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.

  • Review Article
  • Published:

Cellular senescence in ageing: from mechanisms to therapeutic opportunities

Subjects

Abstract

Cellular senescence, first described in vitro in 1961, has become a focus for biotech companies that target it to ameliorate a variety of human conditions. Eminently characterized by a permanent proliferation arrest, cellular senescence occurs in response to endogenous and exogenous stresses, including telomere dysfunction, oncogene activation and persistent DNA damage. Cellular senescence can also be a controlled programme occurring in diverse biological processes, including embryonic development. Senescent cell extrinsic activities, broadly related to the activation of a senescence-associated secretory phenotype, amplify the impact of cell-intrinsic proliferative arrest and contribute to impaired tissue regeneration, chronic age-associated diseases and organismal ageing. This Review discusses the mechanisms and modulators of cellular senescence establishment and induction of a senescence-associated secretory phenotype, and provides an overview of cellular senescence as an emerging opportunity to intervene through senolytic and senomorphic therapies in ageing and ageing-associated diseases.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Senescence drivers and phenotypes.
Fig. 2: SASP regulation.
Fig. 3: Biological consequences of cell senescence.
Fig. 4: Senolytic therapeutic interventions.
Fig. 5: Senomorphic therapeutic interventions.

Similar content being viewed by others

References

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

    CAS  PubMed  Google Scholar 

  2. Hayflick, L. The limited in vitro lifetime of human diploid cell strains. Exp. Cell. Res. 37, 614–636 (1965).

    CAS  PubMed  Google Scholar 

  3. Serrano, M., Hannon, G. J. & Beach, D. A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4. Nature 366, 704–707 (1993).

    CAS  PubMed  Google Scholar 

  4. Alcorta, D. A. et al. Involvement of the cyclin-dependent kinase inhibitor p16 (INK4a) in replicative senescence of normal human fibroblasts. Proc. Natl Acad. Sci. USA 93, 13742–13747 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Serrano, M. et al. Role of the INK4a locus in tumor suppression and cell mortality. Cell 85, 27–37 (1996).

    CAS  PubMed  Google Scholar 

  6. Serrano, M., Lin, A. W., McCurrach, M. E., Beach, D. & Lowe, S. W. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593–602 (1997).

    CAS  PubMed  Google Scholar 

  7. Di Micco, R. et al. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature 444, 638–642 (2006).

    PubMed  Google Scholar 

  8. Bartkova, J. et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 444, 633–637 (2006).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  10. Giaimo, S. & d’Adda di Fagagna, F. Is cellular senescence an example of antagonistic pleiotropy? Aging Cell 11, 378–383 (2012).

    CAS  PubMed  Google Scholar 

  11. Baker, D. J. et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479, 232–236 (2011). This study demonstrates that selective elimination of p16-expressing senescent cells is safe and capable of modulating age-related dysfunction in a premature aged mouse model.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Childs, B. G. et al. Senescent cells: an emerging target for diseases of ageing. Nat. Rev. Drug Discov. 16, 718–735 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Chang, J. et al. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat. Med. 22, 78–83 (2016).

    CAS  PubMed  Google Scholar 

  16. Childs, B. G., Baker, D. J., Kirkland, J. L., Campisi, J. & van Deursen, J. M. Senescence and apoptosis: dueling or complementary cell fates? EMBO Rep. 15, 1139–1153 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Kirschner, K. et al. Phenotype specific analyses reveal distinct regulatory mechanism for chronically activated p53. PLoS Genet. 11, e1005053 (2015).

    PubMed  PubMed Central  Google Scholar 

  18. Milanovic, M. et al. Senescence-associated reprogramming promotes cancer stemness. Nature 553, 96–100 (2018).

    CAS  PubMed  Google Scholar 

  19. Herranz, N. & Gil, J. Mechanisms and functions of cellular senescence. J. Clin. Invest. 128, 1238–1246 (2018).

    PubMed  PubMed Central  Google Scholar 

  20. Ovadya, Y. et al. Impaired immune surveillance accelerates accumulation of senescent cells and aging. Nat. Commun. 9, 5435 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Jackson, S. P. & Bartek, J. The DNA-damage response in human biology and disease. Nature 461, 1071–1078 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Fumagalli, M., Rossiello, F., Mondello, C. & d’Adda di Fagagna, F. Stable cellular senescence is associated with persistent DDR activation. PLoS ONE 9, e110969 (2014).

    PubMed  PubMed Central  Google Scholar 

  23. Galbiati, A., Beausejour, C. & d’Adda di Fagagna, F. A novel single-cell method provides direct evidence of persistent DNA damage in senescent cells and aged mammalian tissues. Aging Cell 16, 422–427 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. d’Adda di Fagagna, F. et al. A DNA damage checkpoint response in telomere-initiated senescence. Nature 426, 194–198 (2003). This study demonstrates that the activation of the DDR pathways by critically short telomeres is key in the enforcement of cellular senescence.

    PubMed  Google Scholar 

  25. Mallette, F. A. & Ferbeyre, G. The DNA damage signaling pathway connects oncogenic stress to cellular senescence. Cell Cycle 6, 1831–1836 (2007).

    CAS  PubMed  Google Scholar 

  26. Beausejour, C. M. et al. Reversal of human cellular senescence: roles of the p53 and p16 pathways. EMBO J. 22, 4212–4222 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Dulic, V., Beney, G. E., Frebourg, G., Drullinger, L. F. & Stein, G. H. Uncoupling between phenotypic senescence and cell cycle arrest in aging p21-deficient fibroblasts. Mol. Cell. Biol. 20, 6741–6754 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Kamijo, T. et al. Tumor suppression at the mouse INK4a locus mediated by the alternative reading frame product p19ARF. Cell 91, 649–659 (1997).

    CAS  PubMed  Google Scholar 

  29. Sherr, C. J. Divorcing ARF and p53: an unsettled case. Nat. Rev. Cancer 6, 663–673 (2006).

    CAS  PubMed  Google Scholar 

  30. Kamijo, T. et al. Loss of the ARF tumor suppressor reverses premature replicative arrest but not radiation hypersensitivity arising from disabled atm function. Cancer Res. 59, 2464–2469 (1999).

    CAS  PubMed  Google Scholar 

  31. Velimezi, G. et al. Functional interplay between the DNA-damage-response kinase ATM and ARF tumour suppressor protein in human cancer. Nat. Cell Biol. 15, 967–977 (2013).

    CAS  PubMed  Google Scholar 

  32. Herbig, U., Jobling, W. A., Chen, B. P., Chen, D. J. & Sedivy, J. M. Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21(CIP1), but not p16(INK4a). Mol. Cell 14, 501–513 (2004).

    CAS  PubMed  Google Scholar 

  33. Hemann, M. T., Strong, M. A., Hao, L. Y. & Greider, C. W. The shortest telomere, not average telomere length, is critical for cell viability and chromosome stability. Cell 107, 67–77 (2001).

    CAS  PubMed  Google Scholar 

  34. Bodnar, A. G. et al. Extension of life-span by introduction of telomerase into normal human cells. Science 279, 349–352 (1998).

    CAS  PubMed  Google Scholar 

  35. Fumagalli, M. et al. Telomeric DNA damage is irreparable and causes persistent DNA-damage-response activation. Nat. Cell Biol. 14, 355–365 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. 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).

    PubMed  Google Scholar 

  37. Bae, N. S. & Baumann, P. A RAP1/TRF2 complex inhibits nonhomologous end-joining at human telomeric DNA ends. Mol. Cell 26, 323–334 (2007).

    CAS  PubMed  Google Scholar 

  38. Rossiello, F. et al. DNA damage response inhibition at dysfunctional telomeres by modulation of telomeric DNA damage response RNAs. Nat. Commun. 8, 13980 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Halazonetis, T. D., Gorgoulis, V. G. & Bartek, J. An oncogene-induced DNA damage model for cancer development. Science 319, 1352–1355 (2008).

    CAS  PubMed  Google Scholar 

  40. Alimonti, A. et al. A novel type of cellular senescence that can be enhanced in mouse models and human tumor xenografts to suppress prostate tumorigenesis. J. Clin. Invest. 120, 681–693 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Parisotto, M. et al. PTEN deletion in luminal cells of mature prostate induces replication stress and senescence in vivo. J. Exp. Med. 215, 1749–1763 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Astle, M. V. et al. AKT induces senescence in human cells via mTORC1 and p53 in the absence of DNA damage: implications for targeting mTOR during malignancy. Oncogene 31, 1949–1962 (2012).

    CAS  PubMed  Google Scholar 

  43. Chan, K. T. et al. A functional genetic screen defines the AKT-induced senescence signaling network. Cell Death Differ. 27, 725–741 (2020).

    CAS  PubMed  Google Scholar 

  44. Suram, A. et al. Oncogene-induced telomere dysfunction enforces cellular senescence in human cancer precursor lesions. EMBO J. 31, 2839–2851 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Ogrunc, M. et al. Oncogene-induced reactive oxygen species fuel hyperproliferation and DNA damage response activation. Cell Death Differ. 21, 998–1012 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Chapman, J., Fielder, E. & Passos, J. F. Mitochondrial dysfunction and cell senescence: deciphering a complex relationship. FEBS Lett. 593, 1566–1579 (2019).

    CAS  PubMed  Google Scholar 

  47. Wiley, C. D. et al. Mitochondrial dysfunction induces senescence with a distinct secretory phenotype. Cell Metab. 23, 303–314 (2016).

    CAS  PubMed  Google Scholar 

  48. Correia-Melo, C. et al. Mitochondria are required for pro-ageing features of the senescent phenotype. EMBO J. 35, 724–742 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Narita, M. et al. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell 113, 703–716 (2003).

    CAS  PubMed  Google Scholar 

  50. Zhang, R. et al. Formation of MacroH2A-containing senescence-associated heterochromatin foci and senescence driven by ASF1a and HIRA. Dev. Cell 8, 19–30 (2005).

    CAS  PubMed  Google Scholar 

  51. Chandra, T. et al. Independence of repressive histone marks and chromatin compaction during senescent heterochromatic layer formation. Mol. Cell 47, 203–214 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Di Micco, R. et al. Interplay between oncogene-induced DNA damage response and heterochromatin in senescence and cancer. Nat. Cell Biol. 13, 292–302 (2011).

    PubMed  PubMed Central  Google Scholar 

  53. Zhang, R., Chen, W. & Adams, P. D. Molecular dissection of formation of senescence-associated heterochromatin foci. Mol. Cell. Biol. 27, 2343–2358 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Sadaie, M. et al. Redistribution of the lamin B1 genomic binding profile affects rearrangement of heterochromatic domains and SAHF formation during senescence. Genes Dev. 27, 1800–1808 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Samaraweera, L., Adomako, A., Rodriguez-Gabin, A. & McDaid, H. M. A novel indication for panobinostat as a senolytic drug in NSCLC and HNSCC. Sci. Rep. 7, 1900 (2017).

    PubMed  PubMed Central  Google Scholar 

  56. Munro, J., Barr, N. I., Ireland, H., Morrison, V. & Parkinson, E. K. Histone deacetylase inhibitors induce a senescence-like state in human cells by a p16-dependent mechanism that is independent of a mitotic clock. Exp. Cell. Res. 295, 525–538 (2004).

    CAS  PubMed  Google Scholar 

  57. Swanson, E. C., Manning, B., Zhang, H. & Lawrence, J. B. Higher-order unfolding of satellite heterochromatin is a consistent and early event in cell senescence. J. Cell Biol. 203, 929–942 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Ivanov, A. et al. Lysosome-mediated processing of chromatin in senescence. J. Cell Biol. 202, 129–143 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Dou, Z. et al. Autophagy mediates degradation of nuclear lamina. Nature 527, 105–109 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Dou, Z. et al. Cytoplasmic chromatin triggers inflammation in senescence and cancer. Nature 550, 402–406 (2017). This study demonstrates that cytoplasmic chromatin in senescent and cancer cells activates the innate immunity through the cGAS–STING pathway.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Tsantoulis, P. K. et al. Oncogene-induced replication stress preferentially targets common fragile sites in preneoplastic lesions. A genome-wide study. Oncogene 27, 3256–3264 (2008).

    CAS  PubMed  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  63. Vizioli, M. G. et al. Mitochondria-to-nucleus retrograde signaling drives formation of cytoplasmic chromatin and inflammation in senescence. Genes Dev. 34, 428–445 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Cruickshanks, H. A. et al. Senescent cells harbour features of the cancer epigenome. Nat. Cell Biol. 15, 1495–1506 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Xie, W. et al. DNA methylation patterns separate senescence from transformation potential and indicate cancer risk. Cancer Cell 33, 309–321 e305 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Parry, A. J. et al. NOTCH-mediated non-cell autonomous regulation of chromatin structure during senescence. Nat. Commun. 9, 1840 (2018).

    PubMed  PubMed Central  Google Scholar 

  67. De Cecco, M. et al. Genomes of replicatively senescent cells undergo global epigenetic changes leading to gene silencing and activation of transposable elements. Aging Cell 12, 247–256 (2013).

    PubMed  Google Scholar 

  68. Shah, P. P. et al. Lamin B1 depletion in senescent cells triggers large-scale changes in gene expression and the chromatin landscape. Genes Dev. 27, 1787–1799 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Freund, A., Laberge, R. M., Demaria, M. & Campisi, J. Lamin B1 loss is a senescence-associated biomarker. Mol. Biol. Cell 23, 2066–2075 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Shimi, T. et al. The role of nuclear lamin B1 in cell proliferation and senescence. Genes Dev. 25, 2579–2593 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Tasdemir, N. et al. BRD4 connects enhancer remodeling to senescence immune surveillance. Cancer Discov. 6, 612–629 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Sen, P. et al. Histone acetyltransferase p300 induces de novo super-enhancers to drive cellular senescence. Mol. Cell 73, 684–698 e688 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Acosta, J. C. et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat. Cell Biol. 15, 978–990 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Kuilman, T. et al. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell 133, 1019–1031 (2008).

    CAS  PubMed  Google Scholar 

  75. Coppe, J. P. et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 6, 2853–2868 (2008).

    CAS  PubMed  Google Scholar 

  76. Coppe, J. P., Desprez, P. Y., Krtolica, A. & Campisi, J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu. Rev. Pathol. 5, 99–118 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Coppe, J. P. et al. A human-like senescence-associated secretory phenotype is conserved in mouse cells dependent on physiological oxygen. PLoS ONE 5, e9188 (2010).

    PubMed  PubMed Central  Google Scholar 

  78. Eren, M. et al. PAI-1-regulated extracellular proteolysis governs senescence and survival in Klotho mice. Proc. Natl Acad. Sci. USA 111, 7090–7095 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Ozcan, S. et al. Unbiased analysis of senescence associated secretory phenotype (SASP) to identify common components following different genotoxic stresses. Aging 8, 1316–1329 (2016).

    PubMed  PubMed Central  Google Scholar 

  80. Basisty, N. et al. A proteomic atlas of senescence-associated secretomes for aging biomarker development. PLoS Biol. 18, e3000599 (2020).

    PubMed  PubMed Central  Google Scholar 

  81. Faget, D. V., Ren, Q. & Stewart, S. A. Unmasking senescence: context-dependent effects of SASP in cancer. Nat. Rev. Cancer 19, 439–453 (2019).

    CAS  PubMed  Google Scholar 

  82. Tanaka, T. et al. Plasma proteomic signature of age in healthy humans. Aging Cell 17, e12799 (2018).

    PubMed  PubMed Central  Google Scholar 

  83. Borghesan, M. et al. Small extracellular vesicles are key regulators of non-cell autonomous intercellular communication in senescence via the interferon protein IFITM3. Cell Rep. 27, 3956–3971 e3956 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Jakhar, R. & Crasta, K. Exosomes as emerging pro-tumorigenic mediators of the senescence-associated secretory phenotype. Int. J. Mol. Sci. https://doi.org/10.3390/ijms20102547 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  85. Coppe, J. P. et al. Tumor suppressor and aging biomarker p16(INK4a) induces cellular senescence without the associated inflammatory secretory phenotype. J. Biol. Chem. 286, 36396–36403 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Rodier, F. et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat. Cell Biol. 11, 973–979 (2009). This study demonstrates that persistent DDR activation controls SASP induction in senescent cells.

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Chen, H. et al. MacroH2A1 and ATM play opposing roles in paracrine senescence and the senescence-associated secretory phenotype. Mol. Cell 59, 719–731 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Freund, A., Patil, C. K. & Campisi, J. p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype. EMBO J. 30, 1536–1548 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. White, R. R., Vijg, J. & Do, D. N. A. Double-strand breaks drive aging? Mol. Cell 63, 729–738 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Ou, H. L. & Schumacher, B. DNA damage responses and p53 in the aging process. Blood 131, 488–495 (2018).

    CAS  PubMed  Google Scholar 

  91. Gorbunova, V. & Seluanov, A. DNA double strand break repair, aging and the chromatin connection. Mutat. Res. 788, 2–6 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Shmulevich, R. & Krizhanovsky, V. Cell senescence, DNA damage, and metabolism. Antioxid. Redox Signal. https://doi.org/10.1089/ars.2020.8043 (2020).

    Article  PubMed  Google Scholar 

  93. Acosta, J. C. et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell 133, 1006–1018 (2008).

    CAS  PubMed  Google Scholar 

  94. Hoare, M. et al. NOTCH1 mediates a switch between two distinct secretomes during senescence. Nat. Cell Biol. 18, 979–992 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Kang, C. et al. The DNA damage response induces inflammation and senescence by inhibiting autophagy of GATA4. Science 349, aaa5612 (2015).

    PubMed  PubMed Central  Google Scholar 

  96. Mazzucco, A. E. et al. Genetic interrogation of replicative senescence uncovers a dual role for USP28 in coordinating the p53 and GATA4 branches of the senescence program. Genes Dev. 31, 1933–1938 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Orjalo, A. V., Bhaumik, D., Gengler, B. K., Scott, G. K. & Campisi, J. Cell surface-bound IL-1alpha is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network. Proc. Natl Acad. Sci. USA 106, 17031–17036 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Toso, A. et al. Enhancing chemotherapy efficacy in Pten-deficient prostate tumors by activating the senescence-associated antitumor immunity. Cell Rep. 9, 75–89 (2014).

    CAS  PubMed  Google Scholar 

  99. Xu, M. et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc. Natl Acad. Sci. USA 112, E6301–E6310 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Hernandez-Segura, A. et al. Unmasking transcriptional heterogeneity in senescent cells. Curr. Biol. 27, 2652–2660 e2654 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Vermezovic, J. et al. Notch is a direct negative regulator of the DNA-damage response. Nat. Struct. Mol. Biol. 22, 417–424 (2015).

    CAS  PubMed  Google Scholar 

  102. Adamowicz, M., Vermezovic, J. & d’Adda di Fagagna, F. NOTCH1 inhibits activation of ATM by impairing the formation of an ATM-FOXO3a-KAT5/Tip60 complex. Cell Rep. 16, 2068–2076 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Stathis, A. & Bertoni, F. BET proteins as targets for anticancer treatment. Cancer Discov. 8, 24–36 (2018).

    CAS  PubMed  Google Scholar 

  104. Wakita, M. et al. A BET family protein degrader provokes senolysis by targeting NHEJ and autophagy in senescent cells. Nat. Commun. 11, 1935 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Wang, S. et al. BRD4 inhibitors block telomere elongation. Nucleic Acids Res. 45, 8403–8410 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. 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. 22, 3480–3492 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Capell, B. C. et al. MLL1 is essential for the senescence-associated secretory phenotype. Genes Dev. 30, 321–336 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Aird, K. M. et al. HMGB2 orchestrates the chromatin landscape of senescence-associated secretory phenotype gene loci. J. Cell Biol. 215, 325–334 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Huang, J. et al. DAMPs, ageing, and cancer: the ‘DAMP hypothesis’. Ageing Res. Rev. 24, 3–16 (2015).

    CAS  PubMed  Google Scholar 

  110. Davalos, A. R. et al. p53-dependent release of alarmin HMGB1 is a central mediator of senescent phenotypes. J. Cell Biol. 201, 613–629 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Boumendil, C., Hari, P., Olsen, K. C. F., Acosta, J. C. & Bickmore, W. A. Nuclear pore density controls heterochromatin reorganization during senescence. Genes Dev. 33, 144–149 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Gluck, S. et al. Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence. Nat. Cell Biol. 19, 1061–1070 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Yang, H., Wang, H., Ren, J., Chen, Q. & Chen, Z. J. cGAS is essential for cellular senescence. Proc. Natl Acad. Sci. USA 114, E4612–E4620 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Hopfner, K. P. & Hornung, V. Molecular mechanisms and cellular functions of cGAS-STING signalling. Nat. Rev. Mol. Cell Biol. 21, 501–521 (2020).

    CAS  PubMed  Google Scholar 

  115. Takahashi, A. et al. Downregulation of cytoplasmic DNases is implicated in cytoplasmic DNA accumulation and SASP in senescent cells. Nat. Commun. 9, 1249 (2018).

    PubMed  PubMed Central  Google Scholar 

  116. Haag, S. M. et al. Targeting STING with covalent small-molecule inhibitors. Nature 559, 269–273 (2018).

    CAS  PubMed  Google Scholar 

  117. Broz, P. & Dixit, V. M. Inflammasomes: mechanism of assembly, regulation and signalling. Nat. Rev. Immunol. 16, 407–420 (2016).

    CAS  PubMed  Google Scholar 

  118. Hari, P. et al. The innate immune sensor Toll-like receptor 2 controls the senescence-associated secretory phenotype. Sci. Adv. 5, eaaw0254 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Simon, M. et al. LINE1 derepression in aged wild-type and SIRT6-deficient mice drives inflammation. Cell Metab. 29, 871–885 e875 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Warren, L. A. & Rossi, D. J. Stem cells and aging in the hematopoietic system. Mech. Ageing Dev. 130, 46–53 (2009).

    CAS  PubMed  Google Scholar 

  121. Rossi, D. J. et al. Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature 447, 725–729 (2007).

    CAS  PubMed  Google Scholar 

  122. McNeely, T., Leone, M., Yanai, H. & Beerman, I. DNA damage in aging, the stem cell perspective. Hum. Genet. https://doi.org/10.1007/s00439-019-02047-z (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  123. Sperka, T., Wang, J. & Rudolph, K. L. DNA damage checkpoints in stem cells, ageing and cancer. Nat. Rev. Mol. Cell Biol. 13, 579–590 (2012).

    CAS  PubMed  Google Scholar 

  124. Inomata, K. et al. Genotoxic stress abrogates renewal of melanocyte stem cells by triggering their differentiation. Cell 137, 1088–1099 (2009). This study demonstrates that DNA damage can induce stem cell differentiation.

    CAS  PubMed  Google Scholar 

  125. Wang, J. et al. A differentiation checkpoint limits hematopoietic stem cell self-renewal in response to DNA damage. Cell 158, 1444 (2014).

    CAS  PubMed  Google Scholar 

  126. Schiroli, G. et al. Precise gene editing preserves hematopoietic stem cell function following transient p53-mediated DNA damage response. Cell Stem Cell 24, 551–565 e558 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Flach, J. et al. Replication stress is a potent driver of functional decline in ageing haematopoietic stem cells. Nature 512, 198–202 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Schneider, L. et al. DNA damage in mammalian neural stem cells leads to astrocytic differentiation mediated by BMP2 signaling through JAK-STAT. Stem Cell Rep. 1, 123–138 (2013).

    CAS  Google Scholar 

  129. Zou, Y. et al. Responses of human embryonic stem cells and their differentiated progeny to ionizing radiation. Biochem. Biophys. Res. Commun. 426, 100–105 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Li, M. et al. Distinct regulatory mechanisms and functions for p53-activated and p53-repressed DNA damage response genes in embryonic stem cells. Mol. Cell 46, 30–42 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Storer, M. et al. Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell 155, 1119–1130 (2013).

    CAS  PubMed  Google Scholar 

  132. Munoz-Espin, D. et al. Programmed cell senescence during mammalian embryonic development. Cell 155, 1104–1118 (2013).

    CAS  PubMed  Google Scholar 

  133. Sapieha, P. & Mallette, F. A. Cellular senescence in postmitotic cells: beyond growth arrest. Trends Cell Biol. 28, 595–607 (2018).

    CAS  PubMed  Google Scholar 

  134. Jurk, D. et al. Postmitotic neurons develop a p21-dependent senescence-like phenotype driven by a DNA damage response. Aging Cell 11, 996–1004 (2012).

    CAS  PubMed  Google Scholar 

  135. Ogrodnik, M. et al. Obesity-induced cellular senescence drives anxiety and impairs neurogenesis. Cell Metab. 29, 1061–1077.e1068 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Musi, N. et al. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell 17, e12840 (2018).

    PubMed  PubMed Central  Google Scholar 

  137. Oubaha, M. et al. Senescence-associated secretory phenotype contributes to pathological angiogenesis in retinopathy. Sci. Transl Med. 8, 362ra144 (2016).

    PubMed  Google Scholar 

  138. Minamino, T. et al. A crucial role for adipose tissue p53 in the regulation of insulin resistance. Nat. Med. 15, 1082–1087 (2009).

    CAS  PubMed  Google Scholar 

  139. Farr, J. N. et al. Targeting cellular senescence prevents age-related bone loss in mice. Nat. Med. 23, 1072–1079 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Anderson, R. et al. Length-independent telomere damage drives post-mitotic cardiomyocyte senescence. EMBO J. https://doi.org/10.15252/embj.2018100492 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  141. Burton, D. G. & Krizhanovsky, V. Physiological and pathological consequences of cellular senescence. Cell. Mol. Life Sci. 71, 4373–4386 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Chuprin, A. et al. Cell fusion induced by ERVWE1 or measles virus causes cellular senescence. Genes Dev. 27, 2356–2366 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Gal, H. et al. Molecular pathways of senescence regulate placental structure and function. EMBO J. 38, e100849 (2019).

    PubMed  PubMed Central  Google Scholar 

  144. Davaapil, H., Brockes, J. P. & Yun, M. H. Conserved and novel functions of programmed cellular senescence during vertebrate development. Development 144, 106–114 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Krizhanovsky, V. et al. Senescence of activated stellate cells limits liver fibrosis. Cell 134, 657–667 (2008). This study demonstrates that immune cell targeting of senescent cells limits dysfunction in the liver.

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Kim, K. H., Chen, C. C., Monzon, R. I. & Lau, L. F. Matricellular protein CCN1 promotes regression of liver fibrosis through induction of cellular senescence in hepatic myofibroblasts. Mol. Cell. Biol. 33, 2078–2090 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Jun, J. I. & Lau, L. F. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nat. Cell Biol. 12, 676–685 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Fitzner, B. et al. Senescence determines the fate of activated rat pancreatic stellate cells. J. Cell Mol. Med. 16, 2620–2630 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Da Silva-Álvarez, S. et al. Cell senescence contributes to tissue regeneration in zebrafish. Aging Cell 19, e13052 (2020).

    PubMed  Google Scholar 

  150. Yun, M. H., Davaapil, H. & Brockes, J. P. Recurrent turnover of senescent cells during regeneration of a complex structure. eLife https://doi.org/10.7554/eLife.05505 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  151. Sagiv, A. & Krizhanovsky, V. Immunosurveillance of senescent cells: the bright side of the senescence program. Biogerontology 14, 617–628 (2013).

    CAS  PubMed  Google Scholar 

  152. Freund, A., Orjalo, A. V., Desprez, P. Y. & Campisi, J. Inflammatory networks during cellular senescence: causes and consequences. Trends Mol. Med. 16, 238–246 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Lujambio, A. et al. Non-cell-autonomous tumor suppression by p53. Cell 153, 449–460 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Soriani, A. et al. ATM-ATR-dependent up-regulation of DNAM-1 and NKG2D ligands on multiple myeloma cells by therapeutic agents results in enhanced NK-cell susceptibility and is associated with a senescent phenotype. Blood 113, 3503–3511 (2009).

    CAS  PubMed  Google Scholar 

  155. Xue, W. et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 445, 656–660 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Kang, T. W. et al. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature 479, 547–551 (2011).

    CAS  PubMed  Google Scholar 

  157. Sagiv, A. et al. NKG2D ligands mediate immunosurveillance of senescent cells. Aging 8, 328–344 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Biran, A. et al. Senescent cells communicate via intercellular protein transfer. Genes Dev. 29, 791–802 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Ruscetti, M. et al. NK cell-mediated cytotoxicity contributes to tumor control by a cytostatic drug combination. Science 362, 1416–1422 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Di Mitri, D. et al. Tumour-infiltrating Gr-1+ myeloid cells antagonize senescence in cancer. Nature 515, 134–137 (2014).

    PubMed  Google Scholar 

  161. Eggert, T. et al. Distinct functions of senescence-associated immune responses in liver tumor surveillance and tumor progression. Cancer Cell 30, 533–547 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Yoshimoto, S. et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 499, 97–101 (2013).

    CAS  PubMed  Google Scholar 

  163. Gonzalez-Meljem, J. M. et al. Stem cell senescence drives age-attenuated induction of pituitary tumours in mouse models of paediatric craniopharyngioma. Nat. Commun. 8, 1819 (2017).

    PubMed  PubMed Central  Google Scholar 

  164. Lau, L., Porciuncula, A., Yu, A., Iwakura, Y. & David, G. Uncoupling the senescence-associated secretory phenotype from cell cycle exit via interleukin-1 inactivation unveils its protumorigenic role. Mol. Cell. Biol. 39, e00586-18 (2019).

    PubMed  PubMed Central  Google Scholar 

  165. Demaria, M. et al. Cellular senescence promotes adverse effects of chemotherapy and cancer relapse. Cancer Discov. 7, 165–176 (2017).

    CAS  PubMed  Google Scholar 

  166. Franceschi, C. et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann. N. Y. Acad. Sci. 908, 244–254 (2000).

    CAS  PubMed  Google Scholar 

  167. Jeck, W. R., Siebold, A. P. & Sharpless, N. E. Review: a meta-analysis of GWAS and age-associated diseases. Aging Cell 11, 727–731 (2012).

    CAS  PubMed  Google Scholar 

  168. Melzer, D. Genetic polymorphisms and human aging: association studies deliver. Rejuvenation Res. 11, 523–526 (2008).

    CAS  PubMed  Google Scholar 

  169. Melzer, D. et al. A common variant of the p16(INK4a) genetic region is associated with physical function in older people. Mech. Ageing Dev. 128, 370–377 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Johnson, S. C., Dong, X., Vijg, J. & Suh, Y. Genetic evidence for common pathways in human age-related diseases. Aging Cell 14, 809–817 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. D’Mello, M. J. et al. Association between shortened leukocyte telomere length and cardiometabolic outcomes: systematic review and meta-analysis. Circ. Cardiovasc. Genet. 8, 82–90 (2015).

    PubMed  Google Scholar 

  172. Benetos, A. et al. Tracking and fixed ranking of leukocyte telomere length across the adult life course. Aging Cell 12, 615–621 (2013).

    CAS  PubMed  Google Scholar 

  173. Fabbri, E. et al. Aging and the burden of multimorbidity: associations with inflammatory and anabolic hormonal biomarkers. J. Gerontol. A Biol. Sci. Med. Sci 70, 63–70 (2015).

    CAS  PubMed  Google Scholar 

  174. Xu, M. et al. Senolytics improve physical function and increase lifespan in old age. Nat. Med. 24, 1246–1256 (2018). This study demonstrates that transplanting senescent cells in young mice causes persistent physical dysfunction that can be alleviated by senolytic drugs.

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Childs, B. G. et al. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science 354, 472–477 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Holdt, L. M. et al. Expression of Chr9p21 genes CDKN2B (p15(INK4b)), CDKN2A (p16(INK4a), p14(ARF)) and MTAP in human atherosclerotic plaque. Atherosclerosis 214, 264–270 (2011).

    CAS  PubMed  Google Scholar 

  177. Cavalli, G., Biavasco, R., Borgiani, B. & Dagna, L. Oncogene-induced senescence as a new mechanism of disease: the paradigm of erdheim-chester disease. Front. Immunol. 5, 281 (2014).

    PubMed  PubMed Central  Google Scholar 

  178. Cangi, M. G. et al. BRAFV600E-mutation is invariably present and associated to oncogene-induced senescence in Erdheim-Chester disease. Ann. Rheum. Dis. 74, 1596–1602 (2015).

    CAS  PubMed  Google Scholar 

  179. Sousa-Victor, P., Garcia-Prat, L., Serrano, A. L., Perdiguero, E. & Munoz-Canoves, P. Muscle stem cell aging: regulation and rejuvenation. Trends Endocrinol. Metab. 26, 287–296 (2015).

    CAS  PubMed  Google Scholar 

  180. Gnani, D. et al. An early-senescence state in aged mesenchymal stromal cells contributes to hematopoietic stem and progenitor cell clonogenic impairment through the activation of a pro-inflammatory program. Aging Cell 18, e12933 (2019).

    PubMed  PubMed Central  Google Scholar 

  181. Krizhanovsky, V. & Lowe, S. W. Stem cells: The promises and perils of p53. Nature 460, 1085–1086 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Banito, A. et al. Senescence impairs successful reprogramming to pluripotent stem cells. Genes Dev. 23, 2134–2139 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Li, H. et al. The Ink4/Arf locus is a barrier for iPS cell reprogramming. Nature 460, 1136–1139 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Mosteiro, L. et al. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Science https://doi.org/10.1126/science.aaf4445 (2016).

    Article  PubMed  Google Scholar 

  185. Wajapeyee, N., Serra, R. W., Zhu, X., Mahalingam, M. & Green, M. R. Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7. Cell 132, 363–374 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Takasugi, M. et al. Small extracellular vesicles secreted from senescent cells promote cancer cell proliferation through EphA2. Nat. Commun. 8, 15729 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Katlinskaya, Y. V. et al. Suppression of type I interferon signaling overcomes oncogene-induced senescence and mediates melanoma development and progression. Cell Rep. 15, 171–180 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Bird, T. G. et al. TGFbeta inhibition restores a regenerative response in acute liver injury by suppressing paracrine senescence. Sci. Transl Med. https://doi.org/10.1126/scitranslmed.aan1230 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  189. Ritschka, B. et al. The senescence-associated secretory phenotype induces cellular plasticity and tissue regeneration. Genes Dev. 31, 172–183 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Chiche, A. et al. Injury-induced senescence enables in vivo reprogramming in skeletal muscle. Cell Stem Cell 20, 407–414.e404 (2017).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  192. Baker, D. J., Jin, F. & van Deursen, J. M. The yin and yang of the Cdkn2a locus in senescence and aging. Cell Cycle 7, 2795–2802 (2008).

    CAS  PubMed  Google Scholar 

  193. Baker, D. J. et al. Opposing roles for p16Ink4a and p19Arf in senescence and ageing caused by BubR1 insufficiency. Nat. Cell Biol. 10, 825–836 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Baker, D. J. et al. Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature 530, 184–189 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Laberge, R. M. et al. Mitochondrial DNA damage induces apoptosis in senescent cells. Cell Death Dis. 4, e727 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Chinta, S. J. et al. Cellular senescence is induced by the environmental neurotoxin paraquat and contributes to neuropathology linked to Parkinson’s disease. Cell Rep. 22, 930–940 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Bussian, T. J. et al. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 562, 578–582 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  198. Zhang, P. et al. Senolytic therapy alleviates Aβ-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer’s disease model. Nat. Neurosci. 22, 719–728 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Schafer, M. J. et al. Cellular senescence mediates fibrotic pulmonary disease. Nat. Commun. 8, 14532 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  200. Sagiv, A. et al. p53 in bronchial club cells facilitates chronic lung inflammation by promoting senescence. Cell Rep. 22, 3468–3479 (2018).

    CAS  PubMed  Google Scholar 

  201. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  203. Guerrero, A. et al. Cardiac glycosides are broad-spectrum senolytics. Nat. Metab. 1, 1074–1088 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Baar, M. P. et al. Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell 169, 132–147 e116 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  207. Zhu, Y. et al. Inflammation and the depot-specific secretome of human preadipocytes. Obesity 23, 989–999 (2015).

    CAS  PubMed  Google Scholar 

  208. Roos, C. M. et al. Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell 15, 973–977 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  210. Zhu, Y. et al. The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell 14, 644–658 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  211. 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).

    PubMed  PubMed Central  Google Scholar 

  212. Justice, J. N. et al. Senolytics in idiopathic pulmonary fibrosis: results from a first-in-human, open-label, pilot study. EBioMedicine 40, 554–563 (2019).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  217. Ozsvari, B., Nuttall, J. R., Sotgia, F. & Lisanti, M. P. Azithromycin and roxithromycin define a new family of “senolytic” drugs that target senescent human fibroblasts. Aging 10, 3294–3307 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  218. Munoz-Espin, D. et al. A versatile drug delivery system targeting senescent cells. EMBO Mol. Med. https://doi.org/10.15252/emmm.201809355 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  220. González-Gualda, E. et al. Galacto-conjugation of navitoclax as an efficient strategy to increase senolytic specificity and reduce platelet toxicity. Aging Cell 19, e13142 (2020).

    PubMed  PubMed Central  Google Scholar 

  221. Ovadya, Y. & Krizhanovsky, V. Strategies targeting cellular senescence. J. Clin. Invest. 128, 1247–1254 (2018).

    PubMed  PubMed Central  Google Scholar 

  222. McMichael, E. L. et al. IL-21 enhances natural killer cell response to cetuximab-coated pancreatic tumor cells. Clin. Cancer Res. 23, 489–502 (2017).

    CAS  PubMed  Google Scholar 

  223. Brady, J. et al. The interactions of multiple cytokines control NK cell maturation. J. Immunol. 185, 6679–6688 (2010).

    CAS  PubMed  Google Scholar 

  224. Elpek, K. G., Rubinstein, M. P., Bellemare-Pelletier, A., Goldrath, A. W. & Turley, S. J. Mature natural killer cells with phenotypic and functional alterations accumulate upon sustained stimulation with IL-15/IL-15Ralpha complexes. Proc. Natl Acad. Sci. USA 107, 21647–21652 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  225. Rautela, J. & Huntington, N. D. IL-15 signaling in NK cell cancer immunotherapy. Curr. Opin. Immunol. 44, 1–6 (2017).

    CAS  PubMed  Google Scholar 

  226. Amor, C. et al. Senolytic CAR T cells reverse senescence-associated pathologies. Nature 583, 127–132 (2020). This study demonstrates the feasibility of targeting senescent cells in a variety of conditions by chimeric antigen receptor T cell-mediated therapy.

    CAS  PubMed  PubMed Central  Google Scholar 

  227. Frescas, D. et al. Senescent cells expose and secrete an oxidized form of membrane-bound vimentin as revealed by a natural polyreactive antibody. Proc. Natl Acad. Sci. USA 114, E1668–E1677 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  228. Kim, K. M. et al. Identification of senescent cell surface targetable protein DPP4. Genes Dev. 31, 1529–1534 (2017).

    PubMed  PubMed Central  Google Scholar 

  229. Althubiti, M. et al. Characterization of novel markers of senescence and their prognostic potential in cancer. Cell Death Dis. 5, e1528 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  230. Herranz, N. et al. mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype. Nat. Cell Biol. 17, 1205–1217 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  231. Laberge, R. M. et al. MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat. Cell Biol. 17, 1049–1061 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  232. Zhang, B. et al. The senescence-associated secretory phenotype is potentiated by feedforward regulatory mechanisms involving Zscan4 and TAK1. Nat. Commun. 9, 1723 (2018).

    PubMed  PubMed Central  Google Scholar 

  233. Thapa, R. K. et al. Progressive slowdown/prevention of cellular senescence by CD9-targeted delivery of rapamycin using lactose-wrapped calcium carbonate nanoparticles. Sci. Rep. 7, 43299 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  234. Harrison, D. E. et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 460, 392–395 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  235. Moiseeva, O. et al. Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-kappaB activation. Aging Cell 12, 489–498 (2013).

    CAS  PubMed  Google Scholar 

  236. Maruthur, N. M. et al. Diabetes medications as monotherapy or metformin-based combination therapy for type 2 diabetes: a systematic review and meta-analysis. Ann. Intern. Med. 164, 740–751 (2016).

    PubMed  Google Scholar 

  237. Noren Hooten, N. et al. Metformin-mediated increase in DICER1 regulates microRNA expression and cellular senescence. Aging Cell 15, 572–581 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  238. Lim, H., Park, H. & Kim, H. P. Effects of flavonoids on senescence-associated secretory phenotype formation from bleomycin-induced senescence in BJ fibroblasts. Biochem. Pharmacol. 96, 337–348 (2015).

    CAS  PubMed  Google Scholar 

  239. Nacarelli, T. et al. NAD+ metabolism governs the proinflammatory senescence-associated secretome. Nat. Cell Biol. 21, 397–407 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  240. Liu, S. et al. Simvastatin suppresses breast cancer cell proliferation induced by senescent cells. Sci. Rep. 5, 17895 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  241. Laberge, R. M. et al. Glucocorticoids suppress selected components of the senescence-associated secretory phenotype. Aging Cell 11, 569–578 (2012).

    CAS  PubMed  Google Scholar 

  242. Cohen, S. et al. Treatment of rheumatoid arthritis with anakinra, a recombinant human interleukin-1 receptor antagonist, in combination with methotrexate: results of a twenty-four-week, multicenter, randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 46, 614–624 (2002).

    CAS  PubMed  Google Scholar 

  243. Kuemmerle-Deschner, J. B. et al. Canakinumab (ACZ885, a fully human IgG1 anti-IL-1β mAb) induces sustained remission in pediatric patients with cryopyrin-associated periodic syndrome (CAPS). Arthritis Res. Ther. 13, R34 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  244. Hoffman, H. M. et al. Efficacy and safety of rilonacept (interleukin-1 trap) in patients with cryopyrin-associated periodic syndromes: results from two sequential placebo-controlled studies. Arthritis Rheum. 58, 2443–2452 (2008).

    CAS  PubMed  Google Scholar 

  245. Klareskog, L. et al. Therapeutic effect of the combination of etanercept and methotrexate compared with each treatment alone in patients with rheumatoid arthritis: double-blind randomised controlled trial. Lancet 363, 675–681 (2004).

    CAS  PubMed  Google Scholar 

  246. Jobanputra, P., Barton, P., Bryan, S. & Burls, A. The effectiveness of infliximab and etanercept for the treatment of rheumatoid arthritis: a systematic review and economic evaluation. Health Technol. Assess. 6, 1–110 (2002).

    CAS  PubMed  Google Scholar 

  247. Emery, P. et al. IL-6 receptor inhibition with tocilizumab improves treatment outcomes in patients with rheumatoid arthritis refractory to anti-tumour necrosis factor biologicals: results from a 24-week multicentre randomised placebo-controlled trial. Ann. Rheum. Dis. 67, 1516–1523 (2008).

    CAS  PubMed  Google Scholar 

  248. van Rhee, F. et al. Siltuximab for multicentric Castleman’s disease: a randomised, double-blind, placebo-controlled trial. Lancet Oncol. 15, 966–974 (2014).

    PubMed  Google Scholar 

  249. Harrison, D. E. et al. Acarbose, 17-α-estradiol, and nordihydroguaiaretic acid extend mouse lifespan preferentially in males. Aging Cell 13, 273–282 (2014).

    CAS  PubMed  Google Scholar 

  250. Michelini, F. et al. Damage-induced lncRNAs control the DNA damage response through interaction with DDRNAs at individual double-strand breaks. Nat. Cell Biol. 19, 1400–1411 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  251. Aguado, J. et al. Inhibition of DNA damage response at telomeres improves the detrimental phenotypes of Hutchinson-Gilford progeria syndrome. Nat. Commun. 10, 4990 (2019).

    PubMed  PubMed Central  Google Scholar 

  252. Hall, B. M. et al. Aging of mice is associated with p16(Ink4a)- and beta-galactosidase-positive macrophage accumulation that can be induced in young mice by senescent cells. Aging 8, 1294–1315 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  253. Liu, J. Y. et al. Cells exhibiting strong p16 (INK4a) promoter activation in vivo display features of senescence. Proc. Natl Acad. Sci. USA 116, 2603–2611 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  254. Grosse, L. et al. Defined p16(High) senescent cell types are indispensable for mouse healthspan. Cell Metab. https://doi.org/10.1016/j.cmet.2020.05.002 (2020).

    Article  PubMed  Google Scholar 

  255. Fontana, L., Partridge, L. & Longo, V. D. Extending healthy life span–from yeast to humans. Science 328, 321–326 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  256. Krishnamurthy, J. et al. Ink4a/Arf expression is a biomarker of aging. J. Clin. Invest. 114, 1299–1307 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  257. Fontana, L. et al. The effects of graded caloric restriction: XII. Comparison of mouse to human impact on cellular senescence in the colon. Aging Cell 17, e12746 (2018).

    PubMed  PubMed Central  Google Scholar 

  258. Wang, C. et al. Adult-onset, short-term dietary restriction reduces cell senescence in mice. Aging 2, 555–566 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  259. Satyanarayana, A. et al. Mitogen stimulation cooperates with telomere shortening to activate DNA damage responses and senescence signaling. Mol. Cell. Biol. 24, 5459–5474 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  260. Martinez, P. & Blasco, M. A. Telomere-driven diseases and telomere-targeting therapies. J. Cell Biol. 216, 875–887 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  261. Salvador, L. et al. A natural product telomerase activator lengthens telomeres in humans: a randomized, double blind, and placebo controlled study. Rejuvenation Res. 19, 478–484 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  262. Bar, C., Huber, N., Beier, F. & Blasco, M. A. Therapeutic effect of androgen therapy in a mouse model of aplastic anemia produced by short telomeres. Haematologica 100, 1267–1274 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  263. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  264. Alvarez, D. et al. IPF lung fibroblasts have a senescent phenotype. Am. J. Physiol. Lung Cell Mol. Physiol. 313, L1164–L1173 (2017).

    PubMed  PubMed Central  Google Scholar 

  265. Povedano, J. M. et al. Therapeutic effects of telomerase in mice with pulmonary fibrosis induced by damage to the lungs and short telomeres. eLife https://doi.org/10.7554/eLife.31299 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  266. Munoz-Lorente, M. A. et al. AAV9-mediated telomerase activation does not accelerate tumorigenesis in the context of oncogenic K-Ras-induced lung cancer. PLoS Genet. 14, e1007562 (2018).

    PubMed  PubMed Central  Google Scholar 

  267. Conboy, M. J., Conboy, I. M. & Rando, T. A. Heterochronic parabiosis: historical perspective and methodological considerations for studies of aging and longevity. Aging Cell 12, 525–530 (2013).

    CAS  PubMed  Google Scholar 

  268. Castellano, J. M. et al. Human umbilical cord plasma proteins revitalize hippocampal function in aged mice. Nature 544, 488–492 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  269. Conboy, I. M. & Rando, T. A. Heterochronic parabiosis for the study of the effects of aging on stem cells and their niches. Cell Cycle 11, 2260–2267 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  270. Yousefzadeh, M. J. et al. Heterochronic parabiosis regulates the extent of cellular senescence in multiple tissues. Geroscience https://doi.org/10.1007/s11357-020-00185-1 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  271. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04057872 (2019).

  272. US National Library of Medicine. ClinicalTrials.gov https://www.clinicaltrials.gov/ct2/show/NCT03702920 (2018).

  273. Karin, O., Agrawal, A., Porat, Z., Krizhanovsky, V. & Alon, U. Senescent cell turnover slows with age providing an explanation for the Gompertz law. Nat. Commun. 10, 5495 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  274. Kang, H. T. et al. Chemical screening identifies ATM as a target for alleviating senescence. Nat. Chem. Biol. 13, 616–623 (2017).

    CAS  PubMed  Google Scholar 

  275. Dimri, G. P. et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc. Natl Acad. Sci. USA 92, 9363–9367 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  276. Hall, B. M. et al. p16(Ink4a) and senescence-associated beta-galactosidase can be induced in macrophages as part of a reversible response to physiological stimuli. Aging 9, 1867–1884 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  277. Evangelou, K. et al. Robust, universal biomarker assay to detect senescent cells in biological specimens. Aging Cell 16, 192–197 (2017).

    CAS  PubMed  Google Scholar 

  278. Neurohr, G. E. et al. excessive cell growth causes cytoplasm dilution and contributes to senescence. Cell 176, 1083–1097 e1018 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  279. Biran, A. et al. Quantitative identification of senescent cells in aging and disease. Aging Cell 16, 661–671 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  280. Witkiewicz, A. K., Knudsen, K. E., Dicker, A. P. & Knudsen, E. S. The meaning of p16(ink4a) expression in tumors: functional significance, clinical associations and future developments. Cell Cycle 10, 2497–2503 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  281. Herbig, U., Ferreira, M., Condel, L., Carey, D. & Sedivy, J. M. Cellular senescence in aging primates. Science 311, 1257 (2006).

    CAS  PubMed  Google Scholar 

  282. Lozano-Torres, B. et al. The chemistry of senescence. Nat. Rev. Chem. 3, 426–441 (2019).

    CAS  Google Scholar 

  283. Paez-Ribes, M., González-Gualda, E., Doherty, G. J. & Muñoz-Espín, D. Targeting senescent cells in translational medicine. EMBO Mol. Med. 11, e10234 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  284. Wang, Y. et al. Real-time imaging of senescence in tumors with DNA damage. Sci. Rep. 9, 2102 (2019).

    PubMed  PubMed Central  Google Scholar 

  285. Doherty, J. & Baehrecke, E. H. Life, death and autophagy. Nat. Cell Biol. 20, 1110–1117 (2018).

    CAS  PubMed  Google Scholar 

  286. Narita, M. et al. Spatial coupling of mTOR and autophagy augments secretory phenotypes. Science 332, 966–970 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  287. Young, A. R. et al. Autophagy mediates the mitotic senescence transition. Genes Dev. 23, 798–803 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  288. Dorr, J. R. et al. Synthetic lethal metabolic targeting of cellular senescence in cancer therapy. Nature 501, 421–425 (2013).

    PubMed  Google Scholar 

  289. Garcia-Prat, L. et al. Autophagy maintains stemness by preventing senescence. Nature 529, 37–42 (2016).

    CAS  PubMed  Google Scholar 

  290. Tai, H. et al. Autophagy impairment with lysosomal and mitochondrial dysfunction is an important characteristic of oxidative stress-induced senescence. Autophagy 13, 99–113 (2017).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

R.D.M. is supported by Telethon (TIGET grant E5), a Career Development Award from the Human Frontier Science Program, an Advanced Research Grant from the European Haematology Association, “Pilot and Seed Grant 2015” from San Raffaele Hospital, a Hollis Brownstein Research Grant from the Leukemia Research Foundation, the Interstellar Initiative on Healthy Longevity of the New York Academy of Sciences and the Japan Agency for Medical Research and Development, and the Italian AIRC under MFAG 2019, ID. 23321 project PI Di Micco Raffaella. V.K. is supported by grants from the European Research Council under the European Union’s Seventh Framework Proframme (309688) and under Horizon 2020 (856487), from the Israel Science Foundation (634-15; 2633-17), from the Israel Ministry of Health, Minerva Center “Ageing, from Physical Materials to Human Tissues”, and from the Sagol Institute for Longevity Research. V.K. is an incumbent of the Georg F. Duckwitz Professorial Chair. D.B. is supported by the Ellison Medical Foundation, the Glenn Foundation for Medical Research, the US National Institutes of Health (R01AG053229 and R01AG068076), the Mayo Clinic Children’s Research Center, the Alzheimer’s Disease Research Center at Mayo Clinic and the Cure Alzheimer’s Fund. F.d’A.d.F. is supported by AIRC (application 12971), Fondazione Telethon (GGP17111), PRIN 2015, the InterOmics Project, the AMANDA project Accordo Quadro Regione Lombardia-CNR, a European Research Council advanced grant (322726), a European Research Council proof-of-concept grant (875139), AriSLA (project “DDRNA and ALS”), the AIRC Special Program 5 per mille metastases (Project-21091) and the European Joint Programme on Rare Diseases (EJP RD).The authors apologize for not being able to cite all the important contributions of their colleagues owing to space limitations.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to the writing and revision of the article.

Corresponding authors

Correspondence to Raffaella Di Micco or Fabrizio d’Adda di Fagagna.

Ethics declarations

Competing interests

D.B. is a co-inventor on patent applications licensed to or filed by Unity Biotechnology, a company developing senolytic medicines, including small molecules that selectively eliminate senescent cells. Research in the Baker laboratory has been reviewed by the Mayo Clinic Conflict of Interest Review Board and is being conducted in compliance with Mayo Clinic conflict of interest policies. V.K. is a co-inventor on patent applications in the field of senolytics, some of which are licensed to Sentaur Bio. F.d’A.d.F. is among the inventors on patent applications for the use of antisense oligonucleotides to target DNA damage-induced transcripts. R.D.M. declares no competing interests.

Additional information

Peer review information

Nature Reviews Molecular Cell Biology thanks Daniel Muñoz-Espín and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Glossary

INK-ATTAC

Transgenic mouse model with drug-inducible caspase 8 under the control of a minimal p16 promoter element active in senescent cells to allow selective elimination of p16-expressing senescent cells.

p16-3MR

Transgenic mouse model expressing a trimodal reporter of red fluorescent protein, luciferase and herpes simplex virus thymidine kinase under the control of the p16 promoter to allow tracking and elimination of p16-expressing senescent cells.

Sirtuins

Nicotinamide dinucleotide (NAD+)-dependent deacylases that regulate diverse cellular processes, including DNA repair, inflammation, metabolism and ageing.

Mitochondrial dysfunction-associated senescence

(MiDAS). Mitochondrial damage triggers senescence with a distinct secretory phenotype that lacks IL-1-dependent inflammation.

Exosomes

Extracellular vesicles produced by the endosomal compartment involved in intercellular communication.

HMGB proteins

Non-histone molecules that bind DNA and affect chromatin compaction.

Myeloid skewing

An age-related proportional increase in myeloid cells at the expense of other lineages as observed in the bone marrow and blood.

tau

A protein found in neurons that is important for maintaining microtubule structure in axons. Mutants and hyperphosphorylated forms are found in a variety of neurodegenerative diseases, including Alzheimer disease.

Fibrosis

Pathological accumulation of extracellular matrix in diseases tissue that limits normal tissue function and leads to long-term tissue scaring.

LDL receptor

Mediates entry of LDL into cells. Mutations in the gene encoding this receptor predispose to the development of atherosclerosis.

Cataracts

Clouding of the lens in the eye leading to inability to have clear vision. Surgical intervention to replace diseased lenses is a common medical procedure in aged humans.

Lordokyphosis

Abnormal rearward curvature of the spine, observed both in laboratory mice and in humans.

Lipodystrophy

Abnormal distribution of adipose tissue in the body, can refer to both excessive or insufficient deposition.

HSP90

A molecular chaperone that promotes proper protein folding and degradation, which also contributes to heat stress resilience.

Prodrugs

Compounds that are metabolized into an active drug to modify drug bioavailability and activity.

Perforin

A pore forming protein expressed in cytotoxic T cells and natural killer cells. When these cells execute cytotoxicity, they secrete granules containing perforin, which binds to the target cell’s membrane and forms pores on the target cell in order to allow cytotoxicity.

Chimeric antigen receptor T (CAR T) cells

T cells that have been genetically engineered to express T cell receptor developed to bind a defined target in order to eliminate the cells that have the target on their membrane.

PD1

A protein expressed on the cell surface that inhibits the ability of the immune system to target the cells that express the protein. Inhibition of interaction of PD1 with its ligand is a potent immunotherapy approach.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Di Micco, R., Krizhanovsky, V., Baker, D. et al. Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nat Rev Mol Cell Biol 22, 75–95 (2021). https://doi.org/10.1038/s41580-020-00314-w

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41580-020-00314-w

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