Review Article | | Peer-Reviewed

DNA Damage, Transposable Element Expression and Their Associated Factors in Aging

Received: 3 October 2024     Accepted: 25 October 2024     Published: 29 November 2024
Views:       Downloads:
Abstract

Aging is a spontaneous and permanent physiological process that leads to declines in tissue and cell functions, along with an increased risk of developing various age-related diseases. The primary driving force associated with aging is the accumulation of damaged genetic material in the cell, such as DNA. DNA damage can be caused by endogenous and exogenous factors, which leads to genome instability, mitochondrial dysfunction, epigenetic modifications, and proteostatic disturb. Another driving force associated with aging is the disruption of cellular metabolism. This disruption is closely linked to alterations in the role of metabolic pathways, including insulin/IGF-1 and mTOR, which regulate crucial cellular processes like cell growth, cell proliferation, and apoptosis. The activation of the insulin/IGF-1 signaling pathway highly promotes cell growth and proliferation, while also inhibits autophagy and increasing ROS production. This ultimately leads to accelerated aging. Another crucial signaling pathway is the mTOR signaling pathway. It is responsible for detecting nutrient availability and controlling cell growth and metabolism. The dysregulation of mTOR function can lead to the development of neurodegenerative diseases, which are characterized by the aggregation of protein. Activation of transposable elements is the other driving force of aging, caused by changes in DNA methylation and the loss of heterochromatin. As a result, this leads to DNA damage, genomic instability, and inflammation. The aim of this review is to elucidate the consequence of DNA damage and other associated factors drive aging.

Published in Biochemistry and Molecular Biology (Volume 9, Issue 3)
DOI 10.11648/j.bmb.20240904.11
Page(s) 63-76
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2024. Published by Science Publishing Group

Keywords

Aging, Genome Instability, Molecular Damage, Transposon Element

References
[1] Ferrucci, L., et al., Measuring biological aging in humans: A quest. Aging cell, 2020. 19(2): p. e13080.
[2] Ogrodnik, M., H. Salmonowicz, and V. N. Gladyshev, Integrating cellular senescence with the concept of damage accumulation in aging: Relevance for clearance of senescent cells. Aging cell, 2019. 18(1): p. e12841.
[3] Singh, P. P., et al., The genetics of aging: a vertebrate perspective. Cell, 2019. 177(1): p. 200-220.
[4] Li, Z., et al., Folic acid inhibits aging-induced telomere attrition and apoptosis in astrocytes in vivo and in vitro. Cerebral Cortex, 2022. 32(2): p. 286-297.
[5] Bharath, L. P., et al., Metformin enhances autophagy and normalizes mitochondrial function to alleviate aging-associated inflammation. Cell metabolism, 2020. 32(1): p. 44-55. e6.
[6] Tracy, T. E., et al., Tau interactome maps synaptic and mitochondrial processes associated with neurodegeneration. Cell, 2022. 185(4): p. 712-728. e14.
[7] Lei, J., et al., Exosomes from antler stem cells alleviate mesenchymal stem cell senescence and osteoarthritis. Protein & cell, 2022. 13(3): p. 220-226.
[8] Van der Laan, L., et al., Epigenetic aging biomarkers and occupational exposure to benzene, trichloroethylene and formaldehyde. Environment international, 2022. 158: p. 106871.
[9] Kubben, N. and T. Misteli, Shared molecular and cellular mechanisms of premature ageing and ageing-associated diseases. Nature Reviews Molecular Cell Biology, 2017. 18(10): p. 595-609.
[10] Herrmann, M., et al., Telomere biology and age-related diseases. Clinical Chemistry and Laboratory Medicine (CCLM), 2018. 56(8): p. 1210-1222.
[11] Ovadya, Y. and V. Krizhanovsky, Senescent cells: SASPected drivers of age-related pathologies. Biogerontology, 2014. 15: p. 627-642.
[12] Zhu, Y., et al., Telomere and its role in the aging pathways: telomere shortening, cell senescence and mitochondria dysfunction. Biogerontology, 2019. 20: p. 1-16.
[13] Hood, D. A., et al., Maintenance of skeletal muscle mitochondria in health, exercise, and aging. Annual Review of Physiology, 2019. 81: p. 19-41.
[14] Sun, N., R. J. Youle, and T. Finkel, The mitochondrial basis of aging. Molecular cell, 2016. 61(5): p. 654-666.
[15] Mossad, O., et al., Gut microbiota drives age-related oxidative stress and mitochondrial damage in microglia via the metabolite N 6-carboxymethyllysine. Nature neuroscience, 2022. 25(3): p. 295-305.
[16] Yang, W. and S. Hekimi, A mitochondrial superoxide signal triggers increased longevity in Caenorhabditis elegans. PLoS biology, 2010. 8(12): p. e1000556.
[17] Lee, S.-J., A. B. Hwang, and C. Kenyon, Inhibition of respiration extends C. elegans life span via reactive oxygen species that increase HIF-1 activity. Current Biology, 2010. 20(23): p. 2131-2136.
[18] Schriner, S. E., et al., Extension of murine life span by overexpression of catalase targeted to mitochondria. science, 2005. 308(5730): p. 1909-1911.
[19] López-Otín, C., et al., The hallmarks of aging. Cell, 2013. 153(6): p. 1194-1217.
[20] Hernandez-Segura, A., J. Nehme, and M. Demaria, Hallmarks of cellular senescence. Trends in cell biology, 2018. 28(6): p. 436-453.
[21] Baker, D. J., et al., Naturally occurring p16Ink4a-positive cells shorten healthy lifespan. Nature, 2016. 530(7589): p. 184-189.
[22] De Luca, C., et al., Enhanced expression of LINE-1-encoded ORF2 protein in early stages of colon and prostate transformation. Oncotarget, 2016. 7(4): p. 4048.
[23] Di Ruocco, F., et al., Alu RNA accumulation induces epithelial-to-mesenchymal transition by modulating miR-566 and is associated with cancer progression. Oncogene, 2018. 37(5): p. 627-637.
[24] Rodić, N., et al., Long interspersed element-1 protein expression is a hallmark of many human cancers. The American journal of pathology, 2014. 184(5): p. 1280-1286.
[25] Sun, W., et al., Pathogenic tau-induced piRNA depletion promotes neuronal death through transposable element dysregulation in neurodegenerative tauopathies. Nature neuroscience, 2018. 21(8): p. 1038-1048.
[26] Gao, Y., J. Zhang, and F. Zhao, Circular RNA identification based on multiple seed matching. Briefings in bioinformatics, 2018. 19(5): p. 803-810.
[27] Burns, K. H. and J. D. Boeke, Human transposon tectonics. Cell, 2012. 149(4): p. 740-752.
[28] Curtin, N. J., DNA repair dysregulation from cancer driver to therapeutic target. Nature Reviews Cancer, 2012. 12(12): p. 801-817.
[29] Swenberg, J. A., et al., Endogenous versus exogenous DNA adducts: their role in carcinogenesis, epidemiology, and risk assessment. Toxicological sciences, 2011. 120(suppl_1): p. S130-S145.
[30] Maynard, S., et al., Base excision repair of oxidative DNA damage and association with cancer and aging. Carcinogenesis, 2009. 30(1): p. 2-10.
[31] Vijg, J., From DNA damage to mutations: All roads lead to aging. Ageing Research Reviews, 2021. 68: p. 101316.
[32] Sirbu, B. M. and D. Cortez, DNA damage response: three levels of DNA repair regulation. Cold Spring Harbor perspectives in biology, 2013. 5(8): p. a012724.
[33] Kennedy, B. K., et al., Geroscience: linking aging to chronic disease. Cell, 2014. 159(4): p. 709-713.
[34] Schumacher, B., et al., The central role of DNA damage in the ageing process. Nature, 2021. 592(7856): p. 695-703.
[35] Fagagna, F. d. A. d., et al., A DNA damage checkpoint response in telomere-initiated senescence. Nature, 2003. 426(6963): p. 194-198.
[36] Vermeij, W., et al., Restricted diet delays accelerated ageing and genomic stress in DNA-repair-deficient mice. Nature, 2016. 537(7620): p. 427-431.
[37] Vijg, J., Somatic mutations, genome mosaicism, cancer and aging. Current opinion in genetics & development, 2014. 26: p. 141-149.
[38] Fitsiou, E., et al., Cellular senescence and the senescence-associated secretory phenotype as drivers of skin photoaging. Journal of Investigative Dermatology, 2021. 141(4): p. 1119-1126.
[39] Thompson, L. H., Recognition, signaling, and repair of DNA double-strand breaks produced by ionizing radiation in mammalian cells: the molecular choreography. Mutation Research/Reviews in Mutation Research, 2012. 751(2): p. 158-246.
[40] Yousefzadeh, M., et al., DNA damage—how and why we age? Elife, 2021. 10: p. e62852.
[41] Kaiser, A. M. and L. D. Attardi, Deconstructing networks of p53-mediated tumor suppression in vivo. Cell Death & Differentiation, 2018. 25(1): p. 93-103.
[42] Tateossian, H., et al., Interactions between the otitis media gene, Fbxo11, and p53 in the mouse embryonic lung. Disease models & mechanisms, 2015. 8(12): p. 1531-1542.
[43] Rinon, A., et al., p53 coordinates cranial neural crest cell growth and epithelial-mesenchymal transition/delamination processes. Development, 2011. 138(9): p. 1827-1838.
[44] Menck, C. F. and V. Munford, DNA repair diseases: What do they tell us about cancer and aging? Genetics and Molecular Biology, 2014. 37: p. 220-233.
[45] Wood, R. D., Fifty years since DNA repair was linked to cancer. 2018, Nature Publishing Group UK London.
[46] Arancio, W., et al., Epigenetic involvement in Hutchinson-Gilford progeria syndrome: a mini-review. Gerontology, 2014. 60(3): p. 197-203.
[47] Burla, R., et al., Genomic instability and DNA replication defects in progeroid syndromes. Nucleus, 2018. 9(1): p. 368-379.
[48] Burtner, C. R. and B. K. Kennedy, Progeria syndromes and ageing: what is the connection? Nature reviews Molecular cell biology, 2010. 11(8): p. 567-578.
[49] Loi, M., et al., Barrier-to-autointegration factor (BAF) involvement in prelamin A-related chromatin organization changes. Oncotarget, 2016. 7(13): p. 15662.
[50] Valentin-Vega, Y. A., et al., Mitochondrial dysfunction in ataxia-telangiectasia. Blood, The Journal of the American Society of Hematology, 2012. 119(6): p. 1490-1500.
[51] Rothblum-Oviatt, C., et al., Ataxia telangiectasia: a review. Orphanet journal of rare diseases, 2016. 11: p. 1-21.
[52] Kudlow, B. A., B. K. Kennedy, and R. J. Monnat Jr, Werner and Hutchinson–Gilford progeria syndromes: mechanistic basis of human progeroid diseases. Nature reviews Molecular cell biology, 2007. 8(5): p. 394-404.
[53] Sugimoto, M., A cascade leading to premature aging phenotypes including abnormal tumor profiles in Werner syndrome. International journal of molecular medicine, 2014. 33(2): p. 247-253.
[54] Nguyen, G. H., et al., Regulation of gene expression by the BLM helicase correlates with the presence of G-quadruplex DNA motifs. Proceedings of the National Academy of Sciences, 2014. 111(27): p. 9905-9910.
[55] De Renty, C. and N. A. Ellis, Bloom’s syndrome: Why not premature aging?: A comparison of the BLM and WRN helicases. Ageing research reviews, 2017. 33: p. 36-51.
[56] Maciejowski, J. and T. de Lange, Telomeres in cancer: tumour suppression and genome instability. Nature reviews Molecular cell biology, 2017. 18(3): p. 175-186.
[57] de Lange, T., Shelterin-mediated telomere protection. Annual review of genetics, 2018. 52: p. 223-247.
[58] Ishikawa, F., Portrait of replication stress viewed from telomeres. Cancer science, 2013. 104(7): p. 790-794.
[59] Günes, C. and K. L. Rudolph, The role of telomeres in stem cells and cancer. Cell, 2013. 152(3): p. 390-393.
[60] Wang, J. Y., et al., Shorter telomere length increases age‐related tumor risks in von Hippel‐Lindau disease patients. Cancer Medicine, 2017. 6(9): p. 2131-2141.
[61] Fumagalli, M., et al., Telomeric DNA damage is irreparable and causes persistent DNA-damage-response activation. Nature cell biology, 2012. 14(4): p. 355-365.
[62] Monaghan, P. and S. E. Ozanne, Somatic growth and telomere dynamics in vertebrates: relationships, mechanisms and consequences. Philosophical Transactions of the Royal Society B: Biological Sciences, 2018. 373(1741): p. 20160446.
[63] Halliwell, B. and J. M. Gutteridge, Free radicals in biology and medicine. 2015: Oxford university press, USA.
[64] Von Zglinicki, T., Oxidative stress shortens telomeres. Trends in biochemical sciences, 2002. 27(7): p. 339-344.
[65] Haussmann, M. F. and B. J. Heidinger, Telomere dynamics may link stress exposure and ageing across generations. Biology letters, 2015. 11(11): p. 20150396.
[66] Angelier, F., et al., Do glucocorticoids mediate the link between environmental conditions and telomere dynamics in wild vertebrates? A review. General and comparative endocrinology, 2018. 256: p. 99-111.
[67] Costantini, D., V. Marasco, and A. P. Møller, A meta-analysis of glucocorticoids as modulators of oxidative stress in vertebrates. Journal of Comparative Physiology B, 2011. 181: p. 447-456.
[68] Choi, J., S. R. Fauce, and R. B. Effros, Reduced telomerase activity in human T lymphocytes exposed to cortisol. Brain, behavior, and immunity, 2008. 22(4): p. 600-605.
[69] Fang, E. F., et al., Defective mitophagy in XPA via PARP-1 hyperactivation and NAD+/SIRT1 reduction. Cell, 2014. 157(4): p. 882-896.
[70] Gibson, B. A. and W. L. Kraus, New insights into the molecular and cellular functions of poly (ADP-ribose) and PARPs. Nature reviews Molecular cell biology, 2012. 13(7): p. 411-424.
[71] Gonzales-Ebsen, A. C., N. Gregersen, and R. K. Olsen, Linking telomere loss and mitochondrial dysfunction in chronic disease. Front Biosci (Landmark Ed), 2017. 22: p. 117-127.
[72] Cadet, J. and J. R. Wagner, DNA base damage by reactive oxygen species, oxidizing agents, and UV radiation. Cold Spring Harbor perspectives in biology, 2013. 5(2): p. a012559.
[73] Pfeiffer, V. and J. Lingner, TERRA promotes telomere shortening through exonuclease 1–mediated resection of chromosome ends. PLoS genetics, 2012. 8(6): p. e1002747.
[74] Arnoult, N., A. Van Beneden, and A. Decottignies, Telomere length regulates TERRA levels through increased trimethylation of telomeric H3K9 and HP1α. Nature structural & molecular biology, 2012. 19(9): p. 948-956.
[75] Sen, P., et al., Epigenetic mechanisms of longevity and aging. Cell, 2016. 166(4): p. 822-839.
[76] Nikolac Perkovic, M., et al., Epigenetics of Alzheimer’s disease. Biomolecules, 2021. 11(2): p. 195.
[77] Moore, L. D., T. Le, and G. Fan, DNA methylation and its basic function. Neuropsychopharmacology, 2013. 38(1): p. 23-38.
[78] Hervouet, E., et al., Specific or not specific recruitment of DNMTs for DNA methylation, an epigenetic dilemma. Clinical epigenetics, 2018. 10: p. 1-18.
[79] Jeltsch, A., et al., Mechanism and biological role of Dnmt2 in nucleic acid methylation. RNA biology, 2017. 14(9): p. 1108-1123.
[80] Zarakowska, E., et al., Oxidation products of 5-methylcytosine are decreased in senescent cells and tissues of progeroid mice. The Journals of Gerontology: Series A, 2018. 73(8): p. 1003-1009.
[81] Cagan, A., et al., Somatic mutation rates scale with lifespan across mammals. Nature, 2022. 604(7906): p. 517-524.
[82] Stratton, M. R., P. J. Campbell, and P. A. Futreal, The cancer genome. Nature, 2009. 458(7239): p. 719-724.
[83] Horvath, S., DNA methylation age of human tissues and cell types. Genome biology, 2013. 14: p. 1-20.
[84] Vijg, J. and X. Dong, Pathogenic mechanisms of somatic mutation and genome mosaicism in aging. Cell, 2020. 182(1): p. 12-23.
[85] Paluvai, H., E. Di Giorgio, and C. Brancolini, The histone code of senescence. Cells, 2020. 9(2): p. 466.
[86] Yi, S.-J. and K. Kim, New insights into the role of histone changes in aging. International journal of molecular sciences, 2020. 21(21): p. 8241.
[87] Wang, Y., Q. Yuan, and L. Xie, Histone modifications in aging: the underlying mechanisms and implications. Current stem cell research & therapy, 2018. 13(2): p. 125-135.
[88] Kirfel, P., A. Vilcinskas, and M. Skaljac, Lysine acetyltransferase p300/cbp plays an important role in reproduction, embryogenesis and longevity of the pea aphid Acyrthosiphon pisum. Insects, 2020. 11(5): p. 265.
[89] Sen, P., et al., H3K36 methylation promotes longevity by enhancing transcriptional fidelity. Genes & development, 2015. 29(13): p. 1362-1376.
[90] Jeon, H.-J., et al., Effect of heterochromatin stability on intestinal stem cell aging in Drosophila. Mechanisms of Ageing and Development, 2018. 173: p. 50-60.
[91] Li, C.-L., et al., Region-specific H3K9me3 gain in aged somatic tissues in Caenorhabditis elegans. PLoS Genetics, 2021. 17(9): p. e1009432.
[92] Wood, J. G., et al., Chromatin remodeling in the aging genome of Drosophila. Aging cell, 2010. 9(6): p. 971-978.
[93] Zhang, W., et al., A Werner syndrome stem cell model unveils heterochromatin alterations as a driver of human aging. Science, 2015. 348(6239): p. 1160-1163.
[94] Wu, Z., et al., Differential stem cell aging kinetics in Hutchinson-Gilford progeria syndrome and Werner syndrome. Protein & cell, 2018. 9(4): p. 333-350.
[95] Liu, Z., et al., Large-scale chromatin reorganization reactivates placenta-specific genes that drive cellular aging. Developmental cell, 2022. 57(11): p. 1347-1368. e12.
[96] Liu, X., et al., Mechanism of chromatin remodelling revealed by the Snf2-nucleosome structure. Nature, 2017. 544(7651): p. 440-445.
[97] Zhu, D., et al., NuRD mediates mitochondrial stress–induced longevity via chromatin remodeling in response to acetyl-CoA level. Science advances, 2020. 6(31): p. eabb2529.
[98] Kauppila, T. E., J. H. Kauppila, and N.-G. Larsson, Mammalian mitochondria and aging: an update. Cell metabolism, 2017. 25(1): p. 57-71.
[99] Nacarelli, T., et al., NAD+ metabolism governs the proinflammatory senescence-associated secretome. Nature cell biology, 2019. 21(3): p. 397-407.
[100] Bakula, D. and M. Scheibye-Knudsen, MitophAging: mitophagy in aging and disease. Frontiers in cell and developmental biology, 2020. 8: p. 239.
[101] Zhunina, O. A., et al., The role of mitochondrial dysfunction in vascular disease, tumorigenesis, and diabetes. Frontiers in Molecular Biosciences, 2021. 8: p. 671908.
[102] Bagkos, G., K. Koufopoulos, and C. Piperi, A new model for mitochondrial membrane potential production and storage. Medical hypotheses, 2014. 83(2): p. 175-181.
[103] Passos, J. F., et al., Feedback between p21 and reactive oxygen production is necessary for cell senescence. Molecular systems biology, 2010. 6(1): p. 347.
[104] Nelson, G., et al., The senescent bystander effect is caused by ROS-activated NF-κB signalling. Mechanisms of ageing and development, 2018. 170: p. 30-36.
[105] Trifunovic, A., et al., Somatic mtDNA mutations cause aging phenotypes without affecting reactive oxygen species production. Proceedings of the National Academy of Sciences, 2005. 102(50): p. 17993-17998.
[106] Ryzhkova, A. I., et al., Mitochondrial diseases caused by mtDNA mutations: a mini-review. Therapeutics and clinical risk management, 2018: p. 1933-1942.
[107] Vermulst, M., et al., DNA deletions and clonal mutations drive premature aging in mitochondrial mutator mice. Nature genetics, 2008. 40(4): p. 392-394.
[108] Kolesar, J. E., et al., Defects in mitochondrial DNA replication and oxidative damage in muscle of mtDNA mutator mice. Free Radical Biology and Medicine, 2014. 75: p. 241-251.
[109] Marchi, S., et al., Mitochondrial control of inflammation. Nature Reviews Immunology, 2023. 23(3): p. 159-173.
[110] de Oliveira, L. G., et al., Unraveling the link between mitochondrial dynamics and neuroinflammation. Frontiers in Immunology, 2021. 12: p. 624919.
[111] Bartolomé, A., et al., MTORC1 regulates both general autophagy and mitophagy induction after oxidative phosphorylation uncoupling. Molecular and cellular biology, 2017. 37(23): p. e00441-17.
[112] Dikic, I. and Z. Elazar, Mechanism and medical implications of mammalian autophagy. Nature reviews Molecular cell biology, 2018. 19(6): p. 349-364.
[113] Lee, J., S. Giordano, and J. Zhang, Autophagy, mitochondria and oxidative stress: cross-talk and redox signalling. Biochemical Journal, 2012. 441(2): p. 523-540.
[114] Li, L., Y. Chen, and S. B. Gibson, Starvation-induced autophagy is regulated by mitochondrial reactive oxygen species leading to AMPK activation. Cellular signalling, 2013. 25(1): p. 50-65.
[115] Rabinovitch, R. C., et al., AMPK maintains cellular metabolic homeostasis through regulation of mitochondrial reactive oxygen species. Cell reports, 2017. 21(1): p. 1-9.
[116] Aman, Y., et al., Autophagy in healthy aging and disease. Nature aging, 2021. 1(8): p. 634-650.
[117] Leidal, A. M., B. Levine, and J. Debnath, Autophagy and the cell biology of age-related disease. Nature cell biology, 2018. 20(12): p. 1338-1348.
[118] Hansen, M., D. C. Rubinsztein, and D. W. Walker, Autophagy as a promoter of longevity: insights from model organisms. Nature reviews Molecular cell biology, 2018. 19(9): p. 579-593.
[119] Hewitt, G. and V. I. Korolchuk, Repair, reuse, recycle: the expanding role of autophagy in genome maintenance. Trends in cell biology, 2017. 27(5): p. 340-351.
[120] Wang, Y., et al., Autophagy regulates chromatin ubiquitination in DNA damage response through elimination of SQSTM1/p62. Molecular cell, 2016. 63(1): p. 34-48.
[121] Mathew, R., et al., Autophagy suppresses tumorigenesis through elimination of p62. Cell, 2009. 137(6): p. 1062-1075.
[122] Hewitt, G., et al., SQSTM1/p62 mediates crosstalk between autophagy and the UPS in DNA repair. Autophagy, 2016. 12(10): p. 1917-1930.
[123] Gillespie, D. A. and K. M. Ryan, Autophagy is critically required for DNA repair by homologous recombination. Molecular & Cellular Oncology, 2016. 3(1): p. e1030538.
[124] Qiang, L., et al., Autophagy positively regulates DNA damage recognition by nucleotide excision repair. Autophagy, 2016. 12(2): p. 357-368.
[125] Mathew, R., et al., Autophagy suppresses tumor progression by limiting chromosomal instability. Genes & development, 2007. 21(11): p. 1367-1381.
[126] Morsli, S., G. J. Doherty, and D. Muñoz-Espín, Activatable senoprobes and senolytics: Novel strategies to detect and target senescent cells. Mechanisms of Ageing and Development, 2022. 202: p. 111618.
[127] Ohtani, N., The roles and mechanisms of senescence-associated secretory phenotype (SASP): can it be controlled by senolysis? Inflammation and regeneration, 2022. 42(1): p. 11.
[128] Zhou, D., M. Borsa, and A. K. Simon, Hallmarks and detection techniques of cellular senescence and cellular ageing in immune cells. Aging Cell, 2021. 20(2): p. e13316.
[129] Mittelbrunn, M. and G. Kroemer, Hallmarks of T cell aging. Nature immunology, 2021. 22(6): p. 687-698.
[130] Desdín-Micó, G., et al., T cells with dysfunctional mitochondria induce multimorbidity and premature senescence. Science, 2020. 368(6497): p. 1371-1376.
[131] Henriques, C. M. and M. G. Ferreira, Consequences of telomere shortening during lifespan. Current opinion in cell biology, 2012. 24(6): p. 804-808.
[132] Sun, Y., J.-P. Coppé, and E. W.-F. Lam, Cellular senescence: the sought or the unwanted? Trends in Molecular Medicine, 2018. 24(10): p. 871-885.
[133] Indo, H. P., et al., A mitochondrial superoxide theory for oxidative stress diseases and aging. Journal of clinical biochemistry and nutrition, 2015. 56(1): p. 1-7.
[134] Balaban, R. S., S. Nemoto, and T. Finkel, Mitochondria, oxidants, and aging. cell, 2005. 120(4): p. 483-495.
[135] Campisi, J., Aging, cellular senescence, and cancer. Annual review of physiology, 2013. 75: p. 685-705.
[136] Abbas, M., et al., Endothelial microparticles from acute coronary syndrome patients induce premature coronary artery endothelial cell aging and thrombogenicity: role of the Ang II/AT1 receptor/NADPH oxidase-mediated activation of MAPKs and PI3-kinase pathways. Circulation, 2017. 135(3): p. 280-296.
[137] Muñoz-Espín, D. and M. Serrano, Cellular senescence: from physiology to pathology. Nature reviews Molecular cell biology, 2014. 15(7): p. 482-496.
[138] Lanigan, F., J. Geraghty, and A. Bracken, Transcriptional regulation of cellular senescence. Oncogene, 2011. 30(26): p. 2901-2911.
[139] Di Micco, R., et al., Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nature reviews Molecular cell biology, 2021. 22(2): p. 75-95.
[140] Fumagalli, M., et al., Stable cellular senescence is associated with persistent DDR activation. PloS one, 2014. 9(10): p. e110969.
[141] Bernadotte, A., V. M. Mikhelson, and I. M. Spivak, Markers of cellular senescence. Telomere shortening as a marker of cellular senescence. Aging (Albany NY), 2016. 8(1): p. 3.
[142] Yue, L. and H. Yao, Mitochondrial dysfunction in inflammatory responses and cellular senescence: pathogenesis and pharmacological targets for chronic lung diseases. British journal of pharmacology, 2016. 173(15): p. 2305-2318.
[143] Wiley, C. D., et al., Mitochondrial dysfunction induces senescence with a distinct secretory phenotype. Cell metabolism, 2016. 23(2): p. 303-314.
[144] Gewirtz, D. A., Autophagy and senescence: a partnership in search of definition. Autophagy, 2013. 9(5): p. 808-812.
[145] He, Q., et al., Chromosomal instability-induced senescence potentiates cell non-autonomous tumourigenic effects. Oncogenesis, 2018. 7(8): p. 62.
[146] Storer, M., et al., Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell, 2013. 155(5): p. 1119-1130.
[147] Demaria, M., et al., An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Developmental cell, 2014. 31(6): p. 722-733.
[148] Helman, A., et al., p16Ink4a-induced senescence of pancreatic beta cells enhances insulin secretion. Nature medicine, 2016. 22(4): p. 412-420.
[149] Liu, Y., et al., Expression of p16INK4a in peripheral blood T‐cells is a biomarker of human aging. Aging cell, 2009. 8(4): p. 439-448.
[150] Roos, C. M., et al., Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging cell, 2016. 15(5): p. 973-977.
[151] Chandra, A., et al., Targeted reduction of senescent cell burden alleviates focal radiotherapy‐related bone loss. Journal of Bone and Mineral Research, 2020. 35(6): p. 1119-1131.
[152] Ogrodnik, M., et al., Cellular senescence drives age-dependent hepatic steatosis. Nature communications, 2017. 8(1): p. 15691.
[153] Alimirah, F., et al., Cellular senescence promotes skin carcinogenesis through p38MAPK and p44/42MAPK signaling. Cancer research, 2020. 80(17): p. 3606-3619.
[154] Chinta, S. J., et al., Cellular senescence is induced by the environmental neurotoxin paraquat and contributes to neuropathology linked to Parkinson’s disease. Cell reports, 2018. 22(4): p. 930-940.
[155] Zhang, P., et al., Senolytic therapy alleviates Aβ-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer’s disease model. Nature neuroscience, 2019. 22(5): p. 719-728.
[156] Kirkland, J. and T. Tchkonia, Senolytic drugs: from discovery to translation. Journal of internal medicine, 2020. 288(5): p. 518-536.
[157] Johnson, S. C., Nutrient sensing, signaling and ageing: the role of IGF-1 and mTOR in ageing and age-related disease. Biochemistry and Cell Biology of Ageing: Part I Biomedical Science, 2018: p. 49-97.
[158] Templeman, N. M. and C. T. Murphy, Regulation of reproduction and longevity by nutrient-sensing pathways. Journal of Cell Biology, 2018. 217(1): p. 93-106.
[159] Johnson, S. C., P. S. Rabinovitch, and M. Kaeberlein, mTOR is a key modulator of ageing and age-related disease. Nature, 2013. 493(7432): p. 338-345.
[160] Bartke, A., L. Y. Sun, and V. Longo, Somatotropic signaling: trade-offs between growth, reproductive development, and longevity. Physiological reviews, 2013. 93(2): p. 571-598.
[161] Ock, S., et al., Deletion of IGF-1 receptors in cardiomyocytes attenuates cardiac aging in male mice. Endocrinology, 2016. 157(1): p. 336-345.
[162] Laplante, M. and D. M. Sabatini, mTOR signaling in growth control and disease. cell, 2012. 149(2): p. 274-293.
[163] González, A. and M. N. Hall, Nutrient sensing and TOR signaling in yeast and mammals. The EMBO journal, 2017. 36(4): p. 397-408.
[164] Kirtonia, A., et al., Overexpression of laminin-5 gamma-2 promotes tumorigenesis of pancreatic ductal adenocarcinoma through EGFR/ERK1/2/AKT/mTOR cascade. Cellular and Molecular Life Sciences, 2022. 79(7): p. 362.
[165] He, Y., et al., Targeting signaling pathways in prostate cancer: Mechanisms and clinical trials. Signal Transduction and Targeted Therapy, 2022. 7(1): p. 198.
[166] Carroll, B., et al., Persistent mTORC1 signaling in cell senescence results from defects in amino acid and growth factor sensing. Journal of Cell Biology, 2017. 216(7): p. 1949-1957.
[167] Nacarelli, T., A. Azar, and C. Sell, Mitochondrial stress induces cellular senescence in an mTORC1-dependent manner. Free Radical Biology and Medicine, 2016. 95: p. 133-154.
[168] Perluigi, M., F. Di Domenico, and D. A. Butterfield, mTOR signaling in aging and neurodegeneration: at the crossroad between metabolism dysfunction and impairment of autophagy. Neurobiology of disease, 2015. 84: p. 39-49.
[169] Lin, S.-C. and D. G. Hardie, AMPK: sensing glucose as well as cellular energy status. Cell metabolism, 2018. 27(2): p. 299-313.
[170] Salminen, A. and K. Kaarniranta, AMP-activated protein kinase (AMPK) controls the aging process via an integrated signaling network. Ageing research reviews, 2012. 11(2): p. 230-241.
[171] Toyama, E. Q., et al., AMP-activated protein kinase mediates mitochondrial fission in response to energy stress. Science, 2016. 351(6270): p. 275-281.
[172] Moqrich, A., Peripheral pain-sensing neurons: from molecular diversity to functional specialization. Cell Reports, 2014. 6(2): p. 245-246.
[173] Ning, Y.-C., et al., Short-term calorie restriction protects against renal senescence of aged rats by increasing autophagic activity and reducing oxidative damage. Mechanisms of ageing and development, 2013. 134(11-12): p. 570-579.
[174] Ma, Y., et al., Autophagy controls mesenchymal stem cell properties and senescence during bone aging. Aging cell, 2018. 17(1): p. e12709.
[175] García-Prat, L., et al., Autophagy maintains stemness by preventing senescence. Nature, 2016. 529(7584): p. 37-42.
[176] Garcia, D. and R. J. Shaw, AMPK: mechanisms of cellular energy sensing and restoration of metabolic balance. Molecular cell, 2017. 66(6): p. 789-800.
[177] Brunet, A., et al., Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. science, 2004. 303(5666): p. 2011-2015.
[178] Liu, Y., et al., New insights for cellular and molecular mechanisms of aging and aging-related diseases: herbal medicine as potential therapeutic approach. Oxidative Medicine and Cellular Longevity, 2019. 2019.
[179] Zhao, L., et al., Sirtuins and their biological relevance in aging and age-related diseases. Aging and disease, 2020. 11(4): p. 927.
[180] Kapahi, P., M. Kaeberlein, and M. Hansen, Dietary restriction and lifespan: Lessons from invertebrate models. Ageing research reviews, 2017. 39: p. 3-14.
[181] Kanfi, Y., et al., The sirtuin SIRT6 regulates lifespan in male mice. Nature, 2012. 483(7388): p. 218-221.
[182] Wątroba, M. and D. Szukiewicz, The role of sirtuins in aging and age-related diseases. Advances in medical sciences, 2016. 61(1): p. 52-62.
[183] Fernandes, S. A. and C. Demetriades, The multifaceted role of nutrient sensing and mTORC1 signaling in physiology and aging. Frontiers in Aging, 2021. 2: p. 707372.
[184] Jönsson, M. E., et al., Transposable elements: a common feature of neurodevelopmental and neurodegenerative disorders. Trends in Genetics, 2020. 36(8): p. 610-623.
[185] Wicker, T., et al., Impact of transposable elements on genome structure and evolution in bread wheat. Genome biology, 2018. 19: p. 1-18.
[186] Sedivy, J. M., et al., Death by transposition–the enemy within? Bioessays, 2013. 35(12): p. 1035-1043.
[187] Liu, C.-H., et al., Contribution of human retroviruses to disease development—a focus on the HIV–and HERV–cancer relationships and treatment strategies. Viruses, 2020. 12(8): p. 852.
[188] de Cubas, A. A., et al., DNA hypomethylation promotes transposable element expression and activation of immune signaling in renal cell cancer. JCI insight, 2020. 5(11).
[189] Römer, C., Viruses and endogenous retroviruses as roots for neuroinflammation and neurodegenerative diseases. Frontiers in Neuroscience, 2021. 15: p. 648629.
[190] Ochirov, C., The involvement of human endogenous retroviruses K (HERV-K) in aging processes via induction of inflammation. 2019, PeerJ Preprints.
[191] Bogu, G. K., et al., Atlas of transcriptionally active transposable elements in human adult tissues. BioRxiv, 2019: p. 714212.
[192] Volkman, H. E. and D. B. Stetson, The enemy within: endogenous retroelements and autoimmune disease. Nature immunology, 2014. 15(5): p. 415-422.
[193] Elsner, D., K. Meusemann, and J. Korb, Longevity and transposon defense, the case of termite reproductives. Proceedings of the National Academy of Sciences, 2018. 115(21): p. 5504-5509.
[194] Wahl, D., et al., Healthy aging interventions reduce repetitive element transcripts. The Journals of Gerontology: Series A, 2021. 76(5): p. 805-810.
[195] Gorbunova, V., et al., The role of retrotransposable elements in ageing and age-associated diseases. Nature, 2021. 596(7870): p. 43-53.
Cite This Article
  • APA Style

    Yeshanew, T. M., begashew, B. A., Birhane, N., Getie, B. (2024). DNA Damage, Transposable Element Expression and Their Associated Factors in Aging. Biochemistry and Molecular Biology, 9(3), 63-76. https://doi.org/10.11648/j.bmb.20240904.11

    Copy | Download

    ACS Style

    Yeshanew, T. M.; begashew, B. A.; Birhane, N.; Getie, B. DNA Damage, Transposable Element Expression and Their Associated Factors in Aging. Biochem. Mol. Biol. 2024, 9(3), 63-76. doi: 10.11648/j.bmb.20240904.11

    Copy | Download

    AMA Style

    Yeshanew TM, begashew BA, Birhane N, Getie B. DNA Damage, Transposable Element Expression and Their Associated Factors in Aging. Biochem Mol Biol. 2024;9(3):63-76. doi: 10.11648/j.bmb.20240904.11

    Copy | Download

  • @article{10.11648/j.bmb.20240904.11,
      author = {Temesgen Mitiku Yeshanew and Betelhem Abebe begashew and Nega Birhane and Birhan Getie},
      title = {DNA Damage, Transposable Element Expression and Their Associated Factors in Aging
    },
      journal = {Biochemistry and Molecular Biology},
      volume = {9},
      number = {3},
      pages = {63-76},
      doi = {10.11648/j.bmb.20240904.11},
      url = {https://doi.org/10.11648/j.bmb.20240904.11},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.bmb.20240904.11},
      abstract = {Aging is a spontaneous and permanent physiological process that leads to declines in tissue and cell functions, along with an increased risk of developing various age-related diseases. The primary driving force associated with aging is the accumulation of damaged genetic material in the cell, such as DNA. DNA damage can be caused by endogenous and exogenous factors, which leads to genome instability, mitochondrial dysfunction, epigenetic modifications, and proteostatic disturb. Another driving force associated with aging is the disruption of cellular metabolism. This disruption is closely linked to alterations in the role of metabolic pathways, including insulin/IGF-1 and mTOR, which regulate crucial cellular processes like cell growth, cell proliferation, and apoptosis. The activation of the insulin/IGF-1 signaling pathway highly promotes cell growth and proliferation, while also inhibits autophagy and increasing ROS production. This ultimately leads to accelerated aging. Another crucial signaling pathway is the mTOR signaling pathway. It is responsible for detecting nutrient availability and controlling cell growth and metabolism. The dysregulation of mTOR function can lead to the development of neurodegenerative diseases, which are characterized by the aggregation of protein. Activation of transposable elements is the other driving force of aging, caused by changes in DNA methylation and the loss of heterochromatin. As a result, this leads to DNA damage, genomic instability, and inflammation. The aim of this review is to elucidate the consequence of DNA damage and other associated factors drive aging.
    },
     year = {2024}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - DNA Damage, Transposable Element Expression and Their Associated Factors in Aging
    
    AU  - Temesgen Mitiku Yeshanew
    AU  - Betelhem Abebe begashew
    AU  - Nega Birhane
    AU  - Birhan Getie
    Y1  - 2024/11/29
    PY  - 2024
    N1  - https://doi.org/10.11648/j.bmb.20240904.11
    DO  - 10.11648/j.bmb.20240904.11
    T2  - Biochemistry and Molecular Biology
    JF  - Biochemistry and Molecular Biology
    JO  - Biochemistry and Molecular Biology
    SP  - 63
    EP  - 76
    PB  - Science Publishing Group
    SN  - 2575-5048
    UR  - https://doi.org/10.11648/j.bmb.20240904.11
    AB  - Aging is a spontaneous and permanent physiological process that leads to declines in tissue and cell functions, along with an increased risk of developing various age-related diseases. The primary driving force associated with aging is the accumulation of damaged genetic material in the cell, such as DNA. DNA damage can be caused by endogenous and exogenous factors, which leads to genome instability, mitochondrial dysfunction, epigenetic modifications, and proteostatic disturb. Another driving force associated with aging is the disruption of cellular metabolism. This disruption is closely linked to alterations in the role of metabolic pathways, including insulin/IGF-1 and mTOR, which regulate crucial cellular processes like cell growth, cell proliferation, and apoptosis. The activation of the insulin/IGF-1 signaling pathway highly promotes cell growth and proliferation, while also inhibits autophagy and increasing ROS production. This ultimately leads to accelerated aging. Another crucial signaling pathway is the mTOR signaling pathway. It is responsible for detecting nutrient availability and controlling cell growth and metabolism. The dysregulation of mTOR function can lead to the development of neurodegenerative diseases, which are characterized by the aggregation of protein. Activation of transposable elements is the other driving force of aging, caused by changes in DNA methylation and the loss of heterochromatin. As a result, this leads to DNA damage, genomic instability, and inflammation. The aim of this review is to elucidate the consequence of DNA damage and other associated factors drive aging.
    
    VL  - 9
    IS  - 3
    ER  - 

    Copy | Download

Author Information
  • Department of Medical Biotechnology, Institute of Biotechnology, Dambi Dollo University, Dambi Dollo, Oromia Ethiopia;Department of Medical Biotechnology, Institute of Biotechnology, University of Gondar, Gondar, Ethiopia

  • Department of Medical Biotechnology, Institute of Biotechnology, Dambi Dollo University, Dambi Dollo, Oromia Ethiopia;Department of Medical Biotechnology, Institute of Biotechnology, University of Gondar, Gondar, Ethiopia

  • Department of Medical Biotechnology, Institute of Biotechnology, University of Gondar, Gondar, Ethiopia

  • Department of Medical Biotechnology, Institute of Biotechnology, University of Gondar, Gondar, Ethiopia

  • Sections