Genomic Instability and Epigenetic Changes during Aging
Abstract
:1. Introduction
2. Telomere Shortening
3. DNA Damage Accumulation
4. Decreased DNA Repair Capacity
5. Histone Modification Changes
5.1. Histone Methylation
5.2. Histone Acetylation
5.3. Histone Phosphorylation and Ubiquitination
5.4. Histone Variants
6. DNA Methylation Changes
7. Non-Coding RNAs
8. RNA Modifications and Changes in the Epitranscriptome
9. Chromatin Organization Changes
9.1. Heterochromatin Changes
9.2. Chromatin Remodeling
9.3. Nucleosome and Histone Abundance
10. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
DNA | Desoxyribonucleic acid |
TOR | Target of Rapamycin |
RNA | Ribonucleic acid |
ROS | Reactive Oxygen Species |
UV | Ultraviolet |
8-OHdG | 8-hydroxydeoxyguanosine |
rDNA | Ribosomal DNA |
ERC | Extrachromosomal Ribosomal DNA Circles |
mtDNA | Mitochondrial DNA |
DDR | DNA Damage Repair |
MMR | Mismatch Repair |
BER | Base Excision Repair |
NER | Nucleotide Excision Repair |
DSB | Double Strand Break |
NHEJ | Non-Homologous End Joining |
ADP | Adenosine Diphosphate |
HMT | Histone Methyltransferase |
UPRmt | Mitochondrial Unfolded Protein Response |
HAT | Histone Acetyltransferase |
HDAC | Histone Deacetylase |
DNMT | DNA Methyltransferase |
5-mC | 5-methylcytosine |
CpG | Cytosine-phospho-guanine |
DNAm | DNA Methylation |
rRNA | Ribosomal RNA |
CR | Caloric Restriction |
ncRNA | Non-coding RNA |
miRNA | microRNA |
lncRNA | Long non-coding RNA |
IGF | Insulin-like Growth Factor |
rRNA | Ribosomal RNA |
RNAi | RNA interference |
tRNA | Transfer RNA |
mRNA | Messenger RNA |
SAHF | Senescence-associated Heterochromatin Foci |
ChIP | Chromatin Immunoprecipitation |
HSC | Hematopoietic Stem Cell |
MSC | Mesenchymal Stem Cell |
NuRD | Nucleosome Remodeling and histone Deacetylase |
References
- Sciubba, J.D. Population aging as a global issue. In Oxford Research Encyclopedia of International Studies; Oxford University Press: Oxford, UK, 2020; ISBN 9780190846626. [Google Scholar]
- Jarzebski, M.P.; Elmqvist, T.; Gasparatos, A.; Fukushi, K.; Eckersten, S.; Haase, D.; Goodness, J.; Khoshkar, S.; Saito, O.; Takeuchi, K.; et al. Ageing and population shrinking: Implications for sustainability in the urban century. npj Urban Sustain. 2021, 1, 17. [Google Scholar] [CrossRef]
- Eurostat. Population Structure Indicators at National Level. 2023. Available online: https://ec.europa.eu/eurostat/databrowser/view/DEMO_PJANIND__custom_964289/bookmark/table?lang=en&bookmarkId=599174db-325f-429b-87ba-6af6b18e9ca9 (accessed on 26 June 2023).
- Khan, A.H.; Zou, Z.; Xiang, Y.; Chen, S.; Tian, X.-L. Conserved signaling pathways genetically associated with longevity across the species. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 1745–1755. [Google Scholar] [CrossRef]
- López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. Hallmarks of aging: An expanding universe. Cell 2023, 186, 243–278. [Google Scholar] [CrossRef] [PubMed]
- Cheung, P.; Vallania, F.; Warsinske, H.C.; Donato, M.; Schaffert, S.; Chang, S.E.; Dvorak, M.; Dekker, C.L.; Davis, M.M.; Utz, P.J.; et al. Single-cell chromatin modification profiling reveals increased epigenetic variations with aging. Cell 2018, 173, 1385–1397.e14. [Google Scholar] [CrossRef]
- Fraga, M.F.; Ballestar, E.; Paz, M.F.; Ropero, S.; Setien, F.; Ballestar, M.L.; Heine-Suñer, D.; Cigudosa, J.C.; Urioste, M.; Benitez, J.; et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc. Natl. Acad. Sci. USA 2005, 102, 10604–10609. [Google Scholar] [CrossRef]
- Poulsen, P.; Esteller, M.; Vaag, A.; Fraga, M.F. The epigenetic basis of twin discordance in age-related diseases. Pediatr. Res. 2007, 61, 38R–42R. [Google Scholar] [CrossRef] [PubMed]
- Huang, B.; Jiang, C.; Zhang, R. Epigenetics: The language of the cell? Epigenomics 2014, 6, 73–88. [Google Scholar] [CrossRef]
- Patel, J.; Baptiste, B.A.; Kim, E.; Hussain, M.; Croteau, D.L.; Bohr, V.A. DNA damage and mitochondria in cancer and aging. Carcinogenesis 2020, 41, 1625–1634. [Google Scholar] [CrossRef]
- Aman, Y.; Schmauck-Medina, T.; Hansen, M.; Morimoto, R.I.; Simon, A.K.; Bjedov, I.; Palikaras, K.; Simonsen, A.; Johansen, T.; Tavernarakis, N.; et al. Autophagy in healthy aging and disease. Nat. Aging 2021, 1, 634–650. [Google Scholar] [CrossRef]
- Hipp, M.S.; Kasturi, P.; Hartl, F.U. The proteostasis network and its decline in ageing. Nat. Rev. Mol. Cell Biol. 2019, 20, 421–435. [Google Scholar] [CrossRef]
- Lima, T.; Li, T.Y.; Mottis, A.; Auwerx, J. Pleiotropic effects of mitochondria in aging. Nat. Aging 2022, 2, 199–213. [Google Scholar] [CrossRef] [PubMed]
- de Lucia, C.; Murphy, T.; Steves, C.J.; Dobson, R.J.B.; Proitsi, P.; Thuret, S. Lifestyle mediates the role of nutrient-sensing pathways in cognitive aging: Cellular and epidemiological evidence. Commun. Biol. 2020, 3, 157. [Google Scholar] [CrossRef] [PubMed]
- Ovadya, Y.; Krizhanovsky, V. Senescent cells: SASPected drivers of age-related pathologies. Biogerontology 2014, 15, 627–642. [Google Scholar] [CrossRef]
- Lee, Y.H.; Park, J.Y.; Lee, H.; Song, E.S.; Kuk, M.U.; Joo, J.; Oh, S.; Kwon, H.W.; Park, J.T.; Park, S.C. Targeting mitochondrial metabolism as a strategy to treat senescence. Cells 2021, 10, 3003. [Google Scholar] [CrossRef] [PubMed]
- Gonzales-Ebsen, A.C.; Gregersen, N.; Olsen, R.K. Linking telomere loss and mitochondrial dysfunction in chronic disease. Front. Biosci. 2017, 22, 117–127. [Google Scholar] [CrossRef] [PubMed]
- Liu, L.; Trimarchi, J.R.; Smith, P.J.S.; Keefe, D.L. Mitochondrial dysfunction leads to telomere attrition and genomic instability. Aging Cell 2002, 1, 40–46. [Google Scholar] [CrossRef]
- Saretzki, G.; Murphy, M.P.; von Zglinicki, T. MitoQ counteracts telomere shortening and elongates lifespan of fibroblasts under mild oxidative stress. Aging Cell 2003, 2, 141–143. [Google Scholar] [CrossRef]
- Zheng, Q.; Huang, J.; Wang, G. Mitochondria, telomeres and telomerase subunits. Front. Cell Dev. Biol. 2019, 7, 274. [Google Scholar] [CrossRef]
- Kumar, N.; Qian, W.; Van Houten, B. Sick mitochondria cause telomere damage: Implications for disease. Mol. Cell. Oncol. 2019, 7, 1678362. [Google Scholar] [CrossRef] [PubMed]
- Guo, N.; Parry, E.M.; Li, L.-S.; Kembou, F.; Lauder, N.; Hussain, M.A.; Berggren, P.-O.; Armanios, M. Short telomeres compromise β-cell signaling and survival. PLoS ONE 2011, 6, e17858. [Google Scholar] [CrossRef]
- Sahin, E.; Colla, S.; Liesa, M.; Moslehi, J.; Müller, F.L.; Guo, M.; Cooper, M.; Kotton, D.; Fabian, A.J.; Walkey, C.; et al. Telomere dysfunction induces metabolic and mitochondrial compromise. Nature 2011, 470, 359–365. [Google Scholar] [CrossRef]
- Schank, M.; Zhao, J.; Wang, L.; Li, Z.; Cao, D.; Nguyen, L.N.; Yao, Z.Q. Telomeric injury by KML001 in human T cells induces mitochondrial dysfunction through the p53-PGC-1alpha pathway. Cell Death Dis. 2020, 11, 1030. [Google Scholar] [CrossRef] [PubMed]
- Tzanetakou, I.P.; Nzietchueng, R.; Perrea, D.N.; Benetos, A. Telomeres and their role in aging and longevity. Curr. Vasc. Pharmacol. 2014, 12, 726–734. [Google Scholar] [CrossRef] [PubMed]
- Blackburn, E.H.; Collins, K. Telomerase: An RNP enzyme synthesizes DNA. Cold Spring Harb. Perspect. Biol. 2011, 3, a003558. [Google Scholar] [CrossRef] [PubMed]
- Shay, J.W. Role of telomeres and telomerase in aging and cancer. Cancer Discov. 2016, 6, 584–593. [Google Scholar] [CrossRef]
- Shay, J.W.; Wright, W.E. Telomeres and telomerase: Three decades of progress. Nat. Rev. Genet. 2019, 20, 299–309. [Google Scholar] [CrossRef]
- Harley, C.B.; Futcher, A.B.; Greider, C.W. Telomeres shorten during ageing of human fibroblasts. Nature 1990, 345, 458–460. [Google Scholar] [CrossRef]
- Epel, E.S.; Blackburn, E.H.; Lin, J.; Dhabhar, F.S.; Adler, N.E.; Morrow, J.D.; Cawthon, R.M. Accelerated telomere shortening in response to life stress. Proc. Natl. Acad. Sci. USA 2004, 101, 17312–17315. [Google Scholar] [CrossRef]
- Whittemore, K.; Vera, E.; Martínez-Nevado, E.; Sanpera, C.; Blasco, M.A. Telomere shortening rate predicts species life span. Proc. Natl. Acad. Sci. USA 2019, 116, 15122–15127. [Google Scholar] [CrossRef]
- Lin, J.; Epel, E. Stress and telomere shortening: Insights from cellular mechanisms. Ageing Res. Rev. 2022, 73, 101507. [Google Scholar] [CrossRef]
- Qian, W.; Kumar, N.; Roginskaya, V.; Fouquerel, E.; Opresko, P.L.; Shiva, S.; Watkins, S.C.; Kolodieznyi, D.; Bruchez, M.P.; Van Houten, B. Chemoptogenetic damage to mitochondria causes rapid telomere dysfunction. Proc. Natl. Acad. Sci. USA 2019, 116, 18435–18444. [Google Scholar] [CrossRef]
- Metcalfe, N.B.; Olsson, M. How telomere dynamics are influenced by the balance between mitochondrial efficiency, reactive oxygen species production and DNA damage. Mol. Ecol. 2022, 31, 6040–6052. [Google Scholar] [CrossRef]
- Collins, K.; Mitchell, J.R. Telomerase in the human organism. Oncogene 2002, 21, 564–579. [Google Scholar] [CrossRef]
- Henriques, C.M.; Ferreira, M.G. Consequences of telomere shortening during lifespan. Curr. Opin. Cell Biol. 2012, 24, 804–808. [Google Scholar] [CrossRef]
- Rossiello, F.; Jurk, D.; Passos, J.F.; d’Adda di Fagagna, F. Telomere dysfunction in ageing and age-related diseases. Nat. Cell Biol. 2022, 24, 135–147. [Google Scholar] [CrossRef] [PubMed]
- Blasco, M.A. Mice with bad ends: Mouse models for the study of telomeres and telomerase in cancer and aging. EMBO J. 2005, 24, 1095–1103. [Google Scholar] [CrossRef]
- Demanelis, K.; Jasmine, F.; Chen, L.S.; Chernoff, M.; Tong, L.; Delgado, D.; Zhang, C.; Shinkle, J.; Sabarinathan, M.; Lin, H.; et al. Determinants of telomere length across human tissues. Science 2020, 369, 1333. [Google Scholar] [CrossRef]
- Muñoz-Lorente, M.A.; Cano-Martin, A.C.; Blasco, M.A. Mice with hyper-long telomeres show less metabolic aging and longer lifespans. Nat. Commun. 2019, 10, 4723. [Google Scholar] [CrossRef]
- Harari, Y.; Zadok-Laviel, S.; Kupiec, M. Long telomeres do not affect cellular fitness in yeast. mBio 2017, 8, e01314-17. [Google Scholar] [CrossRef] [PubMed]
- Raices, M.; Maruyama, H.; Dillin, A.; Karlseder, J. Uncoupling of longevity and telomere length in C. elegans. PLoS Genet. 2005, 1, e30. [Google Scholar] [CrossRef]
- Walter, M.F.; Biessmann, M.R.; Benitez, C.; Török, T.; Mason, J.M.; Biessmann, H. Effects of telomere length in Drosophila melanogaster on life span, fecundity, and fertility. Chromosoma 2007, 116, 41–51. [Google Scholar] [CrossRef]
- Samper, E.; Flores, J.M.; Blasco, M.A. Restoration of telomerase activity rescues chromosomal instability and premature aging in Terc−/− mice with short telomeres. EMBO Rep. 2001, 2, 800–807. [Google Scholar] [CrossRef]
- Bernardes De Jesus, B.; Vera, E.; Schneeberger, K.; Tejera, A.M.; Ayuso, E.; Bosch, F.; Blasco, M.A. Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer. EMBO Mol. Med. 2012, 4, 691–704. [Google Scholar] [CrossRef] [PubMed]
- Flores, I.; Benetti, R.; Blasco, M.A. Telomerase regulation and stem cell behaviour. Curr. Opin. Cell Biol. 2006, 18, 254–260. [Google Scholar] [CrossRef]
- Blasco, M.A. Telomere length, stem cells and aging. Nat. Chem. Biol. 2007, 3, 640–649. [Google Scholar] [CrossRef] [PubMed]
- Vijg, J. From DNA damage to mutations: All roads lead to aging. Ageing Res. Rev. 2021, 68, 101316. [Google Scholar] [CrossRef]
- Schumacher, B.; Pothof, J.; Vijg, J.; Hoeijmakers, J.H.J. The central role of DNA damage in the ageing process. Nature 2021, 592, 695–703. [Google Scholar] [CrossRef] [PubMed]
- Yousefzadeh, M.; Henpita, C.; Vyas, R.; Soto-Palma, C.; Robbins, P.; Niedernhofer, L. DNA damage—How and why we age? eLife 2021, 10, e62852. [Google Scholar] [CrossRef]
- Lindahl, T. Instability and decay of the primary structure of DNA. Nature 1993, 362, 709–715. [Google Scholar] [CrossRef]
- Niedernhofer, L.J.; Gurkar, A.U.; Wang, Y.; Vijg, J.; Hoeijmakers, J.H.J.; Robbins, P.D. Nuclear genomic instability and aging. Annu. Rev. Biochem. 2018, 87, 295–322. [Google Scholar] [CrossRef]
- Lubberts, S.; Meijer, C.; Demaria, M.; Gietema, J.A. Early ageing after cytotoxic treatment for testicular cancer and cellular senescence: Time to act. Crit. Rev. Oncol. Hematol. 2020, 151, 102963. [Google Scholar] [CrossRef]
- Killock, D. Chemotherapy: Life gained, years lost? Nat. Rev. Clin. Oncol. 2014, 11, 303. [Google Scholar] [CrossRef]
- Roos, W.P.; Kaina, B. DNA damage-induced cell death by apoptosis. Trends Mol. Med. 2006, 12, 440–450. [Google Scholar] [CrossRef]
- d’Adda Di Fagagna, F. Living on a break: Cellular senescence as a DNA-damage response. Nat. Rev. Cancer 2008, 8, 512–522. [Google Scholar] [CrossRef]
- Baker, D.J.; Childs, B.G.; Durik, M.; Wijers, M.E.; Sieben, C.J.; Zhong, J.; Saltness, R.A.; Jeganathan, K.B.; Verzosa, G.C.; Pezeshki, A.; et al. Naturally occurring p16Ink4a-positive cells shorten healthy lifespan. Nature 2016, 530, 184–189. [Google Scholar] [CrossRef] [PubMed]
- Gorbunova, V.; Seluanov, A. DNA double strand break repair, aging and the chromatin connection. Mutat. Res. 2016, 788, 2–6. [Google Scholar] [CrossRef] [PubMed]
- Gorbunova, V.; Seluanov, A.; Mao, Z.; Hine, C. Changes in DNA repair during aging. Nucleic Acids Res. 2007, 35, 7466–7474. [Google Scholar] [CrossRef]
- Lu, T.; Pan, Y.; Kao, S.-Y.; Li, C.; Kohane, I.; Chan, J.; Yankner, B.A. Gene regulation and DNA damage in the ageing human brain. Nature 2004, 429, 883–891. [Google Scholar] [CrossRef]
- Arvanitaki, E.S.; Stratigi, K.; Garinis, G.A. DNA damage, inflammation and aging: Insights from mice. Front. Aging 2022, 3, 973781. [Google Scholar] [CrossRef] [PubMed]
- Dollé, M.E.T.; Snyder, W.K.; Gossen, J.A.; Lohman, P.H.M.; Vijg, J. Distinct spectra of somatic mutations accumulated with age in mouse heart and small intestine. Proc. Natl. Acad. Sci. USA 2000, 97, 8403–8408. [Google Scholar] [CrossRef] [PubMed]
- Brazhnik, K.; Sun, S.; Alani, O.; Kinkhabwala, M.; Wolkoff, A.W.; Maslov, A.Y.; Dong, X.; Vijg, J. Single-cell analysis reveals different age-related somatic mutation profiles between stem and differentiated cells in human liver. Sci. Adv. 2020, 6, eaax2659. [Google Scholar] [CrossRef] [PubMed]
- Risques, R.A.; Kennedy, S.R. Aging and the rise of somatic cancer-associated mutations in normal tissues. PLoS Genet. 2018, 14, e1007108. [Google Scholar] [CrossRef] [PubMed]
- Ren, P.; Dong, X.; Vijg, J. Age-related somatic mutation burden in human tissues. Front. Aging 2022, 3, 1018119. [Google Scholar] [CrossRef] [PubMed]
- Lodato, M.A.; Rodin, R.E.; Bohrson, C.L.; Coulter, M.E.; Barton, A.R.; Kwon, M.; Sherman, M.A.; Vitzthum, C.M.; Luquette, L.J.; Yandava, C.N.; et al. Aging and neurodegeneration are associated with increased mutations in single human neurons. Science 2018, 359, 555–559. [Google Scholar] [CrossRef]
- Zhang, L.; Dong, X.; Lee, M.; Maslov, A.Y.; Wang, T.; Vijg, J. Single-cell whole-genome sequencing reveals the functional landscape of somatic mutations in B lymphocytes across the human lifespan. Proc. Natl. Acad. Sci. USA 2019, 116, 9014–9019. [Google Scholar] [CrossRef]
- Cagan, A.; Baez-Ortega, A.; Brzozowska, N.; Abascal, F.; Coorens, T.H.H.; Sanders, M.A.; Lawson, A.R.J.; Harvey, L.M.R.; Bhosle, S.; Jones, D.; et al. Somatic mutation rates scale with lifespan across mammals. Nature 2022, 604, 517–524. [Google Scholar] [CrossRef]
- O’Hagan, H.M.; Wang, W.; Sen, S.; DeStefano Shields, C.; Lee, S.S.; Zhang, Y.W.; Clements, E.G.; Cai, Y.; Van Neste, L.; Easwaran, H.; et al. Oxidative damage targets complexes containing dna methyltransferases, sirt1, and polycomb members to promoter cpg islands. Cancer Cell 2011, 20, 606–619. [Google Scholar] [CrossRef]
- Lee, J.W.; Ong, E.B.B. Genomic instability and cellular senescence: Lessons from the budding yeast. Front. Cell Dev. Biol. 2021, 8, 619126. [Google Scholar] [CrossRef]
- Kobayashi, T. How does genome instability affect lifespan?: Roles of rDNA and telomeres. Genes Cells 2011, 16, 617–624. [Google Scholar] [CrossRef]
- Sinclair, D.A.; Guarente, L. Extrachromosomal rDNA circles—A cause of aging in yeast. Cell 1997, 91, 1033–1042. [Google Scholar] [CrossRef]
- Mansisidor, A.; Molinar, T.; Srivastava, P.; Dartis, D.D.; Pino Delgado, A.; Blitzblau, H.G.; Klein, H.; Hochwagen, A. Genomic Copy-Number Loss Is Rescued by Self-Limiting Production of DNA Circles. Mol. Cell 2018, 72, 583–593.e4. [Google Scholar] [CrossRef] [PubMed]
- Morlot, S.; Song, J.; Léger-Silvestre, I.; Matifas, A.; Gadal, O.; Charvin, G. Excessive rDNA Transcription Drives the Disruption in Nuclear Homeostasis during Entry into Senescence in Budding Yeast. Cell Rep. 2019, 28, 408–422.e4. [Google Scholar] [CrossRef] [PubMed]
- Denoth-Lippuner, A.; Krzyzanowski, M.K.; Stober, C.; Barral, Y. Role of SAGA in the asymmetric segregation of DNA circles during yeast ageing. eLife 2014, 3, e03790. [Google Scholar] [CrossRef]
- Kasselimi, E.; Pefani, D.-E.; Taraviras, S.; Lygerou, Z. Ribosomal DNA and the nucleolus at the heart of aging. Trends Biochem. Sci. 2022, 47, 328–341. [Google Scholar] [CrossRef] [PubMed]
- Smith, J.S. The long and short of rDNA and yeast replicative aging. Proc. Natl. Acad. Sci. USA 2022, 119, e2205124119. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Jiang, Y.; Paxman, J.; O’Laughlin, R.; Klepin, S.; Zhu, Y.; Pillus, L.; Tsimring, L.S.; Hasty, J.; Hao, N. A programmable fate decision landscape underlies single-cell aging in yeast. Science 2020, 369, 325–329. [Google Scholar] [CrossRef] [PubMed]
- Hotz, M.; Thayer, N.H.; Hendrickson, D.G.; Schinski, E.L.; Xu, J.; Gottschling, D.E. rDNA array length is a major determinant of replicative lifespan in budding yeast. Proc. Natl. Acad. Sci. USA 2022, 119, e2119593119. [Google Scholar] [CrossRef]
- Payea, M.J.; Anerillas, C.; Tharakan, R.; Gorospe, M. Translational Control during Cellular Senescence. Mol. Cell. Biol. 2021, 41, e00512-20. [Google Scholar] [CrossRef]
- Warmerdam, D.O.; Wolthuis, R.M.F. Keeping ribosomal DNA intact: A repeating challenge. Chromosome Res. 2019, 27, 57–72. [Google Scholar] [CrossRef]
- Watada, E.; Li, S.; Hori, Y.; Fujiki, K.; Shirahige, K.; Inada, T.; Kobayashi, T. Age-Dependent Ribosomal DNA Variations in Mice. Mol. Cell. Biol. 2020, 40, e00368-20. [Google Scholar] [CrossRef]
- Lu, K.L.; Nelson, J.O.; Watase, G.J.; Warsinger-Pepe, N.; Yamashita, Y.M. Transgenerational dynamics of rDNA copy number in Drosophila male germline stem cells. eLife 2018, 7, e32421. [Google Scholar] [CrossRef] [PubMed]
- Shastri, N.; Tsai, Y.-C.; Hile, S.; Jordan, D.; Powell, B.; Chen, J.; Maloney, D.; Dose, M.; Lo, Y.; Anastassiadis, T.; et al. Genome-wide Identification of Structure-Forming Repeats as Principal Sites of Fork Collapse upon ATR Inhibition. Mol. Cell 2018, 72, 222–238.e11. [Google Scholar] [CrossRef] [PubMed]
- Flach, J.; Bakker, S.T.; Mohrin, M.; Conroy, P.C.; Pietras, E.M.; Reynaud, D.; Alvarez, S.; Diolaiti, M.E.; Ugarte, F.; Forsberg, E.C.; et al. Replication stress is a potent driver of functional decline in ageing haematopoietic stem cells. Nature 2014, 512, 198–202. [Google Scholar] [CrossRef]
- Petropoulos, M.; Champeris Tsaniras, S.; Taraviras, S.; Lygerou, Z. Replication licensing aberrations, replication stress, and genomic instability. Trends Biochem. Sci. 2019, 44, 752–764. [Google Scholar] [CrossRef] [PubMed]
- Ren, R.; Deng, L.; Xue, Y.; Suzuki, K.; Zhang, W.; Yu, Y.; Wu, J.; Sun, L.; Gong, X.; Luan, H.; et al. Visualization of aging-associated chromatin alterations with an engineered TALE system. Cell Res. 2017, 27, 483–504. [Google Scholar] [CrossRef]
- Kauppila, T.E.S.; Kauppila, J.H.K.; Larsson, N.-G. Mammalian mitochondria and aging: An update. Cell Metab. 2017, 25, 57–71. [Google Scholar] [CrossRef]
- Trifunovic, A.; Wredenberg, A.; Falkenberg, M.; Spelbrink, J.N.; Rovio, A.T.; Bruder, C.E.; Bohlooly-Y, M.; Gidlöf, S.; Oldfors, A.; Wibom, R.; et al. Premature ageing in mice expressing defective mitochondrial DNA polymerase. Nature 2004, 429, 417–423. [Google Scholar] [CrossRef]
- Kujoth, G.C.; Hiona, A.; Pugh, T.D.; Someya, S.; Panzer, K.; Wohlgemuth, S.E.; Hofer, T.; Seo, A.Y.; Sullivan, R.; Jobling, W.A.; et al. Mitochondrial DNA mutations, oxidative stress, and apoptosis in mammalian aging. Science 2005, 309, 481–484. [Google Scholar] [CrossRef]
- Bua, E.; Johnson, J.; Herbst, A.; Delong, B.; McKenzie, D.; Salamat, S.; Aiken, J.M. Mitochondrial DNA-deletion mutations accumulate intracellularly to detrimental levels in aged human skeletal muscle fibers. Am. J. Hum. Genet. 2006, 79, 469–480. [Google Scholar] [CrossRef]
- Vermulst, M.; Bielas, J.H.; Kujoth, G.C.; Ladiges, W.C.; Rabinovitch, P.S.; Prolla, T.A.; Loeb, L.A. Mitochondrial point mutations do not limit the natural lifespan of mice. Nat. Genet. 2007, 39, 540–543. [Google Scholar] [CrossRef]
- O’Hara, R.; Tedone, E.; Ludlow, A.; Huang, E.; Arosio, B.; Mari, D.; Shay, J.W. Quantitative mitochondrial DNA copy number determination using droplet digital PCR with single-cell resolution. Genome Res. 2019, 29, 1878–1888. [Google Scholar] [CrossRef]
- Ameur, A.; Stewart, J.B.; Freyer, C.; Hagström, E.; Ingman, M.; Larsson, N.-G.; Gyllensten, U. Ultra-deep sequencing of mouse mitochondrial DNA: Mutational patterns and their origins. PLoS Genet. 2011, 7, e1002028. [Google Scholar] [CrossRef] [PubMed]
- Dabin, J.; Fortuny, A.; Polo, S.E. Epigenome maintenance in response to dna damage. Mol. Cell 2016, 62, 712–727. [Google Scholar] [CrossRef]
- Wang, S.; Meyer, D.H.; Schumacher, B. H3K4me2 regulates the recovery of protein biosynthesis and homeostasis following DNA damage. Nat. Struct. Mol. Biol. 2020, 27, 1165–1177. [Google Scholar] [CrossRef]
- Ito, T.; Teo, Y.V.; Evans, S.A.; Neretti, N.; Sedivy, J.M. Regulation of Cellular Senescence by Polycomb Chromatin Modifiers through Distinct DNA Damage- and Histone Methylation-Dependent Pathways. Cell Rep. 2018, 22, 3480–3492. [Google Scholar] [CrossRef]
- Sedelnikova, O.A.; Horikawa, I.; Zimonjic, D.B.; Popescu, N.C.; Bonner, W.M.; Barrett, J.C. Senescing human cells and ageing mice accumulate DNA lesions with unrepairable double-strand breaks. Nat. Cell Biol. 2004, 6, 168–170. [Google Scholar] [CrossRef] [PubMed]
- Bahar, R.; Hartmann, C.H.; Rodriguez, K.A.; Denny, A.D.; Busuttil, R.A.; Dollé, M.E.T.; Calder, R.B.; Chisholm, G.B.; Pollock, B.H.; Klein, C.A.; et al. Increased cell-to-cell variation in gene expression in ageing mouse heart. Nature 2006, 441, 1011–1014. [Google Scholar] [CrossRef]
- Weissman, L.; Jo, D.-G.; Sørensen, M.M.; de Souza-Pinto, N.C.; Markesbery, W.R.; Mattson, M.P.; Bohr, V.A. Defective DNA base excision repair in brain from individuals with Alzheimer’s disease and amnestic mild cognitive impairment. Nucleic Acids Res. 2007, 35, 5545–5555. [Google Scholar] [CrossRef]
- Sepe, S.; Milanese, C.; Gabriels, S.; Derks, K.W.J.; Payan-Gomez, C.; van IJcken, W.F.J.; Rijksen, Y.M.A.; Nigg, A.L.; Moreno, S.; Cerri, S.; et al. Inefficient DNA Repair Is an Aging-Related Modifier of Parkinson’s Disease. Cell Rep. 2016, 15, 1866–1875. [Google Scholar] [CrossRef]
- Obulesu, M.; Rao, D.M. DNA damage and impairment of DNA repair in Alzheimer’s disease. Int. J. Neurosci. 2010, 120, 397–403. [Google Scholar] [CrossRef] [PubMed]
- Bucholtz, N.; Demuth, I. DNA-repair in mild cognitive impairment and Alzheimer’s disease. DNA Repair 2013, 12, 811–816. [Google Scholar] [CrossRef]
- Lopes, A.F.C.; Bozek, K.; Herholz, M.; Trifunovic, A.; Rieckher, M.; Schumacher, B. A C. elegans model for neurodegeneration in Cockayne syndrome. Nucleic Acids Res. 2020, 48, 10973–10985. [Google Scholar] [CrossRef] [PubMed]
- Sepe, S.; Payan-Gomez, C.; Milanese, C.; Hoeijmakers, J.H.; Mastroberardino, P.G. Nucleotide excision repair in chronic neurodegenerative diseases. DNA Repair 2013, 12, 568–577. [Google Scholar] [CrossRef] [PubMed]
- Edifizi, D.; Nolte, H.; Babu, V.; Castells-Roca, L.; Mueller, M.M.; Brodesser, S.; Krüger, M.; Schumacher, B. Multilayered reprogramming in response to persistent DNA damage in C. elegans. Cell Rep. 2017, 20, 2026–2043. [Google Scholar] [CrossRef] [PubMed]
- Hoeijmakers, J.H.J. DNA damage, aging, and cancer. N. Engl. J. Med. 2009, 361, 1475–1485. [Google Scholar] [CrossRef]
- Huang, R.; Zhou, P.-K. DNA damage repair: Historical perspectives, mechanistic pathways and clinical translation for targeted cancer therapy. Signal Transduct. Target. Ther. 2021, 6, 254. [Google Scholar] [CrossRef] [PubMed]
- Chatterjee, N.; Walker, G.C. Mechanisms of DNA damage, repair, and mutagenesis. Environ. Mol. Mutagen. 2017, 58, 235–263. [Google Scholar] [CrossRef]
- Garm, C.; Moreno-Villanueva, M.; Bürkle, A.; Petersen, I.; Bohr, V.A.; Christensen, K.; Stevnsner, T. Age and gender effects on DNA strand break repair in peripheral blood mononuclear cells. Aging Cell 2013, 12, 58–66. [Google Scholar] [CrossRef]
- Ju, Y.-J.; Lee, K.-H.; Park, J.-E.; Yi, Y.-S.; Yun, M.-Y.; Ham, Y.-H.; Kim, T.-J.; Choi, H.M.; Han, G.J.; Lee, J.-H.; et al. Decreased expression of DNA repair proteins Ku70 and Mre11 is associated with aging and may contribute to the cellular senescence. Exp. Mol. Med. 2006, 38, 686–693. [Google Scholar] [CrossRef] [PubMed]
- Mao, Z.; Tian, X.; Van Meter, M.; Ke, Z.; Gorbunova, V.; Seluanov, A. Sirtuin 6 (Sirt6) rescues the decline of homologous recombination repair during replicative senescence. Proc. Natl. Acad. Sci. USA 2012, 109, 11800–11805. [Google Scholar] [CrossRef] [PubMed]
- Vaidya, A.; Mao, Z.; Tian, X.; Spencer, B.; Seluanov, A.; Gorbunova, V. Knock-in reporter mice demonstrate that dna repair by non-homologous end joining declines with age. PLoS Genet. 2014, 10, e1004511. [Google Scholar] [CrossRef]
- Mojumdar, A.; Mair, N.; Adam, N.; Cobb, J.A. Changes in DNA double-strand break repair during aging correlate with an increase in genomic mutations. J. Mol. Biol. 2022, 434, 167798. [Google Scholar] [CrossRef]
- Park, J.-S.; Jeon, H.-J.; Pyo, J.-H.; Kim, Y.-S.; Yoo, M.-A. Deficiency in DNA damage response of enterocytes accelerates intestinal stem cell aging in Drosophila. Aging 2018, 10, 322–338. [Google Scholar] [CrossRef] [PubMed]
- Delabaere, L.; Ertl, H.A.; Massey, D.J.; Hofley, C.M.; Sohail, F.; Bienenstock, E.J.; Sebastian, H.; Chiolo, I.; LaRocque, J.R. Aging impairs double-strand break repair by homologous recombination in Drosophila germ cells. Aging Cell 2017, 16, 320–328. [Google Scholar] [CrossRef]
- Guedj, A.; Geiger-Maor, A.; Galun, E.; Benyamini, H.; Nevo, Y.; Elgavish, S.; Amsalem, H.; Rachmilewitz, J. Early age decline in DNA repair capacity in the liver: In depth profile of differential gene expression. Aging 2016, 8, 3131–3146. [Google Scholar] [CrossRef]
- Meyer, J.N.; Boyd, W.A.; Azzam, G.A.; Haugen, A.C.; Freedman, J.H.; Van Houten, B. Decline of nucleotide excision repair capacity in aging Caenorhabditis elegans. Genome Biol. 2007, 8, R70. [Google Scholar] [CrossRef]
- Menck, C.F.; Munford, V. DNA repair diseases: What do they tell us about cancer and aging? Genet. Mol. Biol. 2014, 37, 220–233. [Google Scholar] [CrossRef] [PubMed]
- Burla, R.; La Torre, M.; Merigliano, C.; Vernì, F.; Saggio, I. Genomic instability and DNA replication defects in progeroid syndromes. Nucleus 2018, 9, 368–379. [Google Scholar] [CrossRef] [PubMed]
- Martin, G.M.; Oshima, J. Lessons from human progeroid syndromes. Nature 2000, 408, 263–266. [Google Scholar] [CrossRef]
- de Magalhães, J.P. Open-minded scepticism: Inferring the causal mechanisms of human ageing from genetic perturbations. Ageing Res. Rev. 2005, 4, 1–22. [Google Scholar] [CrossRef]
- Gonzalo, S.; Kreienkamp, R. DNA repair defects and genome instability in Hutchinson-Gilford Progeria Syndrome. Curr. Opin. Cell Biol. 2015, 34, 75–83. [Google Scholar] [CrossRef]
- Oshima, J.; Sidorova, J.M.; Monnat, R.J., Jr. Werner syndrome: Clinical features, pathogenesis and potential therapeutic interventions. Ageing Res. Rev. 2017, 33, 105–114. [Google Scholar] [CrossRef] [PubMed]
- Karikkineth, A.C.; Scheibye-Knudsen, M.; Fivenson, E.; Croteau, D.L.; Bohr, V.A. Cockayne syndrome: Clinical features, model systems and pathways. Ageing Res. Rev. 2017, 33, 3–17. [Google Scholar] [CrossRef] [PubMed]
- Niedernhofer, L.J.; Garinis, G.A.; Raams, A.; Lalai, A.S.; Robinson, A.R.; Appeldoorn, E.; Odijk, H.; Oostendorp, R.; Ahmad, A.; van Leeuwen, W.; et al. A new progeroid syndrome reveals that genotoxic stress suppresses the somatotroph axis. Nature 2006, 444, 1038–1043. [Google Scholar] [CrossRef] [PubMed]
- Garagnani, P.; Marquis, J.; Delledonne, M.; Pirazzini, C.; Marasco, E.; Kwiatkowska, K.M.; Iannuzzi, V.; Bacalini, M.G.; Valsesia, A.; Carayol, J.; et al. Whole-genome sequencing analysis of semi-supercentenarians. eLife 2021, 10, e57849. [Google Scholar] [CrossRef]
- Tian, X.; Firsanov, D.; Zhang, Z.; Cheng, Y.; Luo, L.; Tombline, G.; Tan, R.; Simon, M.; Henderson, S.; Steffan, J.; et al. SIRT6 Is Responsible for More Efficient DNA Double-Strand Break Repair in Long-Lived Species. Cell 2019, 177, 622–638.e22. [Google Scholar] [CrossRef]
- Garschall, K.; Dellago, H.; Gáliková, M.; Schosserer, M.; Flatt, T.; Grillari, J. Ubiquitous overexpression of the DNA repair factor dPrp19 reduces DNA damage and extends Drosophila life span. npj Aging Mech. Dis. 2017, 3, 5. [Google Scholar] [CrossRef] [PubMed]
- Shaposhnikov, M.; Proshkina, E.; Shilova, L.; Zhavoronkov, A.; Moskalev, A. Lifespan and Stress Resistance in Drosophila with Overexpressed DNA Repair Genes. Sci. Rep. 2015, 5, 15299. [Google Scholar] [CrossRef]
- Torres, I.O.; Fujimori, D.G. Functional coupling between writers, erasers and readers of histone and DNA methylation. Curr. Opin.Struct. Biol. 2015, 35, 68–75. [Google Scholar] [CrossRef]
- Kouzarides, T. Chromatin modifications and their function. Cell 2007, 128, 693–705. [Google Scholar] [CrossRef]
- Iwasaki, W.; Miya, Y.; Horikoshi, N.; Osakabe, A.; Taguchi, H.; Tachiwana, H.; Shibata, T.; Kagawa, W.; Kurumizaka, H. Contribution of histone N-terminal tails to the structure and stability of nucleosomes. FEBS Open Bio 2013, 3, 363–369. [Google Scholar] [CrossRef]
- Kane, A.E.; Sinclair, D.A. Epigenetic changes during aging and their reprogramming potential. Crit. Rev. Biochem. Mol. Biol. 2019, 54, 61–83. [Google Scholar] [CrossRef]
- Galow, A.-M.; Peleg, S. How to slow down the ticking clock: Age-associated epigenetic alterations and related interventions to extend life span. Cells 2022, 11, 468. [Google Scholar] [CrossRef] [PubMed]
- Yi, S.-J.; Kim, K. New Insights into the Role of Histone Changes in Aging. Int. J. Mol. Sci. 2020, 21, 8241. [Google Scholar] [CrossRef] [PubMed]
- Greer, E.L.; Shi, Y. Histone methylation: A dynamic mark in health, disease and inheritance. Nat. Rev. Genet. 2012, 13, 343–357. [Google Scholar] [CrossRef] [PubMed]
- Saul, D.; Kosinsky, R.L. Epigenetics of aging and aging-associated diseases. Int. J. Mol. Sci. 2021, 22, 401. [Google Scholar] [CrossRef] [PubMed]
- Sen, P.; Shah, P.P.; Nativio, R.; Berger, S.L. Epigenetic mechanisms of longevity and aging. Cell 2016, 166, 822–839. [Google Scholar] [CrossRef]
- Benayoun, B.A.; Pollina, E.A.; Brunet, A. Epigenetic regulation of ageing: Linking environmental inputs to genomic stability. Nat. Rev. Mol. Cell Biol. 2015, 16, 593–610. [Google Scholar] [CrossRef] [PubMed]
- Pal, S.; Tyler, J.K. Epigenetics and aging. Sci. Adv. 2016, 2, e1600584. [Google Scholar] [CrossRef]
- Cole, J.J.; Robertson, N.A.; Rather, M.I.; Thomson, J.P.; McBryan, T.; Sproul, D.; Wang, T.; Brock, C.; Clark, W.; Ideker, T.; et al. Diverse interventions that extend mouse lifespan suppress shared age-associated epigenetic changes at critical gene regulatory regions. Genome Biol. 2017, 18, 58. [Google Scholar] [CrossRef]
- Siebold, A.P.; Banerjee, R.; Tie, F.; Kiss, D.L.; Moskowitz, J.; Harte, P.J. Polycomb Repressive Complex 2 and Trithorax modulate Drosophila longevity and stress resistance. Proc. Natl. Acad. Sci. USA 2010, 107, 169–174. [Google Scholar] [CrossRef] [PubMed]
- Maures, T.J.; Greer, E.L.; Hauswirth, A.G.; Brunet, A. The H3K27 demethylase UTX-1 regulates C. elegans lifespan in a germline-independent, insulin-dependent manner. Aging Cell 2011, 10, 980–990. [Google Scholar] [CrossRef] [PubMed]
- Cruz, C.; Della Rosa, M.; Krueger, C.; Gao, Q.; Horkai, D.; King, M.; Field, L.; Houseley, J. Tri-methylation of histone H3 lysine 4 facilitates gene expression in ageing cells. eLife 2018, 7, e34081. [Google Scholar] [CrossRef] [PubMed]
- Pu, M.; Wang, M.; Wang, W.; Velayudhan, S.S.; Lee, S.S. Unique patterns of trimethylation of histone H3 lysine 4 are prone to changes during aging in Caenorhabditis elegans somatic cells. PLoS Genet. 2018, 14, e1007466. [Google Scholar] [CrossRef]
- Greer, E.L.; Maures, T.J.; Hauswirth, A.G.; Green, E.M.; Leeman, D.S.; Maro, G.S.; Han, S.; Banko, M.R.; Gozani, O.; Brunet, A. Members of the H3K4 trimethylation complex regulate lifespan in a germline-dependent manner in C. elegans. Nature 2010, 466, 383–387. [Google Scholar] [CrossRef]
- Cao, Q.; Wang, W.; Williams, J.B.; Yang, F.; Wang, Z.-J.; Yan, Z. Targeting histone K4 trimethylation for treatment of cognitive and synaptic deficits in mouse models of Alzheimer’s disease. Sci. Adv. 2020, 6, eabc8096. [Google Scholar] [CrossRef]
- Sun, D.; Luo, M.; Jeong, M.; Rodriguez, B.; Xia, Z.; Hannah, R.; Wang, H.; Le, T.; Faull, K.F.; Chen, R.; et al. Epigenomic profiling of young and aged HSCs reveals concerted changes during aging that reinforce self-renewal. Cell Stem Cell 2014, 14, 673–688. [Google Scholar] [CrossRef]
- Cheung, I.; Shulha, H.P.; Jiang, Y.; Matevossian, A.; Wang, J.; Weng, Z.; Akbarian, S. Developmental regulation and individual differences of neuronal H3K4me3 epigenomes in the prefrontal cortex. Proc. Natl. Acad. Sci. USA 2010, 107, 8824–8829. [Google Scholar] [CrossRef] [PubMed]
- Adelman, E.R.; Huang, H.-T.; Roisman, A.; Olsson, A.; Colaprico, A.; Qin, T.; Lindsley, R.C.; Bejar, R.; Salomonis, N.; Grimes, H.L.; et al. Aging human hematopoietic stem cells manifest profound epigenetic reprogramming of enhancers that may predispose to leukemia. Cancer Discov. 2019, 9, 1080–1101. [Google Scholar] [CrossRef]
- Bonasio, R.; Tu, S.; Reinberg, D. Molecular signals of epigenetic states. Science 2010, 330, 612–616. [Google Scholar] [CrossRef]
- Shumaker, D.K.; Dechat, T.; Kohlmaier, A.; Adam, S.A.; Bozovsky, M.R.; Erdos, M.R.; Eriksson, M.; Goldman, A.E.; Khuon, S.; Collins, F.S.; et al. Mutant nuclear lamin A leads to progressive alterations of epigenetic control in premature aging. Proc. Natl. Acad. Sci. USA 2006, 103, 8703–8708. [Google Scholar] [CrossRef] [PubMed]
- Baumgart, M.; Groth, M.; Priebe, S.; Savino, A.; Testa, G.; Dix, A.; Ripa, R.; Spallotta, F.; Gaetano, C.; Ori, M.; et al. RNA-seq of the aging brain in the short-lived fish N. furzeri—Conserved pathways and novel genes associated with neurogenesis. Aging Cell 2014, 13, 965–974. [Google Scholar] [CrossRef]
- Liu, L.; Cheung, T.H.; Charville, G.W.; Hurgo, B.M.C.; Leavitt, T.; Shih, J.; Brunet, A.; Rando, T.A. Chromatin modifications as determinants of muscle stem cell quiescence and chronological aging. Cell Rep. 2013, 4, 189–204. [Google Scholar] [CrossRef] [PubMed]
- Guillermo, A.R.R.; Chocian, K.; Gavriilidis, G.; Vandamme, J.; Salcini, A.E.; Mellor, J.; Woollard, A. H3K27 modifiers regulate lifespan in C. elegans in a context-dependent manner. BMC Biol. 2021, 19, 59. [Google Scholar] [CrossRef] [PubMed]
- Jin, C.; Li, J.; Green, C.D.; Yu, X.; Tang, X.; Han, D.; Xian, B.; Wang, D.; Huang, X.; Cao, X.; et al. Histone demethylase UTX-1 regulates C. elegans life span by targeting the insulin/IGF-1 signaling pathway. Cell Metab. 2011, 14, 161–172. [Google Scholar] [CrossRef]
- Ni, Z.; Ebata, A.; Alipanahiramandi, E.; Lee, S.S. Two SET domain containing genes link epigenetic changes and aging in Caenorhabditis elegans: Two worm genes link histone modifications and aging. Aging Cell 2012, 11, 315–325. [Google Scholar] [CrossRef]
- Merkwirth, C.; Jovaisaite, V.; Durieux, J.; Matilainen, O.; Jordan, S.D.; Quiros, P.M.; Steffen, K.K.; Williams, E.G.; Mouchiroud, L.; Tronnes, S.U.; et al. Two Conserved Histone Demethylases Regulate Mitochondrial Stress-Induced Longevity. Cell 2016, 165, 1209–1223. [Google Scholar] [CrossRef]
- Nejman, D.; Straussman, R.; Steinfeld, I.; Ruvolo, M.; Roberts, D.; Yakhini, Z.; Cedar, H. Molecular rules governing de novo methylation in cancer. Cancer Res. 2014, 74, 1475–1483. [Google Scholar] [CrossRef]
- Sen, P.; Dang, W.; Donahue, G.; Dai, J.; Dorsey, J.; Cao, X.; Liu, W.; Cao, K.; Perry, R.; Lee, J.Y.; et al. H3K36 methylation promotes longevity by enhancing transcriptional fidelity. Genes Dev. 2015, 29, 1362–1376. [Google Scholar] [CrossRef]
- Jeon, H.-J.; Kim, Y.-S.; Kim, J.-G.; Heo, K.; Pyo, J.-H.; Yamaguchi, M.; Park, J.-S.; Yoo, M.-A. Effect of heterochromatin stability on intestinal stem cell aging in Drosophila. Mech. Ageing Dev. 2018, 173, 50–60. [Google Scholar] [CrossRef]
- Wood, J.G.; Hillenmeyer, S.; Lawrence, C.; Chang, C.; Hosier, S.; Lightfoot, W.; Mukherjee, E.; Jiang, N.; Schorl, C.; Brodsky, A.S.; et al. Chromatin remodeling in the aging genome of Drosophila. Aging Cell 2010, 9, 971–978. [Google Scholar] [CrossRef] [PubMed]
- Li, C.-L.; Pu, M.; Wang, W.; Chaturbedi, A.; Emerson, F.J.; Lee, S.S. Region-specific H3K9me3 gain in aged somatic tissues in Caenorhabditis elegans. PLoS Genet. 2021, 17, e1009432. [Google Scholar] [CrossRef]
- Djeghloul, D.; Kuranda, K.; Kuzniak, I.; Barbieri, D.; Naguibneva, I.; Choisy, C.; Bories, J.-C.; Dosquet, C.; Pla, M.; Vanneaux, V.; et al. Age-Associated Decrease of the Histone Methyltransferase SUV39H1 in HSC Perturbs Heterochromatin and B Lymphoid Differentiation. Stem Cell Rep. 2016, 6, 970–984. [Google Scholar] [CrossRef] [PubMed]
- Verdone, L.; Agricola, E.; Caserta, M.; Di Mauro, E. Histone acetylation in gene regulation. Brief. Funct. Genom. Proteomic 2006, 5, 209–221. [Google Scholar] [CrossRef] [PubMed]
- Bannister, A.J.; Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 2011, 21, 381–395. [Google Scholar] [CrossRef]
- Kawakami, K.; Nakamura, A.; Ishigami, A.; Goto, S.; Takahashi, R. Age-related difference of site-specific histone modifications in rat liver. Biogerontology 2009, 10, 415–421. [Google Scholar] [CrossRef]
- Klein, H.-U.; McCabe, C.; Gjoneska, E.; Sullivan, S.E.; Kaskow, B.J.; Tang, A.; Smith, R.V.; Xu, J.; Pfenning, A.R.; Bernstein, B.E.; et al. Epigenome-wide study uncovers large-scale changes in histone acetylation driven by tau pathology in aging and Alzheimer’s human brains. Nat. Neurosci. 2019, 22, 37–46. [Google Scholar] [CrossRef]
- Dang, W.; Steffen, K.K.; Perry, R.; Dorsey, J.A.; Johnson, F.B.; Shilatifard, A.; Kaeberlein, M.; Kennedy, B.K.; Berger, S.L. Histone H4 lysine 16 acetylation regulates cellular lifespan. Nature 2009, 459, 802–807. [Google Scholar] [CrossRef]
- Nativio, R.; Donahue, G.; Berson, A.; Lan, Y.; Amlie-Wolf, A.; Tuzer, F.; Toledo, J.B.; Gosai, S.J.; Gregory, B.D.; Torres, C.; et al. Dysregulation of the epigenetic landscape of normal aging in Alzheimer’s disease. Nat. Neurosci. 2018, 21, 497–505. [Google Scholar] [CrossRef]
- Yuan, J.; Pu, M.; Zhang, Z.; Lou, Z. Histone H3-K56 acetylation is important for genomic stability in mammals. Cell Cycle 2009, 8, 1747–1753. [Google Scholar] [CrossRef]
- Li, Q.; Zhou, H.; Wurtele, H.; Davies, B.; Horazdovsky, B.; Verreault, A.; Zhang, Z. Acetylation of histone H3 lysine 56 regulates replication-coupled nucleosome assembly. Cell 2008, 134, 244–255. [Google Scholar] [CrossRef] [PubMed]
- Feser, J.; Truong, D.; Das, C.; Carson, J.J.; Kieft, J.; Harkness, T.; Tyler, J.K. Elevated histone expression promotes life span extension. Mol. Cell 2010, 39, 724–735. [Google Scholar] [CrossRef] [PubMed]
- Soto-Palma, C.; Niedernhofer, L.J.; Faulk, C.D.; Dong, X. Epigenetics, DNA damage, and aging. J. Clin. 2022, 132, e158446. [Google Scholar] [CrossRef] [PubMed]
- Longo, V.D.; Kennedy, B.K. Sirtuins in aging and age-related disease. Cell 2006, 126, 257–268. [Google Scholar] [CrossRef]
- Lee, S.-H.; Lee, J.-H.; Lee, H.-Y.; Min, K.-J. Sirtuin signaling in cellular senescence and aging. BMB Rep. 2019, 52, 24–34. [Google Scholar] [CrossRef]
- Mostoslavsky, R.; Chua, K.F.; Lombard, D.B.; Pang, W.W.; Fischer, M.R.; Gellon, L.; Liu, P.; Mostoslavsky, G.; Franco, S.; Murphy, M.M.; et al. Genomic instability and aging-like phenotype in the absence of mammalian sirt6. Cell 2006, 124, 315–329. [Google Scholar] [CrossRef]
- Chang, A.R.; Ferrer, C.M.; Mostoslavsky, R. Sirt6, a mammalian deacylase with multitasking abilities. Physiol. Rev. 2020, 100, 145–169. [Google Scholar] [CrossRef]
- Kanfi, Y.; Naiman, S.; Amir, G.; Peshti, V.; Zinman, G.; Nahum, L.; Bar-Joseph, Z.; Cohen, H.Y. The sirtuin SIRT6 regulates lifespan in male mice. Nature 2012, 483, 218–221. [Google Scholar] [CrossRef]
- Roichman, A.; Elhanati, S.; Aon, M.A.; Abramovich, I.; Di Francesco, A.; Shahar, Y.; Avivi, M.Y.; Shurgi, M.; Rubinstein, A.; Wiesner, Y.; et al. Restoration of energy homeostasis by SIRT6 extends healthy lifespan. Nat. Commun. 2021, 12, 3208. [Google Scholar] [CrossRef]
- Moazed, D. Enzymatic activities of Sir2 and chromatin silencing. Curr. Opin. Cell Biol. 2001, 13, 232–238. [Google Scholar] [CrossRef]
- Kaeberlein, M.; McVey, M.; Guarente, L. The SIR2/3/4 complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms. Genes Dev. 1999, 13, 2570–2580. [Google Scholar] [CrossRef] [PubMed]
- Guarente, L. Sirtuins, aging, and medicine. N. Engl. J. Med 2011, 364, 2235–2244. [Google Scholar] [CrossRef] [PubMed]
- Haigis, M.C.; Sinclair, D.A. Mammalian sirtuins: Biological insights and disease relevance. Annu. Rev. Pathol. 2010, 5, 253–295. [Google Scholar] [CrossRef] [PubMed]
- Tissenbaum, H.A.; Guarente, L. Increased dosage of a sir-2 gene extends lifespan in Caenorhabditis elegans. Nature 2001, 410, 227–230. [Google Scholar] [CrossRef] [PubMed]
- Rogina, B.; Helfand, S.L. Sir2 mediates longevity in the fly through a pathway related to calorie restriction. Proc. Natl. Acad. Sci. USA 2004, 101, 15998–16003. [Google Scholar] [CrossRef] [PubMed]
- Satoh, A.; Brace, C.S.; Rensing, N.; Cliften, P.; Wozniak, D.F.; Herzog, E.D.; Yamada, K.A.; Imai, S.-I. Sirt1 extends life span and delays aging in mice through the regulation of Nk2 homeobox 1 in the DMH and LH. Cell Metab. 2013, 18, 416–430. [Google Scholar] [CrossRef]
- Kimura, A.; Umehara, T.; Horikoshi, M. Chromosomal gradient of histone acetylation established by Sas2p and Sir2p functions as a shield against gene silencing. Nat. Genet. 2002, 32, 370–377. [Google Scholar] [CrossRef]
- Suka, N.; Luo, K.; Grunstein, M. Sir2p and Sas2p opposingly regulate acetylation of yeast histone H4 lysine16 and spreading of heterochromatin. Nat. Genet. 2002, 32, 378–383. [Google Scholar] [CrossRef]
- Zhang, M.; Poplawski, M.; Yen, K.; Cheng, H.; Bloss, E.; Zhu, X.; Patel, H.; Mobbs, C.V. Role of CBP and SATB-1 in aging, dietary restriction, and insulin-like signaling. PLoS Biol. 2009, 7, e1000245. [Google Scholar] [CrossRef]
- Kirfel, P.; Vilcinskas, A.; Skaljac, M. Lysine Acetyltransferase p300/CBP Plays an Important Role in Reproduction, Embryogenesis and Longevity of the Pea Aphid Acyrthosiphon pisum. Insects 2020, 11, 265. [Google Scholar] [CrossRef]
- Huang, B.; Zhong, D.; Zhu, J.; An, Y.; Gao, M.; Zhu, S.; Dang, W.; Wang, X.; Yang, B.; Xie, Z. Inhibition of histone acetyltransferase GCN5 extends lifespan in both yeast and human cell lines. Aging Cell 2020, 19, e13129. [Google Scholar] [CrossRef]
- Josefowicz, S.Z.; Shimada, M.; Armache, A.; Li, C.H.; Miller, R.M.; Lin, S.; Yang, A.; Dill, B.D.; Molina, H.; Park, H.-S.; et al. Chromatin kinases act on transcription factors and histone tails in regulation of inducible transcription. Mol. Cell 2016, 64, 347–361. [Google Scholar] [CrossRef] [PubMed]
- Rossetto, D.; Avvakumov, N.; Côté, J. Histone phosphorylation: A chromatin modification involved in diverse nuclear events. Epigenetics 2012, 7, 1098–1108. [Google Scholar] [CrossRef] [PubMed]
- Oh, S.; Suganuma, T.; Gogol, M.M.; Workman, J.L. Histone H3 threonine 11 phosphorylation by Sch9 and CK2 regulates chronological lifespan by controlling the nutritional stress response. eLife 2018, 7, e36157. [Google Scholar] [CrossRef] [PubMed]
- Joos, J.P.; Saadatmand, A.R.; Schnabel, C.; Viktorinová, I.; Brand, T.; Kramer, M.; Nattel, S.; Dobrev, D.; Tomancak, P.; Backs, J.; et al. Ectopic expression of S28A-mutated Histone H3 modulates longevity, stress resistance and cardiac function in Drosophila. Sci. Rep. 2018, 8, 2940. [Google Scholar] [CrossRef] [PubMed]
- Rhie, B.-H.; Song, Y.-H.; Ryu, H.-Y.; Ahn, S.H. Cellular aging is associated with increased ubiquitylaiton of histone H2B in yeast telomeric heterochromatin. Biochem. Biophys. Res. Commun. 2013, 439, 570–575. [Google Scholar] [CrossRef]
- McCormick, M.A.; Mason, A.G.; Guyenet, S.J.; Dang, W.; Garza, R.M.; Ting, M.K.; Moller, R.M.; Berger, S.L.; Kaeberlein, M.; Pillus, L.; et al. The SAGA histone deubiquitinase module controls yeast replicative lifespan via Sir2 interaction. Cell Rep. 2014, 8, 477–486. [Google Scholar] [CrossRef]
- Guo, Y.; Chomiak, A.A.; Hong, Y.; Lowe, C.C.; Kopsidas, C.A.; Chan, W.-C.; Andrade, J.; Pan, H.; Zhou, X.; Monuki, E.S.; et al. Histone H2A ubiquitination resulting from Brap loss of function connects multiple aging hallmarks and accelerates neurodegeneration. iScience 2022, 25, 104519. [Google Scholar] [CrossRef]
- Martire, S.; Banaszynski, L.A. The roles of histone variants in fine-tuning chromatin organization and function. Nat. Rev. Mol. Cell Biol. 2020, 21, 522–541. [Google Scholar] [CrossRef]
- Kreiling, J.A.; Tamamori-Adachi, M.; Sexton, A.N.; Jeyapalan, J.C.; Munoz-Najar, U.; Peterson, A.L.; Manivannan, J.; Rogers, E.S.; Pchelintsev, N.A.; Adams, P.D.; et al. Age-associated increase in heterochromatic marks in murine and primate tissues: Age-associated increase in heterochromatic marks. Aging Cell 2011, 10, 292–304. [Google Scholar] [CrossRef]
- Tvardovskiy, A.; Schwämmle, V.; Kempf, S.J.; Rogowska-Wrzesinska, A.; Jensen, O.N. Accumulation of histone variant H3.3 with age is associated with profound changes in the histone methylation landscape. Nucleic Acids Res. 2017, 45, 9272–9289. [Google Scholar] [CrossRef] [PubMed]
- Maze, I.; Wenderski, W.; Noh, K.-M.; Bagot, R.C.; Tzavaras, N.; Purushothaman, I.; Elsässer, S.J.; Guo, Y.; Ionete, C.; Hurd, Y.L.; et al. Critical role of histone turnover in neuronal transcription and plasticity. Neuron 2015, 87, 77–94. [Google Scholar] [CrossRef]
- Piazzesi, A.; Papić, D.; Bertan, F.; Salomoni, P.; Nicotera, P.; Bano, D. Replication-independent histone variant h3.3 controls animal lifespan through the regulation of pro-longevity transcriptional programs. Cell Rep. 2016, 17, 987–996. [Google Scholar] [CrossRef] [PubMed]
- Rogakou, E.P.; Sekeri-Pataryas, K.E. Histone variants of H2A and H3 families are regulated during in vitro aging in the same manner as during differentiation. Exp. Gerontol. 1999, 34, 741–754. [Google Scholar] [CrossRef]
- Contrepois, K.; Coudereau, C.; Benayoun, B.A.; Schuler, N.; Roux, P.-F.; Bischof, O.; Courbeyrette, R.; Carvalho, C.; Thuret, J.-Y.; Ma, Z.; et al. Histone variant H2A.J accumulates in senescent cells and promotes inflammatory gene expression. Nat. Commun. 2017, 8, 14995. [Google Scholar] [CrossRef]
- Robertson, K.D. DNA methylation and human disease. Nat. Rev. Genet. 2005, 6, 597–610. [Google Scholar] [CrossRef] [PubMed]
- Gonzalo, S.; Jaco, I.; Fraga, M.F.; Chen, T.; Li, E.; Esteller, M.; Blasco, M.A. DNA methyltransferases control telomere length and telomere recombination in mammalian cells. Nat. Cell Biol. 2006, 8, 416–424. [Google Scholar] [CrossRef]
- Xu, G.L.; Bestor, T.H.; Bourc’his, D.; Hsieh, C.L.; Tommerup, N.; Bugge, M.; Hulten, M.; Qu, X.; Russo, J.J.; Viegas-Péquignot, E. Chromosome instability and immunodeficiency syndrome caused by mutations in a DNA methyltransferase gene. Nature 1999, 402, 187–191. [Google Scholar] [CrossRef]
- Okano, M.; Bell, D.W.; Haber, D.A.; Li, E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 1999, 99, 247–257. [Google Scholar] [CrossRef]
- Lea, A.J.; Vockley, C.M.; Johnston, R.A.; Del Carpio, C.A.; Barreiro, L.B.; Reddy, T.E.; Tung, J. Genome-wide quantification of the effects of DNA methylation on human gene regulation. eLife 2018, 7, e37513. [Google Scholar] [CrossRef]
- Ahuja, N.; Li, Q.; Mohan, A.L.; Baylin, S.B.; Issa, J.P. Aging and DNA methylation in colorectal mucosa and cancer. Cancer Res. 1998, 58, 5489–5494. [Google Scholar] [PubMed]
- Rakyan, V.K.; Down, T.A.; Maslau, S.; Andrew, T.; Yang, T.-P.; Beyan, H.; Whittaker, P.; McCann, O.T.; Finer, S.; Valdes, A.M.; et al. Human aging-associated DNA hypermethylation occurs preferentially at bivalent chromatin domains. Genome Res. 2010, 20, 434–439. [Google Scholar] [CrossRef]
- Teschendorff, A.E.; Menon, U.; Gentry-Maharaj, A.; Ramus, S.J.; Weisenberger, D.J.; Shen, H.; Campan, M.; Noushmehr, H.; Bell, C.G.; Maxwell, A.P.; et al. Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res. 2010, 20, 440–446. [Google Scholar] [CrossRef] [PubMed]
- Hernandez, D.G.; Nalls, M.A.; Gibbs, J.R.; Arepalli, S.; van der Brug, M.; Chong, S.; Moore, M.; Longo, D.L.; Cookson, M.R.; Traynor, B.J.; et al. Distinct DNA methylation changes highly correlated with chronological age in the human brain. Hum. Mol. Genet. 2011, 20, 1164–1172. [Google Scholar] [CrossRef] [PubMed]
- Bell, J.T.; Tsai, P.-C.; Yang, T.-P.; Pidsley, R.; Nisbet, J.; Glass, D.; Mangino, M.; Zhai, G.; Zhang, F.; Valdes, A.; et al. Epigenome-wide scans identify differentially methylated regions for age and age-related phenotypes in a healthy ageing population. PLoS Genet. 2012, 8, e1002629. [Google Scholar] [CrossRef]
- Christensen, B.C.; Houseman, E.A.; Marsit, C.J.; Zheng, S.; Wrensch, M.R.; Wiemels, J.L.; Nelson, H.H.; Karagas, M.R.; Padbury, J.F.; Bueno, R.; et al. Aging and environmental exposures alter tissue-specific DNA methylation dependent upon CpG island context. PLoS Genet. 2009, 5, e1000602. [Google Scholar] [CrossRef]
- Maegawa, S.; Hinkal, G.; Kim, H.S.; Shen, L.; Zhang, L.; Zhang, J.; Zhang, N.; Liang, S.; Donehower, L.A.; Issa, J.-P.J. Widespread and tissue specific age-related DNA methylation changes in mice. Genome Res. 2010, 20, 332–340. [Google Scholar] [CrossRef]
- Horvath, S.; Zhang, Y.; Langfelder, P.; Kahn, R.S.; Boks, M.P.M.; van Eijk, K.; van den Berg, L.H.; Ophoff, R.A. Aging effects on DNA methylation modules in human brain and blood tissue. Genome Biol. 2012, 13, R97. [Google Scholar] [CrossRef]
- Bollati, V.; Schwartz, J.; Wright, R.; Litonjua, A.; Tarantini, L.; Suh, H.; Sparrow, D.; Vokonas, P.; Baccarelli, A. Decline in genomic DNA methylation through aging in a cohort of elderly subjects. Mech. Ageing Dev. 2009, 130, 234–239. [Google Scholar] [CrossRef]
- Zampieri, M.; Ciccarone, F.; Calabrese, R.; Franceschi, C.; Bürkle, A.; Caiafa, P. Reconfiguration of DNA methylation in aging. Mech Ageing Dev. 2015, 151, 60–70. [Google Scholar] [CrossRef]
- Horvath, S.; Raj, K. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat. Rev. Genet. 2018, 19, 371–384. [Google Scholar] [CrossRef]
- Hernando-Herraez, I.; Evano, B.; Stubbs, T.; Commere, P.H.; Bonder, M.J.; Clark, S.; Andrews, S.; Tajbakhsh, S.; Reik, W. Ageing affects DNA methylation drift and transcriptional cell-to-cell variability in mouse muscle stem cells. Nat. Commun. 2019, 10, 4361. [Google Scholar] [CrossRef] [PubMed]
- Bergsma, T.; Rogaeva, E. DNA methylation clocks and their predictive capacity for aging phenotypes and healthspan. Neurosci. Insights 2020, 15, 2633105520942221. [Google Scholar] [CrossRef] [PubMed]
- Casillas, M.A., Jr.; Lopatina, N.; Andrews, L.G.; Tollefsbol, T.O. Transcriptional control of the DNA methyltransferases is altered in aging and neoplastically-transformed human fibroblasts. Mol. Cell. Biochem. 2003, 252, 33–43. [Google Scholar] [CrossRef] [PubMed]
- Zhou, D.; Robertson, K.D. Role of DNA methylation in genome stability. In Genome Stability; Elsevier: Amsterdam, The Netherlands, 2016; pp. 409–424. ISBN 9780128033098. [Google Scholar]
- Gorbunova, V.; Seluanov, A.; Mita, P.; McKerrow, W.; Fenyö, D.; Boeke, J.D.; Linker, S.B.; Gage, F.H.; Kreiling, J.A.; Petrashen, A.P.; et al. The role of retrotransposable elements in ageing and age-associated diseases. Nature 2021, 596, 43–53. [Google Scholar] [CrossRef]
- Bysani, M.; Perfilyev, A.; De Mello, V.D.; Rönn, T.; Nilsson, E.; Pihlajamäki, J.; Ling, C. Epigenetic alterations in blood mirror age-associated DNA methylation and gene expression changes in human liver. Epigenomics 2017, 9, 105–122. [Google Scholar] [CrossRef]
- Garagnani, P.; Bacalini, M.G.; Pirazzini, C.; Gori, D.; Giuliani, C.; Mari, D.; Di Blasio, A.M.; Gentilini, D.; Vitale, G.; Collino, S.; et al. Methylation of ELOVL 2 gene as a new epigenetic marker of age. Aging Cell 2012, 11, 1132–1134. [Google Scholar] [CrossRef]
- Steegenga, W.T.; Boekschoten, M.V.; Lute, C.; Hooiveld, G.J.; De Groot, P.J.; Morris, T.J.; Teschendorff, A.E.; Butcher, L.M.; Beck, S.; Müller, M. Genome-wide age-related changes in DNA methylation and gene expression in human PBMCs. Age 2014, 36, 1523–1540. [Google Scholar] [CrossRef]
- Horvath, S. DNA methylation age of human tissues and cell types. Genome Biol. 2013, 14, R115. [Google Scholar] [CrossRef]
- Chen, L.; Ganz, P.A.; Sehl, M.E. DNA Methylation, Aging, and Cancer Risk: A Mini-Review. Front. Bioinform. 2022, 2, 847629. [Google Scholar] [CrossRef]
- Heyn, H.; Moran, S.; Esteller, M. Aberrant DNA methylation profiles in the premature aging disorders Hutchinson-Gilford Progeria and Werner syndrome. Epigenetics 2013, 8, 28–33. [Google Scholar] [CrossRef] [PubMed]
- Zhao, M.; Qin, J.; Yin, H.; Tan, Y.; Liao, W.; Liu, Q.; Luo, S.; He, M.; Liang, G.; Shi, Y.; et al. Distinct epigenomes in CD4+ T cells of newborns, middle-ages and centenarians. Sci. Rep. 2016, 6, 38411. [Google Scholar] [CrossRef]
- Tserel, L.; Kolde, R.; Limbach, M.; Tretyakov, K.; Kasela, S.; Kisand, K.; Saare, M.; Vilo, J.; Metspalu, A.; Milani, L.; et al. Age-related profiling of DNA methylation in CD8+ T cells reveals changes in immune response and transcriptional regulator genes. Sci. Rep. 2015, 5, 13107. [Google Scholar] [CrossRef]
- Talens, R.P.; Christensen, K.; Putter, H.; Willemsen, G.; Christiansen, L.; Kremer, D.; Suchiman, H.E.D.; Slagboom, P.E.; Boomsma, D.I.; Heijmans, B.T. Epigenetic variation during the adult lifespan: Cross-sectional and longitudinal data on monozygotic twin pairs. Aging Cell 2012, 11, 694–703. [Google Scholar] [CrossRef] [PubMed]
- McStay, B.; Grummt, I. The epigenetics of rRNA genes: From molecular to chromosome biology. Annu. Rev. Cell Dev. Biol. 2008, 24, 131–157. [Google Scholar] [CrossRef]
- Saka, K.; Ide, S.; Ganley, A.R.D.; Kobayashi, T. Cellular senescence in yeast is regulated by rDNA noncoding transcription. Curr. Biol. 2013, 23, 1794–1798. [Google Scholar] [CrossRef] [PubMed]
- Steffen, K.K.; MacKay, V.L.; Kerr, E.O.; Tsuchiya, M.; Hu, D.; Fox, L.A.; Dang, N.; Johnston, E.D.; Oakes, J.A.; Tchao, B.N.; et al. Yeast life span extension by depletion of 60s ribosomal subunits is mediated by Gcn4. Cell 2008, 133, 292–302. [Google Scholar] [CrossRef]
- Swisshelm, K.; Disteche, C.M.; Thorvaldsen, J.; Nelson, A.; Salk, D. Age-related increase in methylation of ribosomal genes and inactivation of chromosome-specific rRNA gene clusters in mouse. Mutat. Res. 1990, 237, 131–146. [Google Scholar] [CrossRef] [PubMed]
- Oakes, C.C.; Smiraglia, D.J.; Plass, C.; Trasler, J.M.; Robaire, B. Aging results in hypermethylation of ribosomal DNA in sperm and liver of male rats. Proc. Natl. Acad. Sci. USA 2003, 100, 1775–1780. [Google Scholar] [CrossRef]
- D’Aquila, P.; Montesanto, A.; Mandalà, M.; Garasto, S.; Mari, V.; Corsonello, A.; Bellizzi, D.; Passarino, G. Methylation of the ribosomal RNA gene promoter is associated with aging and age-related decline. Aging Cell 2017, 16, 966–975. [Google Scholar] [CrossRef]
- Gensous, N.; Ravaioli, F.; Pirazzini, C.; Gramignoli, R.; Ellis, E.; Storci, G.; Capri, M.; Strom, S.; Laconi, E.; Franceschi, C.; et al. Aging and caloric restriction modulate the DNA methylation profile of the ribosomal RNA locus in human and rat liver. Nutrients 2020, 12, 277. [Google Scholar] [CrossRef]
- Wang, M.; Lemos, B. Ribosomal DNA harbors an evolutionarily conserved clock of biological aging. Genome Res. 2019, 29, 325–333. [Google Scholar] [CrossRef] [PubMed]
- Masser, D.R.; Hadad, N.; Porter, H.L.; Mangold, C.A.; Unnikrishnan, A.; Ford, M.M.; Giles, C.B.; Georgescu, C.; Dozmorov, M.G.; Wren, J.D.; et al. Sexually divergent DNA methylation patterns with hippocampal aging. Aging Cell 2017, 16, 1342–1352. [Google Scholar] [CrossRef]
- Cech, T.R.; Steitz, J.A. The noncoding RNA revolution—Trashing old rules to forge new ones. Cell 2014, 157, 77–94. [Google Scholar] [CrossRef] [PubMed]
- Statello, L.; Guo, C.-J.; Chen, L.-L.; Huarte, M. Gene regulation by long non-coding RNAs and its biological functions. Nat. Rev. Mol. Cell Biol. 2021, 22, 96–118. [Google Scholar] [CrossRef] [PubMed]
- Bushati, N.; Cohen, S.M. microRNA functions. Annu. Rev. Cell Dev. Biol. 2007, 23, 175–205. [Google Scholar] [CrossRef]
- Alles, J.; Fehlmann, T.; Fischer, U.; Backes, C.; Galata, V.; Minet, M.; Hart, M.; Abu-Halima, M.; Grässer, F.A.; Lenhof, H.-P.; et al. An estimate of the total number of true human miRNAs. Nucleic Acids Res. 2019, 47, 3353–3364. [Google Scholar] [CrossRef]
- Kinser, H.E.; Pincus, Z. MicroRNAs as modulators of longevity and the aging process. Hum Genet. 2020, 139, 291–308. [Google Scholar] [CrossRef]
- Smith-Vikos, T.; Slack, F.J. MicroRNAs and their roles in aging. J. Cell Sci. 2012, 125, 7–17. [Google Scholar] [CrossRef] [PubMed]
- Boehm, M.; Slack, F. A developmental timing microRNA and its target regulate life span in C. elegans. Science 2005, 310, 1954–1957. [Google Scholar] [CrossRef]
- De Lencastre, A.; Pincus, Z.; Zhou, K.; Kato, M.; Lee, S.S.; Slack, F.J. MicroRNAs both promote and antagonize longevity in C. elegans. Curr. Biol. 2010, 20, 2159–2168. [Google Scholar] [CrossRef]
- Pincus, Z.; Smith-Vikos, T.; Slack, F.J. MicroRNA predictors of longevity in Caenorhabditis elegans. PLoS Genet. 2011, 7, e1002306. [Google Scholar] [CrossRef]
- Smith-Vikos, T.; de Lencastre, A.; Inukai, S.; Shlomchik, M.; Holtrup, B.; Slack, F.J. MicroRNAs mediate dietary-restriction-induced longevity through PHA-4/FOXA and SKN-1/Nrf transcription factors. Curr. Biol. 2014, 24, 2238–2246. [Google Scholar] [CrossRef] [PubMed]
- Chawla, G.; Deosthale, P.; Childress, S.; Wu, Y.-C.; Sokol, N.S. A let-7-to-miR-125 MicroRNA Switch Regulates Neuronal Integrity and Lifespan in Drosophila. PLoS Genet. 2016, 12, e1006247. [Google Scholar] [CrossRef]
- Gendron, C.M.; Pletcher, S.D. MicroRNAs mir-184 and let-7 alter Drosophila metabolism and longevity. Aging Cell 2017, 16, 1434–1438. [Google Scholar] [CrossRef] [PubMed]
- Liu, N.; Landreh, M.; Cao, K.; Abe, M.; Hendriks, G.-J.; Kennerdell, J.R.; Zhu, Y.; Wang, L.-S.; Bonini, N.M. The microRNA miR-34 modulates ageing and neurodegeneration in Drosophila. Nature 2012, 482, 519–523. [Google Scholar] [CrossRef]
- Verma, P.; Augustine, G.J.; Ammar, M.-R.; Tashiro, A.; Cohen, S.M. A neuroprotective role for microRNA miR-1000 mediated by limiting glutamate excitotoxicity. Nat. Neurosci. 2015, 18, 379–385. [Google Scholar] [CrossRef] [PubMed]
- Du, W.W.; Yang, W.; Fang, L.; Xuan, J.; Li, H.; Khorshidi, A.; Gupta, S.; Li, X.; Yang, B.B. miR-17 extends mouse lifespan by inhibiting senescence signaling mediated by MKP7. Cell Death Dis. 2014, 5, e1355. [Google Scholar] [CrossRef] [PubMed]
- Mori, M.A.; Raghavan, P.; Thomou, T.; Boucher, J.; Robida-Stubbs, S.; Macotela, Y.; Russell, S.J.; Kirkland, J.L.; Blackwell, T.K.; Kahn, C.R. Role of microRNA processing in adipose tissue in stress defense and longevity. Cell Metab. 2012, 16, 336–347. [Google Scholar] [CrossRef]
- Gombar, S.; Jung, H.J.; Dong, F.; Calder, B.; Atzmon, G.; Barzilai, N.; Tian, X.-L.; Pothof, J.; Hoeijmakers, J.H.; Campisi, J.; et al. Comprehensive microRNA profiling in B-cells of human centenarians by massively parallel sequencing. BMC Genom. 2012, 13, 353. [Google Scholar] [CrossRef]
- Serna, E.; Gambini, J.; Borras, C.; Abdelaziz, K.M.; Belenguer, A.; Sanchis, P.; Avellana, J.A.; Rodriguez-Mañas, L.; Viña, J. Centenarians, but not octogenarians, up-regulate the expression of microRNAs. Sci. Rep. 2012, 2, 961. [Google Scholar] [CrossRef] [PubMed]
- Schraml, E.; Grillari, J. From cellular senescence to age-associated diseases: The miRNA connection. Longev. Heal. 2012, 1, 10. [Google Scholar] [CrossRef] [PubMed]
- Huan, T.; Chen, G.; Liu, C.; Bhattacharya, A.; Rong, J.; Chen, B.H.; Seshadri, S.; Tanriverdi, K.; Freedman, J.E.; Larson, M.G.; et al. Age-associated microRNA expression in human peripheral blood is associated with all-cause mortality and age-related traits. Aging Cell 2018, 17, e12687. [Google Scholar] [CrossRef]
- Jin, L.; Song, Q.; Zhang, W.; Geng, B.; Cai, J. Roles of long noncoding RNAs in aging and aging complications. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 1763–1771. [Google Scholar] [CrossRef]
- Kour, S.; Rath, P.C. Long noncoding RNAs in aging and age-related diseases. Ageing Res. Rev. 2016, 26, 1–21. [Google Scholar] [CrossRef]
- He, R.-Z.; Luo, D.-X.; Mo, Y.-Y. Emerging roles of lncRNAs in the post-transcriptional regulation in cancer. Genes Dis. 2019, 6, 6–15. [Google Scholar] [CrossRef] [PubMed]
- Bink, D.I.; Lozano-Vidal, N.; Boon, R.A. Long Non-Coding RNA in Vascular Disease and Aging. Noncoding RNA 2019, 5, 26. [Google Scholar] [CrossRef] [PubMed]
- He, X.; Ou, C.; Xiao, Y.; Han, Q.; Li, H.; Zhou, S. LncRNAs: Key players and novel insights into diabetes mellitus. Oncotarget 2017, 8, 71325–71341. [Google Scholar] [CrossRef]
- Pereira Fernandes, D.; Bitar, M.; Jacobs, F.M.J.; Barry, G. Long Non-Coding RNAs in Neuronal Aging. Noncoding RNA 2018, 4, 12. [Google Scholar] [CrossRef]
- Wood, S.H.; Craig, T.; Li, Y.; Merry, B.; de Magalhães, J.P. Whole transcriptome sequencing of the aging rat brain reveals dynamic RNA changes in the dark matter of the genome. Age 2013, 35, 763–776. [Google Scholar] [CrossRef]
- Marttila, S.; Chatsirisupachai, K.; Palmer, D.; de Magalhães, J.P. Ageing-associated changes in the expression of lncRNAs in human tissues reflect a transcriptional modulation in ageing pathways. Mech. Ageing Dev. 2020, 185, 111177. [Google Scholar] [CrossRef]
- Wagner, A.; Schosserer, M. The epitranscriptome in ageing and stress resistance: A systematic review. Ageing Res. Rev. 2022, 81, 101700. [Google Scholar] [CrossRef] [PubMed]
- McMahon, M.; Forester, C.; Buffenstein, R. Aging through an epitranscriptomic lens. Nat. Aging. 2021, 1, 335–346. [Google Scholar] [CrossRef] [PubMed]
- Schosserer, M.; Minois, N.; Angerer, T.B.; Amring, M.; Dellago, H.; Harreither, E.; Calle-Perez, A.; Pircher, A.; Gerstl, M.P.; Pfeifenberger, S.; et al. Methylation of ribosomal RNA by NSUN5 is a conserved mechanism modulating organismal lifespan. Nat. Commun. 2015, 6, 6158. [Google Scholar] [CrossRef] [PubMed]
- Heissenberger, C.; Rollins, J.A.; Krammer, T.L.; Nagelreiter, F.; Stocker, I.; Wacheul, L.; Shpylovyi, A.; Tav, K.; Snow, S.; Grillari, J.; et al. The ribosomal RNA m5C methyltransferase NSUN-1 modulates healthspan and oogenesis in Caenorhabditis elegans. eLife 2020, 9, e56205. [Google Scholar] [CrossRef]
- Fabrizio, P.; Hoon, S.; Shamalnasab, M.; Galbani, A.; Wei, M.; Giaever, G.; Nislow, C.; Longo, V.D. Genome-wide screen in Saccharomyces cerevisiae identifies vacuolar protein sorting, autophagy, biosynthetic, and tRNA methylation genes involved in life span regulation. PLoS Genet. 2010, 6, e001024. [Google Scholar] [CrossRef] [PubMed]
- Endres, L.; Begley, U.; Clark, R.; Gu, C.; Dziergowska, A.; Małkiewicz, A.; Melendez, J.A.; Dedon, P.C.; Begley, T.J. Alkbh8 regulates selenocysteine-protein expression to protect against reactive oxygen species damage. PLoS ONE 2015, 10, e0131335. [Google Scholar] [CrossRef]
- Kan, L.; Ott, S.; Joseph, B.; Park, E.S.; Dai, W.; Kleiner, R.E.; Claridge-Chang, A.; Lai, E.C. A neural m6A/Ythdf pathway is required for learning and memory in Drosophila. Nat. Commun. 2021, 12, 1458. [Google Scholar] [CrossRef]
- Ozkurede, U.; Kala, R.; Johnson, C.; Shen, Z.; Miller, R.A.; Garcia, G.G. Cap-independent mRNA translation is upregulated in long-lived endocrine mutant mice. J. Mol. Endocrinol. 2019, 63, 123–138. [Google Scholar] [CrossRef]
- Shen, Z.; Hinson, A.; Miller, R.A.; Garcia, G.G. Cap-independent translation: A shared mechanism for lifespan extension by rapamycin, acarbose, and 17α-estradiol. Aging Cell 2021, 20, e13345. [Google Scholar] [CrossRef]
- Allshire, R.C.; Madhani, H.D. Ten principles of heterochromatin formation and function. Nat. Rev. Mol. Cell Biol. 2018, 19, 229–244. [Google Scholar] [CrossRef] [PubMed]
- Termolino, P.; Cremona, G.; Consiglio, M.F.; Conicella, C. Insights into epigenetic landscape of recombination-free regions. Chromosoma 2016, 125, 301–308. [Google Scholar] [CrossRef] [PubMed]
- Tsurumi, A.; Li, W.X. Global heterochromatin loss: A unifying theory of aging? Epigenetics 2012, 7, 680–688. [Google Scholar] [CrossRef] [PubMed]
- Haithcock, E.; Dayani, Y.; Neufeld, E.; Zahand, A.J.; Feinstein, N.; Mattout, A.; Gruenbaum, Y.; Liu, J. Age-related changes of nuclear architecture in Caenorhabditis elegans. Proc. Natl. Acad. Sci. USA 2005, 102, 16690–16695. [Google Scholar] [CrossRef]
- Larson, K.; Yan, S.J.; Tsurumi, A.; Liu, J.; Zhou, J.; Gaur, K.; Guo, D.; Eickbush, T.H.; Li, W.X. Heterochromatin formation promotes longevity and represses ribosomal RNA synthesis. PLoS Genet. 2012, 8, e1002473. [Google Scholar] [CrossRef]
- Lee, J.H.; Kim, E.W.; Croteau, D.L.; Bohr, V.A. Heterochromatin: An epigenetic point of view in aging. Exp. Mol. Med. 2020, 52, 1466–1474. [Google Scholar] [CrossRef] [PubMed]
- Smeal, T.; Claus, J.; Kennedy, B.; Cole, F.; Guarente, L. Loss of transcriptional silencing causes sterility in old mother cells of S. cerevisiae. Cell 1996, 84, 633–642. [Google Scholar] [CrossRef] [PubMed]
- Narita, M.; Nũnez, S.; Heard, E.; Narita, M.; Lin, A.W.; Hearn, S.A.; Spector, D.L.; Hannon, G.J.; Lowe, S.W. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell 2003, 113, 703–716. [Google Scholar] [CrossRef] [PubMed]
- Scaffidi, P.; Misteli, T. Lamin A-dependent nuclear defects in human aging. Science 2006, 312, 1059–1063. [Google Scholar] [CrossRef]
- Pegoraro, G.; Kubben, N.; Wickert, U.; Göhler, H.; Hoffmann, K.; Misteli, T. Ageing-related chromatin defects through loss of the NURD complex. Nat. Cell Biol. 2009, 11, 1261–1267. [Google Scholar] [CrossRef]
- Zhang, W.; Li, J.; Suzuki, K.; Qu, J.; Wang, P.; Zhou, J.; Liu, X.; Ren, R.; Xu, X.; Ocampo, A.; et al. Aging stem cells. A Werner syndrome stem cell model unveils heterochromatin alterations as a driver of human aging. Science 2015, 348, 1160–1163. [Google Scholar] [CrossRef]
- Della Valle, F.; Reddy, P.; Yamamoto, M.; Liu, P.; Saera-Vila, A.; Bensaddek, D.; Zhang, H.; Prieto Martinez, J.; Abassi, L.; Celii, M.; 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, eabl6057. [Google Scholar] [CrossRef]
- Citterio, E.; Van Den Boom, V.; Schnitzler, G.; Kanaar, R.; Bonte, E.; Kingston, R.E.; Hoeijmakers, J.H.; Vermeulen, W. ATP-dependent chromatin remodeling by the Cockayne syndrome B DNA repair-transcription-coupling factor. Mol. Cell. Biol. 2000, 20, 7643–7653. [Google Scholar] [CrossRef]
- Lee, J.-H.; Demarest, T.G.; Babbar, M.; Kim, E.W.; Okur, M.N.; De, S.; Croteau, D.L.; Bohr, V.A. Cockayne syndrome group B deficiency reduces H3K9me3 chromatin remodeler SETDB1 and exacerbates cellular aging. Nucleic Acids Res. 2019, 47, 8548–8562. [Google Scholar] [CrossRef]
- Benayoun, B.A.; Pollina, E.A.; Singh, P.P.; Mahmoudi, S.; Harel, I.; Casey, K.M.; Dulken, B.W.; Kundaje, A.; Brunet, A. Remodeling of epigenome and transcriptome landscapes with aging in mice reveals widespread induction of inflammatory responses. Genome Res. 2019, 29, 697–709. [Google Scholar] [CrossRef]
- Edwards, M.G.; Sarkar, D.; Klopp, R.; Morrow, J.D.; Weindruch, R.; Prolla, T.A. Age-related impairment of the transcriptional responses to oxidative stress in the mouse heart. Physiol. Genom. 2003, 13, 119–127. [Google Scholar] [CrossRef]
- Dutta, N.; Garcia, G.; Higuchi-Sanabria, R. Hijacking cellular stress responses to promote lifespan. Front. Aging 2022, 3, 860404. [Google Scholar] [CrossRef]
- Pascual-Ahuir, A.; González-Cantó, E.; Juyoux, P.; Pable, J.; Poveda-Huertes, D.; Saiz-Balbastre, S.; Squeo, S.; Ureña-Marco, A.; Vanacloig-Pedros, E.; Zaragoza-Infante, L.; et al. Dose dependent gene expression is dynamically modulated by the history, physiology and age of yeast cells. Biochim. Biophys. Acta Gene Regul. Mech. 2019, 1862, 457–471. [Google Scholar] [CrossRef]
- Wood, J.G.; Helfand, S.L. Chromatin structure and transposable elements in organismal aging. Front. Genet. 2013, 4, 274. [Google Scholar] [CrossRef]
- Gorbunova, V.; Boeke, J.D.; Helfand, S.L.; Sedivy, J.M. Human Genomics. Sleeping dogs of the genome. Science 2014, 346, 1187–1188. [Google Scholar] [CrossRef]
- Becker, P.B.; Workman, J.L. Nucleosome remodeling and epigenetics. Cold Spring Harb. Perspect. Biol. 2013, 5, a017905. [Google Scholar] [CrossRef]
- Clapier, C.R.; Iwasa, J.; Cairns, B.R.; Peterson, C.L. Mechanisms of action and regulation of ATP-dependent chromatin-remodelling complexes. Nat. Rev. Mol. Cell Biol. 2017, 18, 407–422. [Google Scholar] [CrossRef] [PubMed]
- Dang, W.; Sutphin, G.L.; Dorsey, J.A.; Otte, G.L.; Cao, K.; Perry, R.M.; Wanat, J.J.; Saviolaki, D.; Murakami, C.J.; Tsuchiyama, S.; et al. Inactivation of yeast Isw2 chromatin remodeling enzyme mimics longevity effect of calorie restriction via induction of genotoxic stress response. Cell Metab. 2014, 19, 952–966. [Google Scholar] [CrossRef] [PubMed]
- De Vaux, V.; Pfefferli, C.; Passannante, M.; Belhaj, K.; von Essen, A.; Sprecher, S.G.; Müller, F.; Wicky, C. The Caenorhabditis elegans LET-418/Mi2 plays a conserved role in lifespan regulation. Aging Cell 2013, 12, 1012–1020. [Google Scholar] [CrossRef] [PubMed]
- Riedel, C.G.; Dowen, R.H.; Lourenco, G.F.; Kirienko, N.V.; Heimbucher, T.; West, J.A.; Bowman, S.K.; Kingston, R.E.; Dillin, A.; Asara, J.M.; et al. DAF-16 employs the chromatin remodeller SWI/SNF to promote stress resistance and longevity. Nat. Cell Biol. 2013, 15, 491–501. [Google Scholar] [CrossRef]
- Oliviero, G.; Kovalchuk, S.; Rogowska-Wrzesinska, A.; Schwämmle, V.; Jensen, O.N. Distinct and diverse chromatin proteomes of ageing mouse organs reveal protein signatures that correlate with physiological functions. eLife 2022, 11, e73524. [Google Scholar] [CrossRef]
- Hu, Z.; Chen, K.; Xia, Z.; Chavez, M.; Pal, S.; Seol, J.H.; Chen, C.C.; Li, W.; Tyler, J.K. Nucleosome loss leads to global transcriptional up-regulation and genomic instability during yeast aging. Genes Dev. 2014, 28, 396–408. [Google Scholar] [CrossRef]
- O’Sullivan, R.J.; Kubicek, S.; Schreiber, S.L.; Karlseder, J. Reduced histone biosynthesis and chromatin changes arising from a damage signal at telomeres. Nat. Struct. Mol. Biol. 2010, 17, 1218–1225. [Google Scholar] [CrossRef]
- Kim, C.; Jin, J.; Ye, Z.; Jadhav, R.R.; Gustafson, C.E.; Hu, B.; Cao, W.; Tian, L.; Weyand, C.M.; Goronzy, J.J. Histone deficiency and accelerated replication stress in T cell aging. J. Clin. Investig. 2021, 131, e143632. [Google Scholar] [CrossRef]
- Chen, Y.; Bravo, J.I.; Son, J.M.; Lee, C.; Benayoun, B.A. Remodeling of the H3 nucleosomal landscape during mouse aging. Transl. Med. Aging 2020, 4, 22–31. [Google Scholar] [CrossRef]
- Lu, Y.-X.; Regan, J.C.; Eßer, J.; Drews, L.F.; Weinseis, T.; Stinn, J.; Hahn, O.; Miller, R.A.; Grönke, S.; Partridge, L. A TORC1-histone axis regulates chromatin organisation and non-canonical induction of autophagy to ameliorate ageing. eLife 2021, 10, e62233. [Google Scholar] [CrossRef]
- MacRae, S.L.; Croken, M.M.; Calder, R.B.; Aliper, A.; Milholland, B.; White, R.R.; Zhavoronkov, A.; Gladyshev, V.N.; Seluanov, A.; Gorbunova, V.; et al. DNA repair in species with extreme lifespan differences. Aging 2015, 7, 1171–1184. [Google Scholar] [CrossRef] [PubMed]
- Tan, L.; Ke, Z.; Tombline, G.; Macoretta, N.; Hayes, K.; Tian, X.; Lv, R.; Ablaeva, J.; Gilbert, M.; Bhanu, N.V.; et al. Naked Mole Rat Cells Have a Stable Epigenome that Resists iPSC Reprogramming. Stem Cell Rep. 2017, 9, 1721–1734. [Google Scholar] [CrossRef] [PubMed]
- Pascual-Torner, M.; Carrero, D.; Pérez-Silva, J.G.; Álvarez-Puente, D.; Roiz-Valle, D.; Bretones, G.; Rodríguez, D.; Maeso, D.; Mateo-González, E.; Español, Y.; et al. Comparative genomics of mortal and immortal cnidarians unveils novel keys behind rejuvenation. Proc. Natl. Acad. Sci. USA 2022, 119, e2118763119. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Z.; Liu, Y.; Feng, Y.; Klepin, S.; Tsimring, L.S.; Pillus, L.; Hasty, J.; Hao, N. Engineering longevity-design of a synthetic gene oscillator to slow cellular aging. Science 2023, 380, 376–381. [Google Scholar] [CrossRef]
Hallmark | Alteration | Consequence | Reference |
---|---|---|---|
Telomere shortening | Telomere attrition | Decreased lifespan | [31,40] |
Mitochondrial dysfunction, Accumulation of ROS | Increase in senescence | [33,34] | |
DNA Damage and Repair | rDNA damage accumulation | Increased genomic instability, decreased lifespan | [74,75] |
Decline of DDR capacity: Upregulation of alt-NHEJ repair | Increased mutagenic alterations, increased genomic instability, decreased lifespan. | [115,116] | |
Histone modification | Loss of repressing marks | Loss of heterochromatin: increased genomic instability, aberrant gene expression | [138] |
Increase in H3K4me3 marks | Activation of aging-related genes | [145] | |
Absence of H3K9 deacetylase | Increased genome instability: reduced lifespan | [180] | |
Increased H2A ubiquitination | Increased histone proteolysis, DNA DSB and genome instability | [200] | |
DNA methylation | Hypermethylation of promoter regions | Degradation of transcriptional networks, genomic instability, decreased lifespan | [224] |
Hypermethylation of rDNA | Ribosomal dysregulation, metabolic control during aging | [244] | |
Chromatin changes | Increased loss of heterochromatin | Increased loss of transcriptional silencing, genomic instability, accelerated aging | [289] |
Altered chromatin protein repertoire | Altered chromatin and gene expression dynamics | [309] | |
ncRNAs | Differential miRNA expression patterns | Age-related tuning of protein expression | [266] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
López-Gil, L.; Pascual-Ahuir, A.; Proft, M. Genomic Instability and Epigenetic Changes during Aging. Int. J. Mol. Sci. 2023, 24, 14279. https://doi.org/10.3390/ijms241814279
López-Gil L, Pascual-Ahuir A, Proft M. Genomic Instability and Epigenetic Changes during Aging. International Journal of Molecular Sciences. 2023; 24(18):14279. https://doi.org/10.3390/ijms241814279
Chicago/Turabian StyleLópez-Gil, Lucía, Amparo Pascual-Ahuir, and Markus Proft. 2023. "Genomic Instability and Epigenetic Changes during Aging" International Journal of Molecular Sciences 24, no. 18: 14279. https://doi.org/10.3390/ijms241814279
APA StyleLópez-Gil, L., Pascual-Ahuir, A., & Proft, M. (2023). Genomic Instability and Epigenetic Changes during Aging. International Journal of Molecular Sciences, 24(18), 14279. https://doi.org/10.3390/ijms241814279