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.

Senolytics improve physical function and increase lifespan in old age

Abstract

Physical function declines in old age, portending disability, increased health expenditures, and mortality. Cellular senescence, leading to tissue dysfunction, may contribute to these consequences of aging, but whether senescence can directly drive age-related pathology and be therapeutically targeted is still unclear. Here we demonstrate that transplanting relatively small numbers of senescent cells into young mice is sufficient to cause persistent physical dysfunction, as well as to spread cellular senescence to host tissues. Transplanting even fewer senescent cells had the same effect in older recipients and was accompanied by reduced survival, indicating the potency of senescent cells in shortening health- and lifespan. The senolytic cocktail, dasatinib plus quercetin, which causes selective elimination of senescent cells, decreased the number of naturally occurring senescent cells and their secretion of frailty-related proinflammatory cytokines in explants of human adipose tissue. Moreover, intermittent oral administration of senolytics to both senescent cell–transplanted young mice and naturally aged mice alleviated physical dysfunction and increased post-treatment survival by 36% while reducing mortality hazard to 65%. Our study provides proof-of-concept evidence that senescent cells can cause physical dysfunction and decreased survival even in young mice, while senolytics can enhance remaining health- and lifespan in old mice.

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: Transplanting small numbers of senescent cells induces physical dysfunction in younger mice.
Fig. 2: Aging exacerbates the effects of senescent cell transplantation.
Fig. 3: Senescent cells reduce resilience to metabolic stress in mice.
Fig. 4: D + Q reduces senescent cell abundance and decreases proinflammatory cytokine secretion in human adipose tissue.
Fig. 5: Eliminating senescent cells both prevents and alleviates physical dysfunction.
Fig. 6: Senolytics extend both health- and lifespan in aged mice.

Similar content being viewed by others

References

  1. Crimmins, E. M. Lifespan and healthspan: past, present, and promise. Gerontologist 55, 901–911 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Fries, J. F. Aging, natural death, and the compression of morbidity. N. Engl. J. Med. 303, 130–135 (1980).

    Article  CAS  PubMed  Google Scholar 

  3. Michaud, M. et al. Proinflammatory cytokines, aging, and age-related diseases. J. Am. Med. Dir. Assoc. 14, 877–882 (2013).

    Article  PubMed  Google Scholar 

  4. Fried, L. P. et al. Frailty in older adults: evidence for a phenotype. J. Gerontol. A Biol. Sci. Med. Sci. 56, M146–M156 (2001).

    Article  CAS  PubMed  Google Scholar 

  5. Collard, R. M., Boter, H., Schoevers, R. A. & Oude Voshaar, R. C. Prevalence of frailty in community-dwelling older persons: a systematic review. J. Am. Geriatr. Soc. 60, 1487–1492 (2012).

    Article  PubMed  Google Scholar 

  6. Song, X., Mitnitski, A. & Rockwood, K. Prevalence and 10-year outcomes of frailty in older adults in relation to deficit accumulation. J. Am. Geriatr. Soc. 58, 681–687 (2010).

    Article  PubMed  Google Scholar 

  7. Xue, Q. L. The frailty syndrome: definition and natural history. Clin. Geriatr. Med. 27, 1–15 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Tchkonia, T., Zhu, Y., van Deursen, J., Campisi, J. & Kirkland, J. L. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J. Clin. Invest. 123, 966–972 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Campisi, J. & d’Adda di Fagagna, F. Cellular senescence: when bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 8, 729–740 (2007).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. Wang, C. et al. DNA damage response and cellular senescence in tissues of aging mice. Aging Cell 8, 311–323 (2009).

    Article  CAS  PubMed  Google Scholar 

  12. Zhu, Y., Armstrong, J. L., Tchkonia, T. & Kirkland, J. L. Cellular senescence and the senescent secretory phenotype in age-related chronic diseases. Curr. Opin. Clin. Nutr. Metab. Care 17, 324–328 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Coppé, 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).

    Article  PubMed  CAS  Google Scholar 

  14. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Xu, M. et al. Targeting senescent cells enhances adipogenesis and metabolic function in old age. eLife 4, e12997 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Xu, M. et al. transplanted senescent cells induce an osteoarthritis-like condition in mice. J. Gerontol. A Biol. Sci. Med. Sci. 72, 780–785 (2017).

    Article  CAS  PubMed  Google Scholar 

  17. Palmer, A. K. et al. cellular senescence in type 2 diabetes: a therapeutic opportunity. Diabetes 64, 2289–2298 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Baker, D. J. et al. Naturally occurring p16Ink4a-positive cells shorten healthy lifespan. Nature 530, 184–189 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Xu, M., Tchkonia, T. & Kirkland, J. L. Perspective: targeting the JAK/STAT pathway to fight age-related dysfunction. Pharmacol. Res. 111, 152–154 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Bitto, A. et al. Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice. eLife 5, e16351 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Tchkonia, T. et al. Mechanisms and metabolic implications of regional differences among fat depots. Cell Metab. 17, 644–656 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Farr, J. N. et al. Identification of senescent cells in the bone microenvironment. J. Bone Miner. Res. 31, 1920–1929 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ryan, J. M., Barry, F. P., Murphy, J. M. & Mahon, B. P. Mesenchymal stem cells avoid allogeneic rejection. J. Inflamm. (Lond.) 2, 8 (2005).

    Article  CAS  Google Scholar 

  27. Tchkonia, T. et al. Fat tissue, aging, and cellular senescence. Aging Cell 9, 667–684 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Nelson, G. et al. A senescent cell bystander effect: senescence-induced senescence. Aging Cell 11, 345–349 (2012).

    Article  CAS  PubMed  Google Scholar 

  30. 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  CAS  Google Scholar 

  31. Walston, J. et al. The physical and biological characterization of a frail mouse model. J. Gerontol. A Biol. Sci. Med. Sci. 63, 391–398 (2008).

    Article  PubMed  Google Scholar 

  32. Schafer, M. J. et al. Exercise prevents diet-induced cellular senescence in adipose tissue. Diabetes 65, 1606–1615 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Mosier, D. E., Stell, K. L., Gulizia, R. J., Torbett, B. E. & Gilmore, G. L. Homozygous scid/scid;beige/beige mice have low levels of spontaneous or neonatal T cell-induced B cell generation. J. Exp. Med. 177, 191–194 (1993).

    Article  CAS  PubMed  Google Scholar 

  34. Kirkland, J. L. & Tchkonia, T. Cellular senescence: a translational perspective. EBioMedicine 21, 21–28 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Stern, J. H., Rutkowski, J. M. & Scherer, P. E. Adiponectin, leptin, and fatty acids in the maintenance of metabolic homeostasis through adipose tissue crosstalk. Cell Metab. 23, 770–784 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Farmer, S. R. Transcriptional control of adipocyte formation. Cell Metab. 4, 263–273 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Reuben, D. B., Judd-Hamilton, L., Harris, T. B. & Seeman, T. E. The associations between physical activity and inflammatory markers in high-functioning older persons: MacArthur Studies of Successful Aging. J. Am. Geriatr. Soc. 51, 1125–1130 (2003).

    Article  PubMed  Google Scholar 

  39. Cohen, H. J., Pieper, C. F., Harris, T., Rao, K. M. & Currie, M. S. The association of plasma IL-6 levels with functional disability in community-dwelling elderly. J. Gerontol. A Biol. Sci. Med. Sci. 52, M201–M208 (1997).

    Article  CAS  PubMed  Google Scholar 

  40. Beyer, I. et al. Inflammation-related muscle weakness and fatigue in geriatric patients. Exp. Gerontol. 47, 52–59 (2012).

    Article  CAS  PubMed  Google Scholar 

  41. Lu, Y. et al. Inflammatory and immune markers associated with physical frailty syndrome: findings from Singapore longitudinal aging studies. Oncotarget 7, 28783–28795 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Kao, T. W. et al. Examining how p16INK4a expression levels are linked to handgrip strength in the elderly. Sci. Rep. 6, 31905 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Justice, J. N. et al. Cellular senescence biomarker p16INK4a+ cell burden in thigh adipose is associated with poor physical function in older women. J. Gerontol. A Biol. Sci. Med. Sci. 73, 939–945 (2017).

    Article  PubMed Central  CAS  Google Scholar 

  44. Christopher, L. J. et al. Metabolism and disposition of dasatinib after oral administration to humans. Drug Metab. Dispos. 36, 1357–1364 (2008).

    Article  CAS  PubMed  Google Scholar 

  45. Graefe, E. U. et al. Pharmacokinetics and bioavailability of quercetin glycosides in humans. J. Clin. Pharmacol. 41, 492–499 (2001).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ogrodnik, M. et al. Cellular senescence drives age-dependent hepatic steatosis. Nat. Commun. 8, 15691 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Baker, D. J. et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479, 232–236 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Srinivas-Shankar, U. & Wu, F. C. Frailty and muscle function: role for testosterone? Front. Horm. Res. 37, 133–149 (2009).

    Article  CAS  PubMed  Google Scholar 

  51. Hall, B. M. et al. p16Ink4a and senescence-associated β-galactosidase can be induced in macrophages as part of a reversible response to physiological stimuli. Aging (Albany NY) 9, 1867 (2017).

    Article  CAS  Google Scholar 

  52. Helman, A. et al. p16Ink4a-induced senescence of pancreatic beta cells enhances insulin secretion. Nat. Med. 22, 412–420 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Costa, L. G., Garrick, J. M., Roquè, P. J. & Pellacani, C. Mechanisms of neuroprotection by quercetin: counteracting oxidative stress and more. Oxid. Med. Cell. Longev. 2016, 2986796 (2016).

    PubMed  PubMed Central  Google Scholar 

  54. Montani, D. et al. Pulmonary arterial hypertension in patients treated by dasatinib. Circulation 125, 2128–2137 (2012).

    Article  CAS  PubMed  Google Scholar 

  55. Kirkland, J. L., Tchkonia, T., Zhu, Y., Niedernhofer, L. J. & Robbins, P. D. The clinical potential of senolytic drugs. J. Am. Geriatr. Soc. 65, 2297–2301 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Kane, A. E. et al. Animal models of frailty: current applications in clinical research. Clin. Interv. Aging 11, 1519–1529 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ness, K. K. et al. Frailty in childhood cancer survivors. Cancer 121, 1540–1547 (2015).

    Article  PubMed  Google Scholar 

  58. Pajvani, U. B. et al. Fat apoptosis through targeted activation of caspase 8: a new mouse model of inducible and reversible lipoatrophy. Nat. Med. 11, 797–803 (2005).

    Article  CAS  PubMed  Google Scholar 

  59. Miller, R. A. et al. An aging interventions testing program: study design and interim report. Aging Cell 6, 565–575 (2007).

    Article  CAS  PubMed  Google Scholar 

  60. Ikeno, Y. et al. Housing density does not influence the longevity effect of calorie restriction. J. Gerontol. A Biol. Sci. Med. Sci. 60, 1510–1517 (2005).

    Article  PubMed  Google Scholar 

  61. Tchkonia, T. et al. Increased TNFα and CCAAT/enhancer-binding protein homologous protein with aging predispose preadipocytes to resist adipogenesis. Am. J. Physiol. Endocrinol. Metab. 293, E1810–E1819 (2007).

    Article  CAS  PubMed  Google Scholar 

  62. Jurk, D. et al. Chronic inflammation induces telomere dysfunction and accelerates ageing in mice. Nat. Commun. 2, 4172 (2014).

    Article  PubMed  CAS  Google Scholar 

  63. Xu, J. Preparation, culture, and immortalization of mouse embryonic fibroblasts. Curr. Protoc. Mol. Biol. 70, 28.1.1–28.1.8 (2005).

    Google Scholar 

Download references

Acknowledgements

The authors are grateful to J. L. Armstrong and L. Thesing for administrative assistance, M. Mahlman for obtaining human adipose tissue samples, Z. Aversa for help with muscle analysis, the Pathology Research Core Lab at Mayo Clinic–Rochester for histology studies, and C. Guo for overall support. This work was supported by the Connor Group (J.L.K.) and Robert J. and Theresa W. Ryan (J.L.K.); the National Institutes of Health (NIH) grants AG13925 (J.L.K.), AG49182 (J.L.K), DK50456 (Adipocyte Subcore, J.L.K.), AG46061 (A.K.P.), AG004875 (S.K.), AG048792 (S.K.), AR070241 (J.N.F.), AR070281 (M.M.W.), AG13319 (Y.I. and G.B.H.), AG050886 (D.B.A.), AG043376 (P.D.R., and L.J.N.), AG056278 (P.D.R. and L.J.N.), and AG044376 (L.J.N.); a Glenn/American Federation for Aging Research (AFAR) BIG Award (J.L.K.); the Glenn Foundation (L.J.N.); and the Ted Nash Long Life and Noaber Foundations (J.L.K.). M.X. received the Glenn/AFAR Postdoctoral Fellowship for Translational Research on Aging and an Irene Diamond Fund/AFAR Postdoctoral Transition Award in Aging.

Author information

Authors and Affiliations

Authors

Contributions

M.X., T.T., and J.L.K. conceived and designed the study. M.X. performed and analyzed most of the transplanted mouse experiments and human adipose tissue explant experiments. N.G. and T.T. contributed to the human adipose tissue explant experiments. J.N.F., D.G.F., J.L.O., and S.K. contributed to the study of aged mice treated with senolytics. A.K.P. contributed to the bioluminescence studies. M.B.O. and D.J. contributed to ear fibroblast isolation. T.P., M.X., T.T., and C.L.I. performed the survival experiments using senolytics in aged mice. B.M.W., M.M.W., C.M.H., N.K.L., H.C., V.D.G., X.H., S.J.W., K.O.J., M.W., L.G.P.L., G.C.V., P.D.R., L.J.N., and J.D.M. contributed to the mouse studies. R.J.S. contributed to androgen measurement. D.B.A. and K.E. contributed to lifespan analysis. G.B.H. and Y.I. contributed to mouse pathology analysis. M.X. and J.L.K. wrote the manuscript with input from all coauthors. J.L.K., M.X., and T.T. oversaw all experimental design, data analyses, and manuscript preparation.

Corresponding authors

Correspondence to Ming Xu, Tamara Tchkonia or James L. Kirkland.

Ethics declarations

Competing interests

J.L.K, T.T., M.X., T.P., N.G., and A.K.P. have a financial interest related to this research. Patents on senolytic drugs (PCT/US2016/041646, filed at the US Patent Office) are held by Mayo Clinic. This research has been reviewed by the Mayo Clinic Conflict of Interest Review Board and was conducted in compliance with Mayo Clinic Conflict of Interest policies. None of the other authors has a relevant financial conflict of interest.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, M., Pirtskhalava, T., Farr, J.N. et al. Senolytics improve physical function and increase lifespan in old age. Nat Med 24, 1246–1256 (2018). https://doi.org/10.1038/s41591-018-0092-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0092-9

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