Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Perspective
  • Published:

Targeting DNA damage in ageing: towards supercharging DNA repair

Abstract

Ageing is the most important risk factor for many common human diseases, including cancer, diabetes, neurodegeneration and cardiovascular disease. Consequently, combating ageing itself has emerged as a rational strategy for addressing age-related multimorbidity. Over the past three decades, multiple genetic and pharmacologic interventions have led to substantial extension of lifespan and healthspan in model organisms. However, it is unclear whether these interventions target the causal mechanisms of ageing or downstream consequences. Ample evidence suggests that DNA damage to the somatic genome is a major causal mechanism of ageing, which compromises essential cellular functions such as transcription and replication, and leads to cellular senescence, apoptosis and mutations. Recently, new concepts have emerged to target the main consequences of DNA damage and enhance DNA repair capacities, thereby extending maintenance of the genome. Here, we review advances in this field and discuss approaches to pharmacologically mitigate the adverse effects of DNA damage to delay ageing, prevent mutation-driven cancer and mitigate age-related degenerative diseases.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Sources and types of DNA damage and the molecular, cellular and organismal consequences.
Fig. 2: Causes of cellular senescence and pharmacological intervention strategies.
Fig. 3: Inflammatory responses to DNA damage.
Fig. 4: Single-enzyme DNA repair reactions.
Fig. 5: Promotion of DNA repair regulator activity by nicotinamide adenine dinucleotide (NAD+).
Fig. 6: Regulation of DNA repair capacities by the DREAM repressor.

Similar content being viewed by others

References

  1. World population prospects 2022: summary of results. United Nations Population Division https://www.un.org/development/desa/pd/content/World-Population-Prospects-2022 (2022).

  2. Current population estimates as of October 1, 2022. Statistics Bureau of Japan https://www.stat.go.jp/english/data/jinsui/2022np/index.html (2022).

  3. Schumacher, B., Pothof, J., Vijg, J. & Hoeijmakers, J. H. J. The central role of DNA damage in the ageing process. Nature 592, 695–703 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhang, L. et al. Maintenance of genome sequence integrity in long- and short-lived rodent species. Sci. Adv. 7, eabj3284 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Cagan, A. et al. Somatic mutation rates scale with lifespan across mammals. Nature 604, 517–524 (2022). The demonstration that somatic mutation rates are highly correlated with species-specific lifespan in mammals suggests that the pace of ageing is dependent on the capacities of DNA repair.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. De Bont, R. & van Larebeke, N. Endogenous DNA damage in humans: a review of quantitative data. Mutagenesis 19, 169–185 (2004).

    Article  PubMed  Google Scholar 

  7. Gates, K. S. An overview of chemical processes that damage cellular DNA: spontaneous hydrolysis, alkylation, and reactions with radicals. Chem. Res. Toxicol. 22, 1747–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Mulderrig, L. et al. Aldehyde-driven transcriptional stress triggers an anorexic DNA damage response. Nature 600, 158–163 (2021). This study suggests that formaldehyde is an endogenous metabolic genotoxin that could lead to DNA damage, increasingly impairing transcription during ageing.

    Article  CAS  PubMed  Google Scholar 

  9. Chatterjee, N. & Walker, G. C. Mechanisms of DNA damage, repair and mutagenesis. Environ. Mol. Mutagen. 58, 235–263 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Rieckher, M. et al. Distinct DNA repair mechanisms prevent formaldehyde toxicity during development, reproduction and aging. Nucleic Acids Res. 52, 8271–8285 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yousefzadeh, M. et al. DNA damage—how and why we age? eLife 10, e62852 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Cosper, P. F., Copeland, S. E., Tucker, J. B. & Weaver, B. A. Chromosome missegregation as a modulator of radiation sensitivity. Semin. Radiat. Oncol. 32, 54–63 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Bulock, C. R., Xing, X. & Shcherbakova, P. V. DNA polymerase δ proofreads errors made by DNA polymerase ε. Proc. Natl Acad. Sci. USA 117, 6035–6041 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ahmad, T. et al. The proofreading exonuclease of leading-strand DNA polymerase epsilon prevents replication fork collapse at broken template strands. Nucleic Acids Res. 51, 12288–12302 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Goodman, M. F. & Woodgate, R. Translesion DNA polymerases. CSH Perspect. Biol. 5, a010363 (2013).

    Google Scholar 

  16. Stoeger, T. et al. Aging is associated with a systemic length-associated transcriptome imbalance. Nat. Aging 2, 1191–1206 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Soheili-Nezhad, S., Ibáñez-Solé, O., Izeta, A., Hoeijmakers, J. H. J. & Stoeger, T. Time is ticking faster for long genes in aging. Trends Genet. 40, 299–312 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ibañez-Solé, O., Barrio, I. & Izeta, A. Age or lifestyle-induced accumulation of genotoxicity is associated with a length-dependent decrease in gene expression. iScience 26, 106368 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Vermeij, W. P. et al. Restricted diet delays accelerated ageing and genomic stress in DNA-repair-deficient mice. Nature 537, 427–431 (2016). This work indicates that calorie restriction could mitigate premature ageing by reducing DNA damage, and it provides a rationale for dietary interventions for progeroid syndromes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Scheibye-Knudsen, M. et al. Cockayne syndrome group A and B proteins converge on transcription-linked resolution of non-B DNA. Proc. Natl Acad. Sci. USA 113, 12502–12507 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Dou, Z. et al. Cytoplasmic chromatin triggers inflammation in senescence and cancer. Nature 550, 402–406 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Garcia-Cao, I. et al. ‘Super p53’ mice exhibit enhanced DNA damage response, are tumor resistant and age normally. EMBO J. 21, 6225–6235 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Tyner, S. D. et al. p53 mutant mice that display early ageing-associated phenotypes. Nature 415, 45–53 (2002).

    Article  CAS  PubMed  Google Scholar 

  24. Krtolica, A., Parrinello, S., Lockett, S., Desprez, P.-Y. & Campisi, J. Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging. Proc. Natl Acad. Sci. USA 98, 12072–12077 (2001). This work demonstrates that SnCs promote growth of cancer cells in their proximity, a foundational finding for the influence of SnCs on their microenvironment.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Rodier, F. et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat. Cell Biol. 11, 973–979 (2009). The first report that SnCs are actively secreting inflammatory cytokines; it coins the term senescence-associated secretory phenotype, in short SASP, for this phenomenon.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Baker, D. J. et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479, 232–236 (2011). This paper shows that elimination of SnCs could extend lifespan, providing the foundational concept for senotherapeutics.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Demaria, M. et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev. Cell 31, 722–733 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Crossley, M. P. et al. R-loop-derived cytoplasmic RNA–DNA hybrids activate an immune response. Nature 613, 187–194 (2023). This work shows that R-loop-derived RNA–DNA hybrids in the cytoplasm activate innate immune signalling, linking DNA damage to inflammatory responses.

    Article  CAS  PubMed  Google Scholar 

  29. Chatzidoukaki, O. et al. R-loops trigger the release of cytoplasmic ssDNAs leading to chronic inflammation upon DNA damage. Sci. Adv. 7, eabj5769 (2021). This work shows that DNA damage accumulation could trigger chronic inflammation via activation of the innate immune response to cytosolic DNA.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Clydesdale, G. J., Dandie, G. W. & Muller, H. K. Ultraviolet light induced injury: immunological and inflammatory effects. Immunol. Cell Biol. 79, 547–568 (2001).

    Article  CAS  PubMed  Google Scholar 

  31. Gulen, M. F. et al. cGAS–STING drives ageing-related inflammation and neurodegeneration. Nature 620, 374–380 (2023). Demonstration that cGAS-STING could trigger neurodegeneration through pro-inflammatory signalling.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Welch, G. & Tsai, L. Mechanisms of DNA damage‐mediated neurotoxicity in neurodegenerative disease. EMBO Rep. 23, e54217 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Polo, S. E. & Almouzni, G. Chromatin dynamics after DNA damage: the legacy of the access-repair-restore model. DNA Repair 36, 114–121 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Fillingham, J., Keogh, M.-C. & Krogan, N. J. γH2AX and its role in DNA double-strand break repair. Biochem. Cell Biol. 84, 568–577 (2006).

    Article  CAS  PubMed  Google Scholar 

  35. Groth, A., Rocha, W., Verreault, A. & Almouzni, G. Chromatin challenges during DNA replication and repair. Cell 128, 721–733 (2007).

    Article  CAS  PubMed  Google Scholar 

  36. Wang, S., Meyer, D. H. & Schumacher, B. H3K4me2 regulates the recovery of protein biosynthesis and homeostasis following DNA damage. Nat. Struct. Mol. Biol. 27, 1165–1177 (2020). Links the ageing hallmarks of DNA damage and its repair to epigenetic regulation and protein homeostasis, by demonstrating that, upon repair of transcription-blocking lesions, the recovery of the expression of protein biosynthesis and homeostasis genes is regulated through the deposition of H3K4me2 along the open reading frames of those genes.

    Article  CAS  PubMed  Google Scholar 

  37. Parreno, V. et al. Transient loss of Polycomb components induces an epigenetic cancer fate. Nature 629, 688–696 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Lu, A. T. et al. Universal DNA methylation age across mammalian tissues. Nat. Aging 3, 1144–1166 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Perez, K. et al. DNA repair-deficient premature aging models display accelerated epigenetic age. Aging Cell 23, e14058 (2024).

    Article  CAS  PubMed  Google Scholar 

  40. Meyer, D. H. & Schumacher, B. Aging clocks based on accumulating stochastic variation. Nat. Aging 4, 871–885 (2024). This work shows that the increased stochasticity alone is sufficient to determine biological age, providing a profound explanation of why the ageing process that is driven by accumulation of random damage advances gradually through time.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Koch, Z., Li, A., Evans, D. S., Cummings, S. & Ideker, T. Somatic mutation as an explanation for epigenetic aging. Nat. Aging 5, 709–719 (2025).

    Article  CAS  PubMed  Google Scholar 

  42. Wang, S., Meyer, D. H. & Schumacher, B. Inheritance of paternal DNA damage by histone-mediated repair restriction. Nature 613, 365–374 (2023). Demonstrates that paternal DNA damage is inaccurately repaired by maternal DNA repair in the zygotes, leading to recurrent genome instability in the offspring as the damage is maintained through heterochromatization, preventing accurate repair.

    Article  CAS  PubMed  Google Scholar 

  43. Ren, P., Dong, X. & Vijg, J. Age-related somatic mutation burden in human tissues. Front. Aging 3, 1018119 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Li, R. et al. A body map of somatic mutagenesis in morphologically normal human tissues. Nature 597, 398–403 (2021).

    Article  CAS  PubMed  Google Scholar 

  45. Brazhnik, K. et al. Single-cell analysis reveals different age-related somatic mutation profiles between stem and differentiated cells in human liver. Sci. Adv. 6, eaax2659 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Rouhani, F. J. et al. Mutational history of a human cell lineage from somatic to induced pluripotent stem cells. PLoS Genet. 12, e1005932 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Thompson, O. et al. Low rates of mutation in clinical grade human pluripotent stem cells under different culture conditions. Nat. Commun. 11, 1528 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Martincorena, I. et al. Universal patterns of selection in cancer and somatic tissues. Cell 171, 1029–1041.e21 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Li, Y. et al. Patterns of somatic structural variation in human cancer genomes. Nature 578, 112–121 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Hamdan, A. & Ewing, A. Unravelling the tumour genome: the evolutionary and clinical impacts of structural variants in tumourigenesis. J. Pathol. 257, 479–493 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Li, C., Chen, L., Pan, G., Zhang, W. & Li, S. C. Deciphering complex breakage-fusion-bridge genome rearrangements with ambigram. Nat. Commun. 14, 5528 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Robinson, P. S. et al. Increased somatic mutation burdens in normal human cells due to defective DNA polymerases. Nat. Genet. 53, 1434–1442 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Albertson, T. M. et al. DNA polymerase ε and δ proofreading suppress discrete mutator and cancer phenotypes in mice. Proc. Natl Acad. Sci. USA 106, 17101–17104 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hegan, D. C. et al. Differing patterns of genetic instability in mice deficient in the mismatch repair genes Pms2, Mlh1, Msh2, Msh3 and Msh6. Carcinogenesis 27, 2402–2408 (2006).

    Article  CAS  PubMed  Google Scholar 

  55. Franco, I., Revêchon, G. & Eriksson, M. Challenges of proving a causal role of somatic mutations in the aging process. Aging Cell 21, e13613 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Carrero, D., Soria-Valles, C. & López-Otín, C. Hallmarks of progeroid syndromes: lessons from mice and reprogrammed cells. Dis. Model. Mech. 9, 719–735 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. 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. 33, 3–17 (2017).

    Article  CAS  PubMed  Google Scholar 

  58. Gonzalo, S. & Kreienkamp, R. DNA repair defects and genome instability in Hutchinson–Gilford progeria syndrome. Curr. Opin. Cell Biol. 34, 75–83 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Hickson, I. D. RecQ helicases: caretakers of the genome. Nat. Rev. Cancer 3, 169–178 (2003).

    Article  CAS  PubMed  Google Scholar 

  60. Gordon, L. B. et al. Association of lonafarnib treatment vs no treatment with mortality rate in patients with Hutchinson–Gilford progeria syndrome. JAMA 319, 1687–1695 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Schumacher, B. et al. Delayed and accelerated aging share common longevity assurance mechanisms. PLoS Genet. 4, e1000161 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Wang, S., Prizment, A., Thyagarajan, B. & Blaes, A. Cancer treatment-induced accelerated aging in cancer survivors: biology and assessment. Cancers 13, 427 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Torgovnick, A. & Schumacher, B. DNA repair mechanisms in cancer development and therapy. Front. Genet. 6, 157 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Lee, J.-M., Ledermann, J. A. & Kohn, E. C. PARP inhibitors for BRCA1/2 mutation-associated and BRCA-like malignancies. Ann. Oncol. 25, 32–40 (2014).

    Article  PubMed  Google Scholar 

  65. Priya, B., Ravi, S. & Kirubakaran, S. Targeting ATM and ATR for cancer therapeutics: inhibitors in clinic. Drug. Discov. Today 28, 103662 (2023).

    Article  CAS  PubMed  Google Scholar 

  66. Glorieux, M., Dok, R. & Nuyts, S. Novel DNA targeted therapies for head and neck cancers: clinical potential and biomarkers. Oncotarget 8, 81662–81678 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Gilmer, T. M. et al. A novel dual ATM/DNA-PK inhibitor, XRD-0394, potently radiosensitizes and potentiates PARP and topoisomerase I inhibitors. Mol. Cancer Ther. 23, 751–765 (2024).

    Article  CAS  PubMed  Google Scholar 

  68. Abad, E., Graifer, D. & Lyakhovich, A. DNA damage response and resistance of cancer stem cells. Cancer Lett. 474, 106–117 (2020).

    Article  CAS  PubMed  Google Scholar 

  69. Somasagara, R. R. et al. Rad6 upregulation promotes stem cell-like characteristics and platinum resistance in ovarian cancer. Biochem. Biophys. Res. Commun. 469, 449–455 (2016).

    Article  CAS  PubMed  Google Scholar 

  70. Hayflick, L. & Moorhead, P. S. The serial cultivation of human diploid cell strains. Exp. Cell Res. 25, 585–621 (1961).

    Article  CAS  PubMed  Google Scholar 

  71. Kotsantis, P., Petermann, E. & Boulton, S. J. Mechanisms of oncogene-induced replication stress: jigsaw falling into place. Cancer Discov. 8, 537–555 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Kumari, R. & Jat, P. Mechanisms of cellular senescence: cell cycle arrest and senescence associated secretory phenotype. Front. Cell Dev. Biol. 9, 645593 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Baker, D. J. et al. BubR1 insufficiency causes early onset of aging-associated phenotypes and infertility in mice. Nat. Genet. 36, 744–749 (2004).

    Article  CAS  PubMed  Google Scholar 

  74. Tripathi, U. et al. SARS-CoV-2 causes senescence in human cells and exacerbates the senescence-associated secretory phenotype through TLR-3. Aging 13, 21838–21854 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Lee, S. et al. Virus-induced senescence is a driver and therapeutic target in COVID-19. Nature 599, 283–289 (2021).

    Article  CAS  PubMed  Google Scholar 

  76. Schmitt, C. A. et al. COVID-19 and cellular senescence. Nat. Rev. Immunol. 23, 251–263 (2023).

    Article  CAS  PubMed  Google Scholar 

  77. Wang, B. et al. Intermittent clearance of p21-highly-expressing cells extends lifespan and confers sustained benefits to health and physical function. Cell Metab. 36, 1795–1805.e6 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. d’Adda di Fagagna, F. Living on a break: cellular senescence as a DNA-damage response. Nat. Rev. Cancer 8, 512–522 (2008).

    Article  PubMed  Google Scholar 

  79. Zhang, L., Pitcher, L. E., Prahalad, V., Niedernhofer, L. J. & Robbins, P. D. Targeting cellular senescence with senotherapeutics: senolytics and senomorphics. FEBS J. 290, 1362–1383 (2023).

    Article  CAS  PubMed  Google Scholar 

  80. Chaib, S., Tchkonia, T. & Kirkland, J. L. Cellular senescence and senolytics: the path to the clinic. Nat. Med. 28, 1556–1568 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Lombardo, L. J. et al. Discovery of N-(2-chloro-6-methyl- phenyl)-2-(6-(4-(2-hydroxyethyl)- piperazin-1-yl)-2-methylpyrimidin-4- ylamino)thiazole-5-carboxamide (BMS-354825), a dual Src/Abl kinase inhibitor with potent antitumor activity in preclinical assays. J. Med. Chem. 47, 6658–6661 (2004).

    Article  CAS  PubMed  Google Scholar 

  82. Pasquale, E. B. Eph receptors and ephrins in cancer: bidirectional signalling and beyond. Nat. Rev. Cancer 10, 165–180 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Nambiar, A. et al. Senolytics dasatinib and quercetin in idiopathic pulmonary fibrosis: results of a phase I, single-blind, single-center, randomized, placebo-controlled pilot trial on feasibility and tolerability. eBioMedicine 90, 104481 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Hickson, L. J. et al. Senolytics decrease senescent cells in humans: preliminary report from a clinical trial of dasatinib plus quercetin in individuals with diabetic kidney disease. EBioMedicine 47, 446–456 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  85. Gonzales, M. M. et al. Senolytic therapy in mild Alzheimer’s disease: a phase 1 feasibility trial. Nat. Med. 29, 2481–2488 (2023). The first clinical trial using senotherapeutics in patients with Alzheimer disease; it supports safety, tolerability and feasibility, marking an advance towards geroprotective therapies that target the senescence response.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Farr, J. N. et al. Effects of intermittent senolytic therapy on bone metabolism in postmenopausal women: a phase 2 randomized controlled trial. Nat. Med. 30, 2605–2612 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Millar, C. L. et al. Rationale and design of STAMINA: senolytics to alleviate mobility issues and neurological impairments in aging, a geroscience feasibility study. Transl. Med. Aging 7, 109–117 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Yousefzadeh, M. J. et al. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine 36, 18–28 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Silva, M. et al. Senolytics to slow progression of sepsis (STOP-sepsis) in elderly patients: study protocol for a multicenter, randomized, adaptive allocation clinical trial. Trials 25, 698 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Tavenier, J. et al. Fisetin as a senotherapeutic agent: evidence and perspectives for age-related diseases. Mech. Ageing Dev. 222, 111995 (2024).

    Article  CAS  PubMed  Google Scholar 

  91. Lim, W., Yang, C., Bazer, F. W. & Song, G. Luteolin inhibits proliferation and induces apoptosis of human placental choriocarcinoma cells by blocking the PI3K/AKT pathway and regulating sterol regulatory element binding protein activity1. Biol. Reprod. 95, 82 (2016).

    Article  PubMed  Google Scholar 

  92. Lu, X., Li, Y., Li, X. & Aisa, H. A. Luteolin induces apoptosis in vitro through suppressing the MAPK and PI3K signaling pathways in gastric cancer. Oncol. Lett. 14, 1993–2000 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Zumerle, S. et al. Targeting senescence induced by age or chemotherapy with a polyphenol-rich natural extract improves longevity and healthspan in mice. Nat. Aging 4, 1231–1248 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Shi, M. et al. Luteolin, a flavone ingredient: anticancer mechanisms, combined medication strategy, pharmacokinetics, clinical trials, and pharmaceutical researches. Phytother. Res. 38, 880–911 (2024).

    Article  CAS  PubMed  Google Scholar 

  95. Cho, H.-J. et al. Nintedanib induces senolytic effect via STAT3 inhibition. Cell Death Dis. 13, 760 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Flaherty, K. R. et al. Nintedanib in progressive fibrosing interstitial lung diseases. N. Engl. J. Med. 381, 1718–1727 (2019).

    Article  CAS  PubMed  Google Scholar 

  97. Raman, L. et al. Nintedanib for non-IPF progressive pulmonary fibrosis: 12-month outcome data from a real-world multicentre observational study. ERJ Open Res. 9, 00423-2022 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  98. Richeldi, L. et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N. Engl. J. Med. 370, 2071–2082 (2014).

    Article  PubMed  Google Scholar 

  99. Deterding, R. et al. Nintedanib in children and adolescents with fibrosing interstitial lung diseases. Eur. Respir. J. 61, 2201512 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Mikyskova, R. et al. STAT3 inhibitor Stattic and its analogues inhibit STAT3 phosphorylation and modulate cytokine secretion in senescent tumour cells. Mol. Med. Rep. 27, 81 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Zhang, W. et al. Targeting the JAK2-STAT3 pathway to inhibit cGAS-STING activation improves neuronal senescence after ischemic stroke. Exp. Neurol. 368, 114474 (2023).

    Article  CAS  PubMed  Google Scholar 

  102. Neuwahl, J. et al. Combined inhibition of class 1-PI3K-alpha and delta isoforms causes senolysis by inducing p21WAF1/CIP1 proteasomal degradation in senescent cells. Cell Death Dis. 15, 373 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Fan, Z. et al. Inhibitor PF-04691502 works as a senolytic to regulate cellular senescence. Exp. Gerontol. 186, 112359 (2024).

    Article  CAS  PubMed  Google Scholar 

  104. Wainberg, Z. A. et al. A multi-arm phase I study of the PI3K/mTOR inhibitors PF-04691502 and gedatolisib (PF-05212384) plus irinotecan or the MEK inhibitor PD-0325901 in advanced cancer. Target. Oncol. 12, 775–785 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  105. Britten, C. D. et al. Phase I study of PF-04691502, a small-molecule, oral, dual inhibitor of PI3K and mTOR, in patients with advanced cancer. Invest. New Drugs 32, 510–517 (2014).

    Article  CAS  PubMed  Google Scholar 

  106. Bowles, D. W. et al. A multicenter phase 1 study of PX-866 in combination with docetaxel in patients with advanced solid tumours. Br. J. Cancer 109, 1085–1092 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Hong, D. S. et al. A multicenter phase I trial of PX-866, an oral irreversible phosphatidylinositol 3-kinase inhibitor, in patients with advanced solid tumors. Clin. Cancer Res. 18, 4173–4182 (2012).

    Article  CAS  PubMed  Google Scholar 

  108. Hotte, S. J. et al. A phase II study of PX-866 in patients with recurrent or metastatic castration-resistant prostate cancer: Canadian cancer trials group study IND205. Clin. Genitourin. Cancer 17, 201–208.e1 (2019).

    Article  PubMed  Google Scholar 

  109. Jimeno, A. et al. A randomized, phase II trial of cetuximab with or without PX-866, an irreversible oral phosphatidylinositol 3-kinase inhibitor, in patients with relapsed or metastatic head and neck squamous cell cancer. Ann. Oncol. 26, 556–561 (2015).

    Article  CAS  PubMed  Google Scholar 

  110. Pitz, M. W. et al. Phase II study of PX-866 in recurrent glioblastoma. Neuro Oncol. 17, 1270–1274 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Hossain, M. T. & Hossain, M. A. Targeting PI3K in cancer treatment: a comprehensive review with insights from clinical outcomes. Eur. J. Pharmacol. 996, 177432 (2025).

    Article  CAS  PubMed  Google Scholar 

  112. Hedges, C. P. et al. Dietary supplementation of clinically utilized PI3K p110α inhibitor extends the lifespan of male and female mice. Nat. Aging 3, 162–172 (2023).

    Article  CAS  PubMed  Google Scholar 

  113. Fuhrmann-Stroissnigg, H. et al. Identification of HSP90 inhibitors as a novel class of senolytics. Nat. Commun. 8, 422 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  114. Mellatyar, H. et al. Targeted cancer therapy through 17-DMAG as an Hsp90 inhibitor: overview and current state of the art. Biomed. Pharmacother. 102, 608–617 (2018).

    Article  CAS  PubMed  Google Scholar 

  115. Rastogi, S. et al. An update on the status of HSP90 inhibitors in cancer clinical trials. Cell Stress Chaperones 29, 519–539 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Honma, Y. et al. Randomized, double-blind, placebo (PL)-controlled, phase III trial of pimitespib (TAS-116), an oral inhibitor of heat shock protein 90 (HSP90), in patients (pts) with advanced gastrointestinal stromal tumor (GIST) refractory to imatinib (IM), sunitinib (SU) and regorafenib (REG). J. Clin. Oncol. 39, 11524 (2021).

    Article  Google Scholar 

  117. Fuhrmann-Stroissnigg, H., Niedernhofer, L. J. & Robbins, P. D. Hsp90 inhibitors as senolytic drugs to extend healthy aging. Cell Cycle 17, 1048–1055 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Yosef, R. et al. Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nat. Commun. 7, 11190 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Chang, J. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat. Med. 22, 78–83 (2016). Chang et al. demonstrate that pharmacological elimination of SnCs could reverse the functional decline of haematopoietic stem cells in irradiated and in aged mice, providing a proof of concept for senotherapeutic treatments in vivo.

    Article  CAS  PubMed  Google Scholar 

  120. Dilley, K., Harb, J., Jalaluddin, M., Hutti, J. E. & Potluri, J. A phase 3, open-label, randomized study evaluating the efficacy and safety of navitoclax plus ruxolitinib versus best available therapy in patients with relapsed/refractory myelofibrosis (TRANSFORM-2). Blood 136, 8 (2020).

    Article  Google Scholar 

  121. Harrison, C. N. et al. Addition of navitoclax to ongoing ruxolitinib therapy for patients with myelofibrosis with progression or suboptimal response: phase II safety and efficacy. J. Clin. Oncol. 40, 1671–1680 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Pemmaraju, N. et al. Transform-1: a randomized, double-blind, placebo-controlled, multicenter, international phase 3 study of navitoclax in combination with ruxolitinib versus ruxolitinib plus placebo in patients with untreated myelofibrosis. Blood 142, 620 (2023).

    Article  Google Scholar 

  123. Tse, C. et al. ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 68, 3421–3428 (2008).

    Article  CAS  PubMed  Google Scholar 

  124. Zhu, Y. et al. New agents that target senescent cells: the flavone, fisetin, and the BCL-XL inhibitors, A1331852 and A1155463. Aging 9, 955–963 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  125. Rysanek, D. et al. Synergism of BCL-2 family inhibitors facilitates selective elimination of senescent cells. Aging 14, 6381–6414 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Mukherjee, N. et al. MCL1 inhibitors S63845/MIK665 plus Navitoclax synergistically kill difficult-to-treat melanoma cells. Cell Death Dis. 11, 443 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Xu, Q. et al. The flavonoid procyanidin C1 has senotherapeutic activity and increases lifespan in mice. Nat. Metab. 3, 1706–1726 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Li, W., He, Y., Zhang, R., Zheng, G. & Zhou, D. The curcumin analog EF24 is a novel senolytic agent. Aging 11, 771–782 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Nayak, D. et al. Development and crystal structures of a potent second-generation dual degrader of BCL-2 and BCL-xL. Nat. Commun. 15, 2743 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Negi, A. & Voisin-Chiret, A. S. Strategies to reduce the on-target platelet toxicity of Bcl-xL inhibitors: PROTACs, SNIPERs and prodrug-based approaches. ChemBioChem 23, e202100689 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Wang, Y. et al. Discovery of piperlongumine as a potential novel lead for the development of senolytic agents. Aging 8, 2915–2926 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Liu, X. et al. Senolytic activity of piperlongumine analogues: synthesis and biological evaluation. Bioorg. Med. Chem. 26, 3925–3938 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Zhang, X. et al. Design and optimization of piperlongumine analogs as potent senolytics. Bioorg. Med. Chem. Lett. 98, 129593 (2024).

    Article  CAS  PubMed  Google Scholar 

  134. Zhang, X. et al. Oxidation resistance 1 is a novel senolytic target. Aging Cell 17, e12780 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  135. Baar, M. P. et al. Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell 169, 132–147.e16 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Jeon, O. H. et al. Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat. Med. 23, 775–781 (2017). In this work, the elimination of SnCs in mice attenuates the development of post-traumatic osteoarthritis, thus providing a rationale for treating age-associated osteoarthritis with senotherapeutics.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Lane, N. et al. A phase 2, randomized, double-blind, placebo-controlled study of senolytic molecule UBX0101 in the treatment of painful knee osteoarthritis. Osteoarthr. Cartil. 29, S52–S53 (2021).

    Article  Google Scholar 

  138. He, Y. et al. Inhibition of USP7 activity selectively eliminates senescent cells in part via restoration of p53 activity. Aging Cell 19, e13117 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Guerrero, A. et al. Galactose-modified duocarmycin prodrugs as senolytics. Aging Cell 19, e13133 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Hu, H.-H. et al. Roles and inhibitors of FAK in cancer: current advances and future directions. Front. Pharmacol. 15, 1274209 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Shin, E.-Y. et al. Integrin-mediated adhesions in regulation of cellular senescence. Sci. Adv. 6, eaay3909 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Genovese, M. C. et al. A phase III, multicenter, randomized, double-blind, placebo-controlled, parallel-group study of 2 dosing regimens of fostamatinib in patients with rheumatoid arthritis with an inadequate response to a tumor necrosis factor-α antagonist. J. Rheumatol. 41, 2120–2128 (2014).

    Article  CAS  PubMed  Google Scholar 

  143. Triana-Martínez, F. et al. Identification and characterization of cardiac glycosides as senolytic compounds. Nat. Commun. 10, 4731 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  144. Amor, C. et al. Senolytic CAR T cells reverse senescence-associated pathologies. Nature 583, 127–132 (2020). Amor et al. demonstrate the ability of CAR T cells to target SnCs in vivo, thus establishing the senotherapeutic potential of senolytic CAR T cell therapies.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Amor, C. et al. Prophylactic and long-lasting efficacy of senolytic CAR T cells against age-related metabolic dysfunction. Nat. Aging 4, 336–349 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Yang, D. et al. NKG2D-CAR T cells eliminate senescent cells in aged mice and nonhuman primates. Sci. Transl. Med. 15, eadd1951 (2023).

    Article  CAS  PubMed  Google Scholar 

  147. Mullard, A. In vivo CAR T cells move into clinical trials. Nat. Rev. Drug Discov. 23, 727–730 (2024).

    Article  CAS  PubMed  Google Scholar 

  148. Suda, M. et al. Senolytic vaccination improves normal and pathological age-related phenotypes and increases lifespan in progeroid mice. Nat. Aging 1, 1117–1126 (2021).

    Article  PubMed  Google Scholar 

  149. Yoshida, S. et al. The CD153 vaccine is a senotherapeutic option for preventing the accumulation of senescent T cells in mice. Nat. Commun. 11, 2482 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Poblocka, M. et al. Targeted clearance of senescent cells using an antibody-drug conjugate against a specific membrane marker. Sci. Rep. 11, 20358 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Takaya, K., Asou, T. & Kishi, K. New senolysis approach via antibody–drug conjugate targeting of the senescent cell marker apolipoprotein D for skin rejuvenation. Int. J. Mol. Sci. 24, 5857 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Huang, W., Hickson, L. J., Eirin, A., Kirkland, J. L. & Lerman, L. O. Cellular senescence: the good, the bad and the unknown. Nat. Rev. Nephrol. 18, 611–627 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Gasek, N. S. et al. Clearance of p21 highly expressing senescent cells accelerates cutaneous wound healing. Nat. Aging 5, 21–27 (2025).

    Article  CAS  PubMed  Google Scholar 

  154. Ferrucci, L. & Fabbri, E. Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty. Nat. Rev. Cardiol. 15, 505–522 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Li, X. et al. Inflammation and aging: signaling pathways and intervention therapies. Signal Transduct. Target. Ther. 8, 239 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  156. Siametis, A. et al. Transcription stress at telomeres leads to cytosolic DNA release and paracrine senescence. Nat. Commun. 15, 4061 (2024). Demonstrates that transcription stress occurring at telomeres could trigger inflammatory signalling and senescence, highlighting the role of telomeric DNA damage in inflammation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Chatzinikolaou, G., Karakasilioti, I. & Garinis, G. A. DNA damage and innate immunity: links and trade-offs. Trends Immunol. 35, 429–435 (2014).

    Article  CAS  PubMed  Google Scholar 

  158. Zhang, X. et al. Cutting edge: Ku70 is a novel cytosolic DNA sensor that induces type III rather than type I IFN. J. Immunol. 186, 4541–4545 (2011).

    Article  CAS  PubMed  Google Scholar 

  159. Ferguson, B. J., Mansur, D. S., Peters, N. E., Ren, H. & Smith, G. L. DNA-PK is a DNA sensor for IRF-3-dependent innate immunity. eLife 1, e00047 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  160. Kondo, T. et al. DNA damage sensor MRE11 recognizes cytosolic double-stranded DNA and induces type I interferon by regulating STING trafficking. Proc. Natl Acad. Sci. USA 110, 2969–2974 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Roth, S. et al. Rad50-CARD9 interactions link cytosolic DNA sensing to IL-1β production. Nat. Immunol. 15, 538–545 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Gorbunova, V. et al. The role of retrotransposable elements in ageing and age-associated diseases. Nature 596, 43–53 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. De Cecco, M. et al. L1 drives IFN in senescent cells and promotes age-associated inflammation. Nature 566, 73–78 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  164. Wu, J. et al. Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science 339, 826–830 (2013).

    Article  CAS  PubMed  Google Scholar 

  165. Sun, L., Wu, J., Du, F., Chen, X. & Chen, Z. J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339, 786–791 (2013). This study and Wu et al. (2013) show that cGAS binds to cytoplasmic DNA and catalyses the production of cGAMP, which acts as a second messenger to trigger a signalling cascade, leading to the activation of an innate immune response.

    Article  CAS  PubMed  Google Scholar 

  166. Ishikawa, H. & Barber, G. N. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature 455, 674–678 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Decout, A., Katz, J. D., Venkatraman, S. & Ablasser, A. The cGAS–STING pathway as a therapeutic target in inflammatory diseases. Nat. Rev. Immunol. 21, 548–569 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Martinez, J. C. et al. cGAS deficient mice display premature aging associated with de-repression of LINE1 elements and inflammation. Preprint at https://doi.org/10.1101/2024.10.10.617645 (2024).

  169. Vincent, J. et al. Small molecule inhibition of cGAS reduces interferon expression in primary macrophages from autoimmune mice. Nat. Commun. 8, 750 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  170. Lama, L. et al. Development of human cGAS-specific small-molecule inhibitors for repression of dsDNA-triggered interferon expression. Nat. Commun. 10, 2261 (2019). Development of a cGAS inhibitor that could provide a pharmacological strategy to mitigate cGAS’s inflammatory signalling outcome.

    Article  PubMed  PubMed Central  Google Scholar 

  171. Udeochu, J. C. et al. Tau activation of microglial cGAS–IFN reduces MEF2C-mediated cognitive resilience. Nat. Neurosci. 26, 737–750 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Chen, M. et al. Design, synthesis, and pharmacological evaluation of spiro[carbazole-3,3′-pyrrolidine] derivatives as cGAS inhibitors for treatment of acute lung injury. J. Med. Chem. 67, 6268–6291 (2024).

    Article  CAS  PubMed  Google Scholar 

  173. Pike, K. et al. Discovery of VENT-03: a novel clinical cGAS inhibitor for the treatment of SLE and other autoimmune diseases. ACR Meeting abstr. 1537, https://acrabstracts.org/abstract/discovery-of-vent-03-a-novel-clinical-cgas-inhibitor-for-the-treatment-of-sle-and-other-autoimmune-diseases/ (2024).

  174. An, J., Woodward, J. J., Sasaki, T., Minie, M. & Elkon, K. B. Cutting edge: antimalarial drugs inhibit IFN-β production through blockade of cyclic GMP-AMP synthase–DNA interaction. J. Immunol. 194, 4089–4093 (2015).

    Article  CAS  PubMed  Google Scholar 

  175. An, J. et al. Inhibition of cyclic GMP-AMP synthase using a novel antimalarial drug derivative in Trex1-deficient mice. Arthritis Rheumatol. 70, 1807–1819 (2018).

    Article  CAS  PubMed  Google Scholar 

  176. Dai, J. et al. Acetylation blocks cGAS activity and inhibits self-DNA-induced autoimmunity. Cell 176, 1447–1460.e14 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Liu, S. et al. Phosphorylation of innate immune adaptor proteins MAVS, STING, and TRIF induces IRF3 activation. Science 347, aaa2630 (2015).

    Article  PubMed  Google Scholar 

  178. Haag, S. M. et al. Targeting STING with covalent small-molecule inhibitors. Nature 559, 269–273 (2018). First discovery and demonstration that drugs targeting STING could be used to treat autoinflammatory disease in mouse models.

    Article  CAS  PubMed  Google Scholar 

  179. Wang, B. et al. The STING inhibitor C-176 attenuates MPTP-induced neuroinflammation and neurodegeneration in mouse parkinsonian models. Int. Immunopharmacol. 124, 110827 (2023).

    Article  CAS  PubMed  Google Scholar 

  180. Humphries, F. et al. Targeting STING oligomerization with small-molecule inhibitors. Proc. Natl Acad. Sci. USA 120, e2305420120 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Barasa, L. et al. Development of LB244, an irreversible STING antagonist. J. Am. Chem. Soc. 145, 20273–20288 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Hong, Z. et al. STING inhibitors target the cyclic dinucleotide binding pocket. Proc. Natl Acad. Sci. USA 118, e2105465118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Hou, Y. et al. SMPDL3A is a cGAMP-degrading enzyme induced by LXR-mediated lipid metabolism to restrict cGAS-STING DNA sensing. Immunity 56, 2492–2507.e10 (2023).

    Article  CAS  PubMed  Google Scholar 

  184. Zou, M. et al. Inhibition of cGAS-STING by JQ1 alleviates oxidative stress-induced retina inflammation and degeneration. Cell Death Differ. 29, 1816–1833 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Zhu, X. et al. The BET PROTAC inhibitor dBET6 protects against retinal degeneration and inhibits the cGAS-STING in response to light damage. J. Neuroinflammation 20, 119 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Thomson, D. W. et al. Discovery of GSK8612, a highly selective and potent TBK1 inhibitor. ACS Med. Chem. Lett. 10, 780–785 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Shi, W. et al. Compound danshen dripping pill effectively alleviates cGAS-STING-triggered diseases by disrupting STING-TBK1 interaction. Phytomedicine 128, 155404 (2024).

    Article  CAS  PubMed  Google Scholar 

  188. Clark, K. et al. Novel cross-talk within the IKK family controls innate immunity. Biochem. J. 434, 93–104 (2011).

    Article  CAS  PubMed  Google Scholar 

  189. Clark, K., Plater, L., Peggie, M. & Cohen, P. Use of the pharmacological inhibitor BX795 to study the regulation and physiological roles of TBK1 and IκB kinase ε: a distinct upstream kinase mediates Ser-172 phosphorylation and activation. J. Biol. Chem. 284, 14136–14146 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Wang, Y. et al. HIV-1 Vif suppresses antiviral immunity by targeting STING. Cell. Mol. Immunol. 19, 108–121 (2022).

    Article  CAS  PubMed  Google Scholar 

  191. Hertzog, J. et al. Varicella-Zoster virus ORF9 is an antagonist of the DNA sensor cGAS. EMBO J. 41, e109217 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Lin, C. et al. How does African swine fever virus evade the cGAS-STING pathway? Pathogens 13, 957 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Yu, L. & Liu, P. Cytosolic DNA sensing by cGAS: regulation, function, and human diseases. Signal Transduct. Target. Ther. 6, 170 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Zhou, J., Zhuang, Z., Li, J. & Feng, Z. Significance of the cGAS-STING pathway in health and disease. Int. J. Mol. Sci. 24, 13316 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Oduro, P. K. et al. The cGAS–STING signaling in cardiovascular and metabolic diseases: future novel target option for pharmacotherapy. Acta Pharm. Sin. B 12, 50–75 (2022).

    Article  CAS  PubMed  Google Scholar 

  196. Gan, Y. et al. The cGAS/STING pathway: a novel target for cancer therapy. Front. Immunol. 12, 795401 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  197. Arvanitaki, E. S. et al. Microglia-derived extracellular vesicles trigger age-related neurodegeneration upon DNA damage. Proc. Natl Acad. Sci. USA 121, e2317402121 (2024). Development of an exosome-based therapeutic strategy to remove DNA damage-driven proinflammatory DNA fragments from the cytoplasm of microglial cells and delay neurodegeneration.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Cong, Y.-S., Wright, W. E. & Shay, J. W. Human telomerase and its regulation. Microbiol. Mol. Biol. Rev. 66, 407–425 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Hewitt, G. et al. Telomeres are favoured targets of a persistent DNA damage response in ageing and stress-induced senescence. Nat. Commun. 3, 708 (2012).

    Article  PubMed  Google Scholar 

  200. Rossiello, F., Jurk, D., Passos, J. F. & d’Adda di Fagagna, F. Telomere dysfunction in ageing and age-related diseases. Nat. Cell Biol. 24, 135–147 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. 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. 10, 4723 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  202. Bernardes de Jesus, B. et al. Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer. EMBO Mol. Med. 4, 691–704 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Harley, C. B. Telomerase and cancer therapeutics. Nat. Rev. Cancer 8, 167–179 (2008).

    Article  CAS  PubMed  Google Scholar 

  204. Kirwan, M. & Dokal, I. Dyskeratosis congenita, stem cells and telomeres. Biochim. Biophys. Acta 1792, 371–379 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Garschall, K. et al. Ubiquitous overexpression of the DNA repair factor dPrp19 reduces DNA damage and extends Drosophila life span. npj Aging Mech. Dis. 3, 5 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  206. Klein, H. L. The consequences of Rad51 overexpression for normal and tumor cells. DNA Repair 7, 686–693 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Cleaver, J. E., Charles, W. C., McDowell, M. L., Sadinski, W. J. & Mitchell, D. L. Overexpression of the XPA repair gene increases resistance to ultraviolet radiation in human cells by selective repair of DNA damage. Cancer Res. 55, 6152–6160 (1995).

    CAS  PubMed  Google Scholar 

  208. Limpose, K. L. et al. Overexpression of the base excision repair NTHL1 glycosylase causes genomic instability and early cellular hallmarks of cancer. Nucleic Acids Res. 46, 4515–4532 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Velegzhaninov, I. O. et al. Radioresistance, DNA damage and DNA repair in cells with moderate overexpression of RPA1. Front. Genet. 11, 855 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Shaposhnikov, M., Proshkina, E., Shilova, L., Zhavoronkov, A. & Moskalev, A. Lifespan and stress resistance in Drosophila with overexpressed DNA repair genes. Sci. Rep. 5, 15299 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Schärer, O. D. Nucleotide excision repair in eukaryotes. CSH Perspect. Biol. 5, a012609 (2013).

    Google Scholar 

  212. Kelner, A. Photoreactivation of ultraviolet-irradiated Escherichia coli, with special reference to the dose-reduction principle and to ultraviolet-induced mutation. J. Bacteriol. 58, 511–522 (1949).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Cellini, A. et al. Directed ultrafast conformational changes accompany electron transfer in a photolyase as resolved by serial crystallography. Nat. Chem. 16, 624–632 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Zhang, M., Wang, L. & Zhong, D. Photolyase: dynamics and electron-transfer mechanisms of DNA repair. Arch. Biochem. Biophys. 632, 158–174 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Schul, W. et al. Enhanced repair of cyclobutane pyrimidine dimers and improved UV resistance in photolyase transgenic mice. EMBO J. 21, 4719–4729 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Jans, J. et al. Powerful skin cancer protection by a CPD-photolyase transgene. Curr. Biol. 15, 105–115 (2005). This work shows that the selective repair of CPDs by a transgenic CPD-photolyase is sufficient to prevent UV-induced carcinogenesis, providing a rationale for using photolyase enzymes to prevent skin cancer.

    Article  CAS  PubMed  Google Scholar 

  217. Yasuda, S. & Sekiguchi, M. T4 endonuclease involved in repair of DNA. Proc. Natl Acad. Sci. USA 67, 1839–1845 (1970).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Dodson, M. L. & Lloyd, R. S. Structure-function studies of the T4 endonuclease V repair enzyme. Mutat. Res. 218, 49–65 (1989).

    Article  CAS  PubMed  Google Scholar 

  219. Luze, H., Nischwitz, S. P., Zalaudek, I., Müllegger, R. & Kamolz, L. P. DNA repair enzymes in sunscreens and their impact on photoageing—a systematic review. Photodermatol. Photoimmunol. Photomed. 36, 424–432 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Yarosh, D. B., Rosenthal, A. & Moy, R. Six critical questions for DNA repair enzymes in skincare products: a review in dialog. Clin. Cosmet. Investig. Dermatol. 12, 617–624 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Slade, D. & Radman, M. Oxidative stress resistance in Deinococcus radiodurans. Microbiol. Mol. Biol. Rev. 75, 133–191 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Makarova, K. S. et al. Genome of the extremely radiation-resistant bacterium Deinococcus radiodurans viewed from the perspective of comparative genomics. Microbiol. Mol. Biol. Rev. 65, 44–79 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Liu, F., Li, N. & Zhang, Y. The radioresistant and survival mechanisms of Deinococcus radiodurans. Radiat. Med. Prot. 4, 70–79 (2023).

    Article  CAS  Google Scholar 

  224. Chavez, C., Cruz-Becerra, G., Fei, J., Kassavetis, G. A. & Kadonaga, J. T. The tardigrade damage suppressor protein binds to nucleosomes and protects DNA from hydroxyl radicals. Elife 8, e47682 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Escarcega, R. D. et al. The tardigrade damage suppressor protein Dsup promotes DNA damage in neurons. Mol. Cell. Neurosci. 125, 103826 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Li, L. et al. Multi-omics landscape and molecular basis of radiation tolerance in a tardigrade. Science 386, eadl0799 (2024). Tardigrades are extremely resistant to DNA damage, and this work provides a set of mechanisms ranging from a protective pigment and a repair mechanism using phase separation to metabolic resupply of NAD+ that these animals evolved to maintain a highly stable genome.

    Article  CAS  PubMed  Google Scholar 

  227. Covarrubias, A. J., Perrone, R., Grozio, A. & Verdin, E. NAD+ metabolism and its roles in cellular processes during ageing. Nat. Rev. Mol. Cell Biol. 22, 119–141 (2021).

    Article  CAS  PubMed  Google Scholar 

  228. Ruszkiewicz, J. A., Bürkle, A. & Mangerich, A. Fueling genome maintenance: on the versatile roles of NAD+ in preserving DNA integrity. J. Biol. Chem. 298, 102037 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Ray Chaudhuri, A. & Nussenzweig, A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat. Rev. Mol. Cell Biol. 18, 610–621 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Houtkooper, R. H., Pirinen, E. & Auwerx, J. Sirtuins as regulators of metabolism and healthspan. Nat. Rev. Mol. Cell Biol. 13, 225–238 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Jeong, S. M. & Haigis, M. C. Sirtuins in cancer: a balancing act between genome stability and metabolism. Mol. Cells 38, 750–758 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Lagunas-Rangel, F. A. Current role of mammalian sirtuins in DNA repair. DNA Repair 80, 85–92 (2019).

    Article  CAS  PubMed  Google Scholar 

  233. Mei, Z. et al. Sirtuins in metabolism, DNA repair and cancer. J. Exp. Clin. Cancer Res. 35, 182 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  234. Oberdoerffer, P. et al. SIRT1 redistribution on chromatin promotes genomic stability but alters gene expression during aging. Cell 135, 907–918 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Vaquero, A. et al. Human SirT1 interacts with histone H1 and promotes formation of facultative heterochromatin. Mol. Cell 16, 93–105 (2004).

    Article  CAS  PubMed  Google Scholar 

  236. Imai, S., Armstrong, C. M., Kaeberlein, M. & Guarente, L. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature 403, 795–800 (2000).

    Article  CAS  PubMed  Google Scholar 

  237. Dobbin, M. M. et al. SIRT1 collaborates with ATM and HDAC1 to maintain genomic stability in neurons. Nat. Neurosci. 16, 1008–1015 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Uhl, M. et al. Role of SIRT1 in homologous recombination. DNA Repair 9, 383–393 (2010).

    Article  CAS  PubMed  Google Scholar 

  239. Jeong, J. et al. SIRT1 promotes DNA repair activity and deacetylation of Ku70. Exp. Mol. Med. 39, 8–13 (2007).

    Article  CAS  PubMed  Google Scholar 

  240. Fan, W. & Luo, J. SIRT1 regulates UV-induced DNA repair through deacetylating XPA. Mol. Cell 39, 247–258 (2010).

    Article  CAS  PubMed  Google Scholar 

  241. Ming, M. et al. Regulation of global genome nucleotide excision repair by SIRT1 through xeroderma pigmentosum C. Proc. Natl Acad. Sci. USA 107, 22623–22628 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Onn, L. et al. SIRT6 is a DNA double-strand break sensor. eLife 9, e51636 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Tian, X. et al. SIRT6 is responsible for more efficient DNA double-strand break repair in long-lived species. Cell 177, 622–638.e22 (2019). Report that, in comparison with short-lived mice, long-lived beavers express a specific form of SIRT6 that could improve the repair of DNA DSBs.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Roichman, A. et al. Restoration of energy homeostasis by SIRT6 extends healthy lifespan. Nat. Commun. 12, 3208 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  245. Xu, Z. et al. SIRT6 rescues the age related decline in base excision repair in a PARP1-dependent manner. Cell Cycle 14, 269–276 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  246. Geng, A. et al. The deacetylase SIRT6 promotes the repair of UV-induced DNA damage by targeting DDB2. Nucleic Acids Res. 48, 9181–9194 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  247. McCord, R. A. et al. SIRT6 stabilizes DNA-dependent protein kinase at chromatin for DNA double-strand break repair. Aging 1, 109–121 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  248. Rezazadeh, S. et al. SIRT6 mono-ADP ribosylates KDM2A to locally increase H3K36me2 at DNA damage sites to inhibit transcription and promote repair. Aging 12, 11165–11184 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Hou, T. et al. SIRT6 coordinates with CHD4 to promote chromatin relaxation and DNA repair. Nucleic Acids Res. 48, 2982–3000 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Simon, M. et al. A rare human centenarian variant of SIRT6 enhances genome stability and interaction with Lamin A. EMBO J. 41, e110393 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Biashad, S. A. et al. SIRT6 activator fucoidan extends healthspan and lifespan in aged wild-type mice. Preprint at https://doi.org/10.1101/2025.03.24.645072 (2025).

  252. Berger, N. A. & Sikorski, G. W. Nicotinamide stimulates repair of DNA damage in human lymphocytes. Biochem. Biophys. Res. Commun. 95, 67–72 (1980).

    Article  CAS  PubMed  Google Scholar 

  253. Weidele, K., Beneke, S. & Bürkle, A. The NAD+ precursor nicotinic acid improves genomic integrity in human peripheral blood mononuclear cells after X-irradiation. DNA Repair 52, 12–23 (2017).

    Article  CAS  PubMed  Google Scholar 

  254. Fang, E. F. et al. Defective mitophagy in XPA via PARP-1 hyperactivation and NAD+/SIRT1 reduction. Cell 157, 882–896 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  255. Hoch, N. C. et al. XRCC1 mutation is associated with PARP1 hyperactivation and cerebellar ataxia. Nature 541, 87–91 (2017). This work establishes that single-strand-repair deficiency becomes pathological due to PARP1 hyperactivation amid unrepaired lesions. Such a response could deprive the cells of NAD+, providing a potential rationale for intervening at the level of PARP1 inhibition or NAD+ supplementation.

    Article  CAS  PubMed  Google Scholar 

  256. Fang, E. F. et al. NAD+ replenishment improves lifespan and healthspan in ataxia telangiectasia models via mitophagy and DNA repair. Cell Metab. 24, 566–581 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  257. Scheibye-Knudsen, M. et al. A high-fat diet and NAD+ activate Sirt1 to rescue premature aging in Cockayne syndrome. Cell Metab. 20, 840–855 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. Okur, M. N. et al. Short-term NAD+ supplementation prevents hearing loss in mouse models of Cockayne syndrome. npj Aging Mech. Dis. 6, 1 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  259. Hou, Y. et al. NAD+ supplementation normalizes key Alzheimer’s features and DNA damage responses in a new AD mouse model with introduced DNA repair deficiency. Proc. Natl Acad. Sci. USA 115, E1876–E1885 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  260. Chen, A. C. et al. A phase 3 randomized trial of nicotinamide for skin-cancer chemoprevention. N. Engl. J. Med. 373, 1618–1626 (2015).

    Article  CAS  PubMed  Google Scholar 

  261. Phelan, M. J., Mulnard, R. A., Gillen, D. L. & Schreiber, S. S. Phase II clinical trial of nicotinamide for the treatment of mild to moderate Alzheimer’s disease. J. Geriatr. Med. Gerontol. 3, 021 (2017).

    Article  Google Scholar 

  262. Presterud, R. et al. Long-term nicotinamide riboside use improves coordination and eye movements in ataxia telangiectasia. Mov. Disord. 39, 360–369 (2024).

    Article  CAS  PubMed  Google Scholar 

  263. Berven, H. et al. NR-SAFE: a randomized, double-blind safety trial of high dose nicotinamide riboside in Parkinson’s disease. Nat. Commun. 14, 7793 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  264. Norheim, K. L. et al. Effect of nicotinamide riboside on airway inflammation in COPD: a randomized, placebo-controlled trial. Nat. Aging 4, 1772–1781 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  265. Reiten, O. K., Wilvang, M. A., Mitchell, S. J., Hu, Z. & Fang, E. F. Preclinical and clinical evidence of NAD+ precursors in health, disease, and ageing. Mech. Ageing Dev. 199, 111567 (2021).

    Article  CAS  PubMed  Google Scholar 

  266. You, W. et al. Structural basis of sirtuin 6 activation by synthetic small molecules. Angew. Chem. Int. Ed. 56, 1007–1011 (2017).

    Article  CAS  Google Scholar 

  267. Zhou, H. et al. Downregulation of Sirt6 by CD38 promotes cell senescence and aging. Aging 14, 9730–9757 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  268. Jiao, F., Zhang, Z., Hu, H., Zhang, Y. & Xiong, Y. SIRT6 activator UBCS039 inhibits thioacetamide-induced hepatic injury in vitro and in vivo. Front. Pharmacol. 13, 837544 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  269. Koblan, L. W. et al. In vivo base editing rescues Hutchinson–Gilford progeria syndrome in mice. Nature 589, 608–614 (2021). This study uses adenine base editors to reverse a pathological HGPS mutation in vivo providing a proof of concept for reversing mutations that lead to progeroid syndromes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  270. Wang, S. et al. Rescue of premature aging defects in Cockayne syndrome stem cells by CRISPR/Cas9-mediated gene correction. Protein Cell 11, 1–22 (2020).

    Article  PubMed  Google Scholar 

  271. Tu, J. et al. Genetic correction of Werner syndrome gene reveals impaired pro‐angiogenic function and HGF insufficiency in mesenchymal stem cells. Aging Cell 19, e13116 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  272. de Luzy, I. R., Lee, M. K., Mobley, W. C. & Studer, L. Lessons from inducible pluripotent stem cell models on neuronal senescence in aging and neurodegeneration. Nat. Aging 4, 309–318 (2024).

    Article  PubMed  PubMed Central  Google Scholar 

  273. Yücel, A. D. & Gladyshev, V. N. The long and winding road of reprogramming-induced rejuvenation. Nat. Commun. 15, 1941 (2024).

    Article  PubMed  PubMed Central  Google Scholar 

  274. Ocampo, A. et al. In vivo amelioration of age-associated hallmarks by partial reprogramming. Cell 167, 1719–1733.e12 (2016). This work demonstrates that partial reprogramming based on the Yamanaka paradigm of cellular reprogramming could be used to mitigate ageing phenotypes in vivo.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  275. Olova, N., Simpson, D. J., Marioni, R. E. & Chandra, T. Partial reprogramming induces a steady decline in epigenetic age before loss of somatic identity. Aging Cell 18, e12877 (2019).

    Article  PubMed  Google Scholar 

  276. Gill, D. et al. Multi-omic rejuvenation of human cells by maturation phase transient reprogramming. eLife 11, e71624 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  277. Zhao, Y. et al. Two supporting factors greatly improve the efficiency of human iPSC generation. Cell Stem Cell 3, 475–479 (2008).

    Article  CAS  PubMed  Google Scholar 

  278. Paine, P. T. et al. Initiation phase cellular reprogramming ameliorates DNA damage in the ERCC1 mouse model of premature aging. Front. Aging 4, 1323194 (2024).

    Article  PubMed  PubMed Central  Google Scholar 

  279. Horvath, S. DNA methylation age of human tissues and cell types. Genome Biol. 14, R115 (2013). This work shows that changes in CpG methylation could indicate the age of human cells and tissues, providing the concept of ageing clocks that could identify changes in biological age. This work is foundational for implementing biological ageing clocks as gerotherapeutic diagnostics to evaluate therapies that could prevent age-associated diseases.

    Article  PubMed  PubMed Central  Google Scholar 

  280. Lopez, M., Gilbert, J., Contreras, J., Halby, L. & Arimondo, P. B. in DNA Methyltransferases — Role and Function 2nd edn, Vol. 1389 (eds Jeltsch, A. & Jurkowska, R. Z.) 471–513 (Springer, 2022).

  281. Mehdipour, P., Chen, R. & De Carvalho, D. D. The next generation of DNMT inhibitors. Nat. Cancer 2, 1000–1001 (2021).

    Article  CAS  PubMed  Google Scholar 

  282. Wasserzug‐Pash, P. et al. Loss of heterochromatin and retrotransposon silencing as determinants in oocyte aging. Aging Cell 21, e13568 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  283. Mitchell, S. J. et al. The SIRT1 activator SRT1720 extends lifespan and improves health of mice fed a standard diet. Cell Rep. 6, 836–843 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  284. Pacholec, M. et al. SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1. J. Biol. Chem. 285, 8340–8351 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  285. Huber, J. L., McBurney, M. W., Distefano, P. S. & McDonagh, T. SIRT1-independent mechanisms of the putative sirtuin enzyme activators SRT1720 and SRT2183. Future Med. Chem. 2, 1751–1759 (2010).

    Article  CAS  PubMed  Google Scholar 

  286. Hubbard, B. P. et al. Evidence for a common mechanism of SIRT1 regulation by allosteric activators. Science 339, 1216–1219 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  287. Dai, H. et al. SIRT1 activation by small molecules: kinetic and biophysical evidence for direct interaction of enzyme and activator. J. Biol. Chem. 285, 32695–32703 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  288. Dai, H. et al. Crystallographic structure of a small molecule SIRT1 activator-enzyme complex. Nat. Commun. 6, 7645 (2015).

    Article  PubMed  Google Scholar 

  289. Mohrin, M. et al. Hematopoietic stem cell quiescence promotes error prone DNA repair and mutagenesis. Cell Stem Cell 7, 174–185 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  290. Milholland, B. et al. Differences between germline and somatic mutation rates in humans and mice. Nat. Commun. 8, 15183 (2017). This study determines that germline mutation rates in mice and humans are at least an order of magnitude lower than somatic mutation rates, indicating profound differences in genome maintenance mechanisms that allow indefinite survival of germline cells, in stark contrast to the single-generation maintenance of the soma.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  291. Ou, H.-L., Kim, C. S., Uszkoreit, S., Wickström, S. A. & Schumacher, B. Somatic niche cells regulate the CEP-1/p53-mediated DNA damage response in primordial germ cells. Dev. Cell 50, 167–183.e8 (2019).

    Article  CAS  PubMed  Google Scholar 

  292. Vermezovic, J., Stergiou, L., Hengartner, M. O. & d’Adda di Fagagna, F. Differential regulation of DNA damage response activation between somatic and germline cells in Caenorhabditis elegans. Cell Death Differ. 19, 1847–1855 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  293. Müller, G. A. & Engeland, K. The central role of CDE/CHR promoter elements in the regulation of cell cycle-dependent gene transcription. FEBS J. 277, 877–893 (2010).

    Article  PubMed  Google Scholar 

  294. Müller, G. A. et al. The CHR promoter element controls cell cycle-dependent gene transcription and binds the DREAM and MMB complexes. Nucleic Acids Res. 40, 1561–1578 (2012).

    Article  PubMed  Google Scholar 

  295. Schmit, F., Cremer, S. & Gaubatz, S. LIN54 is an essential core subunit of the DREAM/LINC complex that binds to the cdc2 promoter in a sequence-specific manner. FEBS J. 276, 5703–5716 (2009).

    Article  CAS  PubMed  Google Scholar 

  296. Marceau, A. H. et al. Structural basis for LIN54 recognition of CHR elements in cell cycle-regulated promoters. Nat. Commun. 7, 12301 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  297. Goetsch, P. D., Garrigues, J. M. & Strome, S. Loss of the Caenorhabditis elegans pocket protein LIN-35 reveals MuvB’s innate function as the repressor of DREAM target genes. PLoS Genet. 13, e1007088 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  298. Bujarrabal-Dueso, A. et al. The DREAM complex functions as conserved master regulator of somatic DNA-repair capacities. Nat. Struct. Mol. Biol. 30, 475–488 (2023). This study identifies the DREAM complex as a repressor of the expression of DNA repair genes of all the different repair pathways and shows that DREAM limits the DNA repair capacities of somatic cells. Genetic or pharmacological targeting of the DREAM complex could boost DNA repair and confer DNA damage resistance, providing a rationale for pharmacological improvement of genome stability.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  299. Litovchick, L. et al. Evolutionarily conserved multisubunit RBL2/p130 and E2F4 protein complex represses human cell cycle-dependent genes in quiescence. Mol. Cell 26, 539–551 (2007).

    Article  CAS  PubMed  Google Scholar 

  300. Korenjak, M. et al. Native E2F/RBF complexes contain Myb-interacting proteins and repress transcription of developmentally controlled E2F target genes. Cell 119, 181–193 (2004).

    Article  CAS  PubMed  Google Scholar 

  301. Harrison, M. M., Ceol, C. J., Lu, X. & Horvitz, H. R. Some C. Elegans class B synthetic multivulva proteins encode a conserved LIN-35 Rb-containing complex distinct from a NuRD-like complex. Proc. Natl Acad. Sci. USA 103, 16782–16787 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  302. Bessler, J. B., Andersen, E. C. & Villeneuve, A. M. Differential localization and independent acquisition of the H3K9me2 and H3K9me3 chromatin modifications in the Caenorhabditis elegans adult germ line. PLoS Genet. 6, e1000830 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  303. Gal, C. et al. DREAM represses distinct targets by cooperating with different THAP domain proteins. Cell Rep. 37, 109835 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  304. Latorre, I. et al. The DREAM complex promotes gene body H2A.Z for target repression. Genes Dev. 29, 495–500 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  305. Sadasivam, S. & DeCaprio, J. A. The DREAM complex: master coordinator of cell cycle-dependent gene expression. Nat. Rev. Cancer 13, 585–595 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  306. Kirienko, N. V. & Fay, D. S. Transcriptome profiling of the C. elegans Rb ortholog reveals diverse developmental roles. Dev. Biol. 305, 674–684 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  307. Engeland, K. Cell cycle arrest through indirect transcriptional repression by p53: I have a DREAM. Cell Death Differ. 25, 114–132 (2018).

    Article  CAS  PubMed  Google Scholar 

  308. Christmann, M. & Kaina, B. Transcriptional regulation of human DNA repair genes following genotoxic stress: trigger mechanisms, inducible responses and genotoxic adaptation. Nucleic Acids Res. 41, 8403–8420 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  309. Christmann, M. et al. Adaptive upregulation of DNA repair genes following benzo(a)pyrene diol epoxide protects against cell death at the expense of mutations. Nucleic Acids Res. 44, 10727–10743 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  310. Guiley, K. Z. et al. Structural mechanisms of DREAM complex assembly and regulation. Genes Dev. 29, 961–974 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  311. Pilkinton, M., Sandoval, R. & Colamonici, O. R. Mammalian Mip/LIN-9 interacts with either the p107, p130/E2F4 repressor complex or B-Myb in a cell cycle-phase-dependent context distinct from the Drosophila dREAM complex. Oncogene 26, 7535–7543 (2007).

    Article  CAS  PubMed  Google Scholar 

  312. Litovchick, L., Florens, L. A., Swanson, S. K., Washburn, M. P. & Decaprio, J. A. DYRK1A protein kinase promotes quiescence and senescence through DREAM complex assembly. Genes Dev. 25, 801–813 (2011). This work determines that the DYRK1A kinase regulates the DREAM complex assembly via phosphorylation of LIN52, thus providing a pharmacological target kinase for the development of DREAM inhibitors.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  313. Göckler, N. et al. Harmine specifically inhibits protein kinase DYRK1A and interferes with neurite formation. FEBS J. 276, 6324–6337 (2009).

    Article  PubMed  Google Scholar 

  314. Ogawa, Y. et al. Development of a novel selective inhibitor of the Down syndrome-related kinase Dyrk1A. Nat. Commun. 1, 86 (2010). Report of a selective inhibitor of DYRK1A that could overcome the limitations in the targeting of this kinase that is amplified in Down syndrome. Down syndrome displays many pathologies of premature ageing, suggesting therapeutic advances could also be applied to age-associated diseases.

    Article  PubMed  Google Scholar 

  315. Antonarakis, S. E. et al. Down syndrome. Nat. Rev. Dis. Primers 6, 9 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  316. Martin, G. M. Genetic syndromes in man with potential relevance to the pathobiology of aging. Birth Defects Orig. Artic. Ser. 14, 5–39 (1978).

    CAS  PubMed  Google Scholar 

  317. Franceschi, C. et al. Accelerated bio-cognitive aging in Down syndrome: state of the art and possible deceleration strategies. Aging Cell 18, e12903 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  318. Murray, A. et al. Dose imbalance of DYRK1A kinase causes systemic progeroid status in Down syndrome by increasing the un-repaired DNA damage and reducing LaminB1 levels. eBioMedicine 94, 104692 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  319. Chen, X.-Q. et al. Mechanistic analysis of age-related clinical manifestations in Down syndrome. Front. Aging Neurosci. 13, 700280 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  320. Horvath, S. et al. Accelerated epigenetic aging in Down syndrome. Aging Cell 14, 491–495 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  321. Peng, L., Baradar, A. A., Aguado, J. & Wolvetang, E. Cellular senescence and premature aging in Down syndrome. Mech. Ageing Dev. 212, 111824 (2023).

    Article  CAS  PubMed  Google Scholar 

  322. Komatsu, T. et al. Reactive oxygen species generation in gingival fibroblasts of Down syndrome patients detected by electron spin resonance spectroscopy. Redox Rep. 11, 71–77 (2006).

    Article  CAS  PubMed  Google Scholar 

  323. Busciglio, J. & Yankner, B. A. Apoptosis and increased generation of reactive oxygen species in Down’s syndrome neurons in vitro. Nature 378, 776–779 (1995).

    Article  CAS  PubMed  Google Scholar 

  324. Meharena, H. S. et al. Down-syndrome-induced senescence disrupts the nuclear architecture of neural progenitors. Cell Stem Cell 29, 116–130.e7 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  325. Murray, A. et al. Brief report: isogenic induced pluripotent stem cell lines from an adult with mosaic Down syndrome model accelerated neuronal ageing and neurodegeneration. Stem Cells 33, 2077–2084 (2015).

    Article  CAS  PubMed  Google Scholar 

  326. Del Bo, R. et al. Down’s syndrome fibroblasts anticipate the accumulation of specific ageing-related mtDNA mutations. Ann. Neurol. 49, 137–138 (2001).

    Article  PubMed  Google Scholar 

  327. Druzhyna, N., Nair, R. G., LeDoux, S. P. & Wilson, G. L. Defective repair of oxidative damage in mitochondrial DNA in Down’s syndrome. Mutat. Res. 409, 81–89 (1998).

    Article  CAS  PubMed  Google Scholar 

  328. Necchi, D. et al. Defective DNA repair and increased chromatin binding of DNA repair factors in Down syndrome fibroblasts. Mutat. Res. 780, 15–23 (2015).

    Article  CAS  PubMed  Google Scholar 

  329. Amiel, A., Goldzak, G., Gaber, E. & Fejgin, M. D. Molecular cytogenetic characteristics of Down syndrome newborns. J. Hum. Genet. 51, 541–547 (2006).

    Article  PubMed  Google Scholar 

  330. Morawiec, Z. et al. DNA damage and repair in children with Down’s syndrome. Mutat. Res. 637, 118–123 (2008).

    Article  CAS  PubMed  Google Scholar 

  331. Raji, N. S. & Rao, K. S. Trisomy 21 and accelerated aging: DNA-repair parameters in peripheral lymphocytes of Down’s syndrome patients. Mech. Ageing Dev. 100, 85–101 (1998).

    Article  CAS  PubMed  Google Scholar 

  332. Thomas, P., Harvey, S., Gruner, T. & Fenech, M. The buccal cytome and micronucleus frequency is substantially altered in Down’s syndrome and normal ageing compared to young healthy controls. Mutat. Res. 638, 37–47 (2008).

    Article  CAS  PubMed  Google Scholar 

  333. Zana, M. et al. Age-dependent oxidative stress-induced DNA damage in Down’s lymphocytes. Biochem. Biophys. Res. Commun. 345, 726–733 (2006).

    Article  CAS  PubMed  Google Scholar 

  334. Wu, C.-I. et al. APP and DYRK1A regulate axonal and synaptic vesicle protein networks and mediate Alzheimer’s pathology in trisomy 21 neurons. Mol. Psychiatry 27, 1970–1989 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  335. Sit, Y. T. et al. Synergistic roles of DYRK1A and GATA1 in trisomy 21 megakaryopoiesis. JCI Insight 8, e172851 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  336. Brault, V. et al. Dyrk1a gene dosage in glutamatergic neurons has key effects in cognitive deficits observed in mouse models of MRD7 and Down syndrome. PLoS Genet. 17, e1009777 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  337. de la Torre, R. et al. Safety and efficacy of cognitive training plus epigallocatechin-3-gallate in young adults with Down’s syndrome (TESDAD): a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet Neurol. 15, 801–810 (2016).

    Article  PubMed  Google Scholar 

  338. Lindberg, M. F. et al. Chemical, biochemical, cellular, and physiological characterization of Leucettinib-21, a Down syndrome and Alzheimer’s disease drug candidate. J. Med. Chem. 66, 15648–15670 (2023).

    Article  CAS  PubMed  Google Scholar 

  339. Liu, T. et al. DYRK1A inhibitors for disease therapy: current status and perspectives. Eur. J. Med. Chem. 229, 114062 (2022).

    Article  CAS  PubMed  Google Scholar 

  340. Biomarkers of Aging Consortium. et al. Challenges and recommendations for the translation of biomarkers of aging. Nat. Aging 4, 1372–1383 (2024).

    Article  PubMed Central  Google Scholar 

  341. Benzing, T. & Schumacher, B. Chronic kidney disease promotes ageing in a multiorgan disease network. Nat. Rev. Nephrol. 19, 542–543 (2023).

    Article  PubMed  Google Scholar 

  342. Huang, J. et al. Functional and multi-omic aging rejuvenation with GLP-1R agonism. Preprint at https://doi.org/10.1101/2024.05.06.592653 (2024).

  343. Partch, C. L. & Sancar, A. in Encyclopedia of Biological Chemistry (eds Lennarz, W. J. & Lane, M. D.) 698–702 (Elsevier, 2004).

  344. Maestre-Reyna, M. et al. Visualizing the DNA repair process by a photolyase at atomic resolution. Science 382, eadd7795 (2023).

    Article  CAS  PubMed  Google Scholar 

  345. Vassylyev, D. G. et al. Atomic model of a pyrimidine dimer excision repair enzyme complexed with a dna substrate: structural basis for damaged DNA recognition. Cell 83, 773–782 (1995).

    Article  CAS  PubMed  Google Scholar 

  346. Weismann, A. The Germ-plasm: a Theory of Heredity (Scribner’s, 1893).

  347. Medawar, P. B. An Unsolved Problem of Biology (H.K. Lewis and Co.,1952).

  348. Williams, G. C. Pleiotropy, natural selection, and the evolution of senescence. Evolution 11, 398–411 (1957).

    Article  Google Scholar 

  349. Campisi, J. Cellular senescence and apoptosis: how cellular responses might influence aging phenotypes. Exp. Gerontol. 38, 5–11 (2003).

    Article  CAS  PubMed  Google Scholar 

  350. Olshansky, S. J., Willcox, B. J., Demetrius, L. & Beltrán-Sánchez, H. Implausibility of radical life extension in humans in the twenty-first century. Nat. Aging 4, 1635–1642 (2024).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

G.A.G. and B.S. acknowledge funding from the Hevolution Foundation (HF-GRO-23-1199212-35) and the ERA Chair Program (101176529). J.V. acknowledges support from NIH grants (P01 AG017242, P01 AG047200, P30 AG038072, U01 ES029519, U01 HL145560, and U19 AG056278). P.D.R. acknowledges grants R01 AG069819, P01 AG043376, U19 AG056278, P01 AG062413, U54 AG079754, U54 AG076041, P01 AI172501, R01 AG076515, R01 AG081293, R01 AG069819 from the National Institutes of Health and the Aligning Science Across Parkinson’s (ASAP-000592) grant administered through the Michael J. Fox Foundation for Parkinson’s Research (MJFF). B.S. acknowledges funding from the European Research Council (ERC-2023-SyG, 101118919), the Deutsche Forschungsgemeinschaft (Reinhart Koselleck-Project 524088035, FOR 5504 project 496650118, FOR 5762 project 531902955, SFB 1678, SFB 1607, CECAD EXC 2030 – 390661388, ANR-DFG project 545378328, and the DFG project grants 558166204, 540136447, 496914708, 437825591, 437407415, 418036758), the José Carreras Leukaemia Foundation, DJCLS 04 R/2023, the Deutsche Krebshilfe (70114555) and the John Templeton Foundation Grant (61734). Figures were generated using Biorender.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to writing the manuscript. 

Corresponding author

Correspondence to Björn Schumacher.

Ethics declarations

Competing interests

J.V. is co-founder of Singulomics Inc. and Mutagentech Inc, and P.D.R. is co-founder of Genascence and Itasca Therapeutics. The other authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Drug Discovery thanks Ron Jachimowicz and the other, anonymous reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Ventus Therapeutics Announces Successful Completion of Phase 1 Clinical Trial of VENT-03, a First-in-Class, Orally Administered cGAS Inhibitor: https://www.ventustx.com/ventus-therapeutics-announces-successful-completion-of-phase-1-clinical-trial-of-vent-03-a-first-in-class-orally-administered-cgas-inhibitor/

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bujarrabal-Dueso, A., Garinis, G.A., Robbins, P.D. et al. Targeting DNA damage in ageing: towards supercharging DNA repair. Nat Rev Drug Discov 24, 785–807 (2025). https://doi.org/10.1038/s41573-025-01212-6

Download citation

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41573-025-01212-6

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing