang W, Li J. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Ther. 2022;7(1):391. doi: https://doi.org/10.1038/s41392-022-01251-0

Fagundes N, Bisso-Machado R, Figueiredo P, Varal M, Zani A. What We Talk About When We Talk About "Junk DNA". Genome Biol Evol. 2022;14(5):evac055. doi: https://doi.org/10.1093/gbe/evac055

Xu T, Cheng D, Zhao Y, Zhang J, Zhu X, Zhang F, et al. Polymorphic tandem DNA repeats activate the human telomerase reverse transcriptase gene. Proc Natl Acad Sci U S A. 2021;118(26):e2019043118. doi: https://doi.org/10.1073/pnas.2019043118

DeBoy EA, Tassia MG, Schratz KE, Yan SM, Cosner ZL, McNally EJ, et al. Familial Clonal Hematopoiesis in a Long Telomere Syndrome. N Engl J Med. 2023;388(26):2422-33. doi: https://doi.org/10.1056/NEJMoa2300503

Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961;25:585-621. doi: 10.1016/0014-4827(61)90192-6

Shay JW. Role of Telomeres and Telomerase in Aging and Cancer. Cancer Discov. 2016;6(6):584-93. doi: https://doi.org/10.1158/2159-8290.CD-16-0062

Robinson NJ, Schiemann WP. Telomerase in Cancer: Function, Regulation, and Clinical Translation. Cancers (Basel). 2022;14(3):808. doi: https://doi.org/10.3390/cancers14030808

Solyom S, Kazazian HH Jr. Mobile elements in the human genome: implications for disease. Genome Med. 2012;4(2):12. doi: https://doi.org/10.1186/gm311

Macciardi F, Giulia Bacalini M, Miramontes R, Boattini A, Taccioli C, Modenini G, et al. A retrotransposon storm marks clinical phenoconversion to late-onset Alzheimer's disease. Geroscience. 2022;44(3):1525-50. doi: https://doi.org/10.1007/s11357-022-00580-w

Gorbunova V, Seluanov A, Mita P, McKerrow W, Fenyö D, Boeke JD, et al. The role of retrotransposable elements in ageing and age-associated diseases. Nature. 2021;596(7870):43-53. doi: https://doi.org/10.1038/s41586-021-03542-y

Tian X, Firsanov D, Zhang Z, Cheng Y, Luo L, Tombline G, et al. SIRT6 Is Responsible for More Efficient DNA Double-Strand Break Repair in Long-Lived Species. Cell. 2019;177(3):622-638.e22. doi: https://doi.org/10.1016/j.cell.2019.03.043

Della Valle F, Reddy P, Yamamoto M, Liu P, Saera-Vila A, Bensaddek D, et al. LINE-1 RNA causes heterochromatin erosion and is a target for amelioration of senescent phenotypes in progeroid syndromes. Sci Transl Med. 2022;14(657):eabl6057. doi: https://doi.org/10.1126/scitranslmed.abl6057

Roichman A, Elhanati S, Aon MA, Abramovich I, Di Francesco A, Shahar Y, et al. Restoration of energy homeostasis by SIRT6 extends healthy lifespan. Nat Commun. 2021;12(1):3208. doi: https://doi.org/10.1038/s41467-021-23545-7

Horvath S. DNA methylation age of human tissues and cell types. Genome Biol. 2013;14(10):R115. doi: https://doi.org/10.1186/gb-2013-14-10-r115

Sen P, Shah PP, Nativio R, Berger SL. Epigenetic Mechanisms of Longevity and Aging. Cell. 2016;166(4):822-39. doi: https://doi.org/10.1016/j.cell.2016.07.050

Levine ME, Lu AT, Quach A, Chen BH, Assimes TL, Bandinelli S, et al. An epigenetic biomarker of aging for lifespan and healthspan. Aging (Albany NY). 2018;10(4):573-91. doi: https://doi.org/10.18632/aging.101414

Loomba R, Gindin Y, Jiang Z, Lawitz E, Caldwell S, Djedjos CS, et al. DNA methylation signatures reflect aging in patients with nonalcoholic steatohepatitis. JCI Insight. 2018;3(2):e96685. doi: https://doi.org/10.1172/jci.insight.96685

Belsky DW, Caspi A, Arseneault L, Baccarelli A, Corcoran DL, Gao X, et al. Quantification of the pace of biological aging in humans through a blood test, the DunedinPoAm DNA methylation algorithm. Elife. 2020;9:e54870. doi: https://doi.org/10.7554/eLife.54870

Cutter AR, Hayes JJ. A brief review of nucleosome structure. FEBS Lett. 2015;589(20 Pt A):2914-22. doi: https://doi.org/10.1016/j.febslet.2015.05.016

Burd CJ, Archer TK. Chromatin architecture defines the glucocorticoid response. Mol Cell Endocrinol. 2013;380(1-2):25-31. doi: https://doi.org/10.1016/j.mce.2013.03.020

Gokey NG, Ward JM, Milliman EJ, Deterding LJ, Trotter KW, Archer TK. The loss of the H1.4 linker histone impacts nascent transcription and chromatin accessibility [Internet]. Biorxiv. 2023. Available from: http://dx.doi.org/10.1101/2023.05.14.540702

Blasco MA. The epigenetic regulation of mammalian telomeres. Nat Rev Genet. 2007;8(4):299-309. doi: https://doi.org/10.1038/nrg2047

Swer PB, Sharma R. ATP-dependent chromatin remodelers in ageing and age-related disorders. Biogerontology. 2021;22(1):1-17. doi: https://doi.org/10.1007/s10522-020-09899-3

Larson K, Yan SJ, Tsurumi A, Liu J, Zhou J, Gaur K, et al. Heterochromatin formation promotes longevity and represses ribosomal RNA synthesis. PLoS Genet. 2012;8(1):e1002473. doi: https://doi.org/10.1371/journal.pgen.1002473

Wu Q, Li B, Liu L, Sun S, Sun S. Centrosome dysfunction: a link between senescence and tumor immunity. Signal Transduct Target Ther. 2020;5(1):107. doi: https://doi.org/10.1038/s41392-020-00214-7

Jackson-Cook C. Constitutional and acquired autosomal aneuploidy. Clin Lab Med. 2011;31(4):481-511, vii. doi: https://doi.org/10.1016/j.cll.2011.08.002

Martin JM, Kellett JM, Kahn J. Aneuploidy in cultured human lymphocytes: I. Age and sex differences. Age Ageing. 1980;9(3):147-53. doi: https://doi.org/10.1093/ageing/9.3.147

Cáceres A, Jene A, Esko T, Pérez-Jurado LA, González JR. Extreme Downregulation of Chromosome Y and Cancer Risk in Men. J Natl Cancer Inst. 2020;112(9):913-20. doi: https://doi.org/10.1093/jnci/djz232

Abdel-Hafiz HA, Schafer JM, Chen X, Xiao T, Gauntner TD, Li Z, et al. Y chromosome loss in cancer drives growth by evasion of adaptive immunity. Nature. 2023 Jul;619(7970):624-31. doi: https://doi.org/10.1038/s41586-023-06234-x

Riaz M, Mattisson J, Polekhina G, Bakshi A, Halvardson J, Danielsson M, et al. A polygenic risk score predicts mosaic loss of chromosome Y in circulating blood cells. Cell Biosci. 2021;11(1):205. doi: https://doi.org/10.1186/s13578-021-00716-z

Sano S, Horitani K, Ogawa H, Halvardson J, Chavkin NW, Wang Y, et al. Hematopoietic loss of Y chromosome leads to cardiac fibrosis and heart failure mortality. Science. 2022;377(6603):292-7. doi: https://doi.org/10.1126/science.abn3100

Hernando-Herraez I, Evano B, Stubbs T, Commere PH, Jan Bonder M, Clark S, et al. Ageing affects DNA methylation drift and transcriptional cell-to-cell variability in mouse muscle stem cells. Nat Commun. 2019 Sep 25;10(1):4361. doi: https://doi.org/10.1038/s41467-019-12293-4

Bhadra M, Howell P, Dutta S, Heintz C, Mair WB. Alternative splicing in aging and longevity. Hum Genet. 2020;139(3):357-69. doi: https://doi.org/10.1007/s00439-019-02094-6

Ferioli M, Zauli G, Maiorano P, Milani D, Mirandola P, Neri LM. Role of physical exercise in the regulation of epigenetic mechanisms in inflammation, cancer, neurodegenerative diseases, and aging process. J Cell Physiol. 2019;234(9):14852-64. doi: https://doi.org/10.1002/jcp.28304

Tabula Muris Consortium. A single-cell transcriptomic atlas characterizes ageing tissues in the mouse. Nature. 2020;583(7817):590-5. doi: https://doi.org/10.1038/s41586-020-2496-1

Liu X, Huang J, Chen T, Wang Y, Xin S, Li J, et al. Yamanaka factors critically regulate the developmental signaling network in mouse embryonic stem cells. Cell Res. 2008;18(12):1177-89. doi: https://doi.org/10.1038/cr.2008.309

Lu Y, Brommer B, Tian X, Krishnan A, Meer M, Wang C, et al. Reprogramming to recover youthful epigenetic information and restore vision. Nature. 2020;588(7836):124-9. doi: https://doi.org/10.1038/s41586-020-2975-4

Kulak NA, Geyer PE, Mann M. Loss-less Nano-fractionator for High Sensitivity, High Coverage Proteomics. Mol Cell Proteomics. 2017;16(4):694-705. doi: https://doi.org/10.1074/mcp.O116.065136

Anfinsen CB. Principles that govern the folding of protein chains. Science. 1973;181(4096):223-30. doi: https://doi.org/10.1126/science.181.4096.223

Balchin D, Hayer-Hartl M, Hartl FU. In vivo aspects of protein folding and quality control. Science. 2016;353(6294):aac4354. doi: https://doi.org/10.1126/science.aac4354

Chiti F, Dobson CM. Protein Misfolding, Amyloid Formation, and Human Disease: A Summary of Progress Over the Last Decade. Annu Rev Biochem. 2017;86:27-68. doi: https://doi.org/10.1146/annurev-biochem-061516-045115

Sontag EM, Samant RS, Frydman J. Mechanisms and Functions of Spatial Protein Quality Control. Annu Rev Biochem. 2017;86:97-122. doi: https://doi.org/10.1146/annurev-biochem-060815-014616

Sabath N, Levy-Adam F, Younis A, Rozales K, Meller A, Hadar S, et al. Cellular proteostasis decline in human senescence. Proc Natl Acad Sci U S A. 2020;117(50):31902-13. doi: https://doi.org/10.1073/pnas.2018138117

Li Y, Xue Y, Xu X, Wang G, Liu Y, Wu H, et al. A mitochondrial FUNDC1/HSC70 interaction organizes the proteostatic stress response at the risk of cell morbidity. EMBO J. 2019;38(3):e98786. doi: https://doi.org/10.15252/embj.201798786

Koyuncu S, Loureiro R, Lee HJ, Wagle P, Krueger M, Vilchez D. Rewiring of the ubiquitinated proteome determines ageing in C. elegans. Nature. 2021;596(7871):285-90. doi: https://doi.org/10.1038/s41586-021-03781-z

Ciechanover A, Orian A, Schwartz AL. Ubiquitin-mediated proteolysis: biological regulation via destruction. Bioessays. 2000;22(5):442-51. doi: https://doi.org/10.1002/(SICI)1521-1878(200005)22:5<442::AID-BIES6>3.0.CO;2-Q

Dikic I, Wakatsuki S, Walters KJ. Ubiquitin-binding domains - from structures to functions. Nat Rev Mol Cell Biol. 2009;10(10):659-71. doi: https://doi.org/10.1038/nrm2767

Klaips CL, Jayaraj GG, Hartl FU. Pathways of cellular proteostasis in aging and disease. J Cell Biol. 2018;217(1):51-63. doi: https://doi.org/10.1083/jcb.201709072

Labbadia J, Morimoto RI. The biology of proteostasis in aging and disease. Annu Rev Biochem. 2015;84:435-64. doi: https://doi.org/10.1146/annurev-biochem-060614-033955

Montibeller L, de Belleroche J. Amyotrophic lateral sclerosis (ALS) and Alzheimer's disease (AD) are characterised by differential activation of ER stress pathways: focus on UPR target genes. Cell Stress Chaperones. 2018;23(5):897-912. doi: https://doi.org/10.1007/s12192-018-0897-y

Shcherbakov D, Nigri M, Akbergenov R, Brilkova M, Mantovani M, Petit PI, et al. Premature aging in mice with error-prone protein synthesis. Sci Adv. 2022;8(9):eabl9051. doi: https://doi.org/10.1126/sciadv.abl9051

Gerashchenko MV, Peterfi Z, Yim SH, Gladyshev VN. Translation elongation rate varies among organs and decreases with age. Nucleic Acids Res. 2021;49(2):e9. doi: https://doi.org/10.1093/nar/gkaa1103

Blagosklonny MV. Hallmarks of cancer and hallmarks of aging. Aging (Albany NY). 2022;14(9):4176-87. doi: https://doi.org/10.18632/aging.204082

Published

2024-02-23

How to Cite

1.
Kletnieks U, Butkevichius A, Hladyshev VV, Sokolovskyi SS. Why do we age? The main signs of aging. Current issues in pharmacy and medicine: science and practice [Internet]. 2024Feb.23 [cited 2024Jul.16];17(1):88-9. Available from: http://pharmed.zsmu.edu.ua/article/view/289224