Senolytics improve physical function and increase lifespan in old age

Ming Xu, Tamar Pirtskhalava, Joshua N Farr, Bettina M Weigand, Allyson K Palmer, Megan M Weivoda, Christina L Inman, Mikolaj B Ogrodnik, Christine M Hachfeld, Daniel G Fraser, Jennifer L Onken, Kurt O Johnson, Grace C Verzosa, Larissa G P Langhi, Moritz Weigl, Nino Giorgadze, Nathan K LeBrasseur, Jordan D Miller, Diana Jurk, Ravinder J Singh, David B Allison, Keisuke Ejima, Gene B Hubbard, Yuji Ikeno, Hajrunisa Cubro, Vesna D Garovic, Xiaonan Hou, S John Weroha, Paul D Robbins, Laura J Niedernhofer, Sundeep Khosla, Tamara Tchkonia, James L Kirkland, Ming Xu, Tamar Pirtskhalava, Joshua N Farr, Bettina M Weigand, Allyson K Palmer, Megan M Weivoda, Christina L Inman, Mikolaj B Ogrodnik, Christine M Hachfeld, Daniel G Fraser, Jennifer L Onken, Kurt O Johnson, Grace C Verzosa, Larissa G P Langhi, Moritz Weigl, Nino Giorgadze, Nathan K LeBrasseur, Jordan D Miller, Diana Jurk, Ravinder J Singh, David B Allison, Keisuke Ejima, Gene B Hubbard, Yuji Ikeno, Hajrunisa Cubro, Vesna D Garovic, Xiaonan Hou, S John Weroha, Paul D Robbins, Laura J Niedernhofer, Sundeep Khosla, Tamara Tchkonia, James L Kirkland

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.

Conflict of interest statement

Competing financial 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) 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.

Figures

Figure 1
Figure 1
Transplanting small numbers of senescent cells induces physical dysfunction in younger mice. (a) Experimental design for transplantation and physical function measurements. (b,c) Representative images of LUC activity of various organs from LUC-negative male mice (n = 3) 5 d post-transplantation with SEN (induced by radiation) and CON preadipocytes from LUC-positive transgenic mice. Scale bars, 10 mm. (d-j) Maximal walking speed (relative to baseline) (d), hanging endurance (e), grip strength (f), daily activity (g), treadmill endurance (h), food intake (i), and change in body weight (BW) (j) of 6-month-old male C57BL/6 mice 1 mo after being injected with PBS, 1×106 non-senescent control (1M CON), 0.2 x106 SEN (0.2M SEN), 0.5×106 SEN (0.5M SEN), or 1×106 SEN (1M SEN) preadipocytes (n = 6 for all groups). Results are means ± s.e.m. (k-m). SA-βgal+ cell numbers (n = 6) (k), p16Ink4a mRNA levels (n = 7) (l), and cells from recipient mice that were TAF+ (>2 TAFs/nucleus) and LUC− (n = 4 mice) (m) in 6-month-old male wildtype (LUC−) C57BL/6 mice 2 mo after being transplanted with 1×106 SEN or CON transgenic constitutively-expressing LUC (LUC+) preadipocytes from transgenic mouse donors. Results are shown as box and whiskers plots, where a box extends from the 25th to 75th percentile with the median shown as a line in the middle, and whiskers indicate smallest and largest values. *P < 0.05; ANOVA with Tukey’s post-hoc comparison (d-j) and two-tailed, unpaired Student’s t-test (k-m).
Figure 2
Figure 2
Aging exacerbates effects of senescent cell transplantation. (a) Experimental design for transplantation and physical function measurements. (b-h) Maximal walking speed (relative to baseline) (b), hanging endurance (c), grip strength (d), body weight change from baseline (e), treadmill endurance (f), daily activity (g), and food intake (h) of 17-month-old male C57BL/6 mice 1 mo after being injected with 0.5 × 106 SEN or CON preadipocytes (n = 8 for both groups). (i) Percent changes in RotaRod (in 6-month-old mice, n = 21 for both SEN and CON; in 17-month-old mice, n = 22 for SEN, n = 20 for CON) and hanging test (in 6-month-old mice, n = 6 for both SEN and CON; in 17-month-old mice, n = 8 for both SEN and CON) in mice transplanted with 0.5 × 106 SEN cells relative to the average of mice transplanted with 0.5 × 106 CON cells at both ages. Results are shown as box and whiskers plots, where a box extends from the 25th to 75th percentile with the median shown as a line in the middle, and whiskers indicate smallest and largest values. (j) One year survival curves of 17-month-old non-transplanted mice (n = 33, N/A) and mice transplanted with 0.5 × 106 SEN (n = 23) or CON (n = 24) preadipocytes. (k) Tumor burden, disease burden, and inflammation at death are shown as means ± s.e.m. after transplanting SEN or CON cells (n = 10 for SEN, n = 7 for CON). (l) Causes of death (n = 10 for SEN, n = 7 for CON). *P <0.05; Two-tailed unpaired Student’s t-test (b-i), Cox proportional hazard regression model (j) and chi-square and Fisher’s exact tests (l).
Figure 3
Figure 3
Senescent cells reduce resilience to metabolic stress in mice. (a) Experimental design for transplantation and physical function measurements. (b-h) Maximal walking speed (relative to baseline) (b), hanging endurance (c), grip strength (d), daily activity (e), food intake (f), body weight change from baseline (g), and treadmill endurance (h) of 8-month-old male C57BL/6 mice 1 mo after being on HFD and injected with 0.4 × 106 SEN or CON preadipocytes (n = 6 for both groups). (i) Percent changes in RotaRod (on NCD, n = 21 for both SEN and CON; on HFD, n = 12 for both SEN and CON) and hanging test (on NCD, n = 6 for both SEN and CON; on HFD, n = 6 for both SEN and CON) in mice transplanted with 0.4-0.5 × 106 SEN cells relative to the average of mice transplanted with 0.4-0.5 × 106 CON cells. (j) Experimental design for transplantation and physical function measurements. (k-q) Maximal walking speed (relative to baseline) (k), hanging endurance (l), grip strength (m), body weight change from baseline (n), treadmill endurance (o), daily activity (p), and food intake (q) of 8-month-old male C57BL/6 mice 1 mo after being on HFD and injected with 1 × 106 SEN or CON autologous ear fibroblasts (n = 10 for both groups). All results are shown as box and whiskers plots, where a box extends from the 25th to 75th percentile with the median shown as a line in the middle, and whiskers indicate smallest and largest values. *P <0.05; Two-tailed unpaired Student’s t-tests (a-q).
Figure 4
Figure 4
D+Q reduces senescent cell abundance and decreases pro-inflammatory cytokine secretion in human adipose tissue. (a) Experimental design. (b) Percent TAF+ cells (n = 5). Blue arrows indicate TAFs. Scale bars, 5μm. (c) Percent p16INK4A-high cells (red arrows), percent p16INK4A+ cells (expressing any detectable level of p16INK4A, green arrows), percent p16INK4A− cells (black arrows), and cell number per field (n = 6). Scale bar, 100μm. (d) Percent SA-βgal+ cells (red arrows) (n = 6). Scale bar, 100μm. (e) Percent cleaved caspase-3+ cells (red arrows) (n = 5). Scale bar, 100μm. (f) Secreted cytokine and adipokine levels in conditioned media (CM) (n = 8). Results are means ± s.e.m. (g) The relative mRNA abundance of key SASP components and markers for adipose tissue function (n = 7). All results are shown as box and whiskers plots, where a box extends from the 25th to 75th percentile with the median shown as a line in the middle, and whiskers indicate smallest and largest values. *P <0.05; Two-tailed Student’s t-tests (a-g).
Figure 5
Figure 5
Eliminating senescent cells both prevents and alleviates physical dysfunction. (a) Experimental design for transplantation and physical function measurements. (b) Representative images of LUC activity in mice 2 days after the last treatment. Scale bars, 15mm. (c) Luminescence of transplanted cells as percent relative to the average of mice treated with V (n =16 for SEN-DQ vs. SEN-V; n =13 for CON-DQ vs. CON-V). (d-f) Maximal walking speed (relative to baseline) (d), hanging endurance (e), and grip strength (f) of 5-month-old male C57BL/6 mice 1 mo after the last drug treatment (n = 7 for SEN-V, CON-V, and SEN-DQ; n = 6 for CON-DQ). (g) Experimental design for transplantation and physical function measurements. (h-j) Maximal walking speed (relative to baseline) (h), hanging endurance (i), and grip strength (j) of 5-month-old male C57BL/6 mice 2 weeks after the last drug treatment (n = 10 for SEN-DQ and SEN-V; n = 14 for CON-V). All results are shown as box and whiskers plots, where a box extends from the 25th to 75th percentile with the median shown as a line in the middle, and whiskers indicate smallest and largest values. *P <0.05; Two-tailed Student’s t-tests (a-j).
Figure 6
Figure 6
Senolytics extend both health- and life-span in aged mice. (a) Experimental design for physical function measurements in 20-month-old male mice treated with D+Q once every 2 weeks (bi-weekly) for 4 months. (b-h) Maximal walking speed (relative to baseline) (b), hanging endurance (c), grip strength (d), body weight change from baseline (e), treadmill endurance (f), daily activity (g), and food intake (h) of 20-month-old male C57BL/6 mice 4 mo after drug initiation (n = 20 for D+Q; n = 13 for V). (i) The relative mRNA abundance for target genes of visceral adipose tissue from 6-month-old non-treated (6m, n = 7), 24-month-old V-treated (24m-V, n = 8), and 24-month-old D+Q-treated (24m-DQ, n = 8) mice. (j) Experimental design for lifespan analyses. (k,l) Post-treatment survival curves (k) and whole-life survival curves (l) of C57BL/6 mice treated bi-weekly with D+Q (n = 71; 40 males, 31 females) or V (n = 76; 41 males, 35 females) starting at 24-27 months of age. Median survival is indicated for all curves. (m) Maximal walking speed and hanging endurance averaged over the last 2 months of life and lifespan for the longest living mice (top 40%) in both groups for both sexes. For male mice, n = 12 for D+Q and n = 12 for V. For female mice, n = 13 for D+Q and n = 13 for V. (n) Disease burden and tumor burden at death. For both sexes, n = 59 for D+Q, n = 62 for V. For males, n = 30 for D+Q, n = 29 for V. For females, n = 29 for D+Q, n = 33 for V. (b-i, m) Results are shown as box and whiskers plots, where a box extends from the 25th to 75th percentile with the median shown as a line in the middle, and whiskers indicate smallest and largest values. (n) Results are shown as mean ± s.e.m. *P <0.05; n.s., not significant; Two-tailed Student’s t-tests (b-i, m-n) and Cox proportional hazard regression model (k-l).

References

    1. Crimmins EM. Lifespan and Healthspan: Past, Present, and Promise. Gerontologist. 2015;55:901–911.
    1. Fries JF. Aging, natural death, and the compression of morbidity. N Engl J Med. 1980;303:130–135.
    1. Michaud M, et al. Proinflammatory cytokines, aging, and age-related diseases. J Am Med Dir Assoc. 2013;14:877–882.
    1. Fried LP, et al. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci. 2001;56:M146–156.
    1. Collard RM, Boter H, Schoevers RA, Oude Voshaar RC. Prevalence of frailty in community-dwelling older persons: a systematic review. Journal of the American Geriatrics Society. 2012;60:1487–1492.
    1. Song X, Mitnitski A, Rockwood K. Prevalence and 10-year outcomes of frailty in older adults in relation to deficit accumulation. Journal of the American Geriatrics Society. 2010;58:681–687.
    1. Xue QL. The frailty syndrome: definition and natural history. Clinics in geriatric medicine. 2011;27:1–15.
    1. Tchkonia T, Zhu Y, van Deursen J, Campisi J, Kirkland JL. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. The Journal of clinical investigation. 2013;123:966–972.
    1. Campisi J, d’Adda di Fagagna F. Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol. 2007;8:729–740.
    1. Wiley CD, et al. Mitochondrial Dysfunction Induces Senescence with a Distinct Secretory Phenotype. Cell Metab. 2016;23:303–314.
    1. Wang C, et al. DNA damage response and cellular senescence in tissues of aging mice. Aging Cell. 2009;8:311–323.
    1. Zhu Y, Armstrong JL, Tchkonia T, Kirkland JL. Cellular senescence and the senescent secretory phenotype in age-related chronic diseases. Current opinion in clinical nutrition and metabolic care. 2014;17:324–328.
    1. Coppe JP, et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 2008;6:2853–2868.
    1. Xu M, et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc Natl Acad Sci U S A. 2015;112:E6301–6310.
    1. Xu M, et al. Targeting senescent cells enhances adipogenesis and metabolic function in old age. Elife. 2015;4:e12997.
    1. Xu M, et al. Transplanted Senescent Cells Induce an Osteoarthritis-Like Condition in Mice. J Gerontol A Biol Sci Med Sci. 2017;72:780–785.
    1. Palmer AK, et al. Cellular Senescence in Type 2 Diabetes: A Therapeutic Opportunity. Diabetes. 2015;64:2289–2298.
    1. Chang J, et al. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nature medicine. 2016;22:78–83.
    1. Childs BG, et al. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science. 2016;354:472–477.
    1. Baker DJ, et al. Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature. 2016;530:184–189.
    1. Xu M, Tchkonia T, Kirkland JL. Perspective: Targeting the JAK/STAT pathway to fight age-related dysfunction. Pharmacol Res. 2016;111:152–154.
    1. Bitto A, et al. Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice. Elife. 2016;5
    1. Tchkonia T, et al. Mechanisms and metabolic implications of regional differences among fat depots. Cell Metab. 2013;17:644–656.
    1. Farr JN, et al. Identification of Senescent Cells in the Bone Microenvironment. J Bone Miner Res. 2016;31:1920–1929.
    1. Schafer MJ, et al. Cellular senescence mediates fibrotic pulmonary disease. Nature communications. 2017;8:14532.
    1. Ryan JM, Barry FP, Murphy JM, Mahon BP. Mesenchymal stem cells avoid allogeneic rejection. J Inflamm (Lond) 2005;2:8.
    1. Tchkonia T, et al. Fat tissue, aging, and cellular senescence. Aging Cell. 2010;9:667–684.
    1. Acosta JC, et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nature cell biology. 2013;15:978–990.
    1. Nelson G, et al. A senescent cell bystander effect: senescence-induced senescence. Aging Cell. 2012;11:345–349.
    1. Hewitt G, et al. Telomeres are favoured targets of a persistent DNA damage response in ageing and stress-induced senescence. Nature communications. 2012;3:708.
    1. Walston J, et al. The physical and biological characterization of a frail mouse model. J Gerontol A Biol Sci Med Sci. 2008;63:391–398.
    1. Schafer MJ, et al. Exercise Prevents Diet-Induced Cellular Senescence in Adipose Tissue. Diabetes. 2016;65:1606–1615.
    1. Mosier DE, Stell KL, Gulizia RJ, Torbett BE, Gilmore GL. Homozygous scid/scid;beige/beige mice have low levels of spontaneous or neonatal T cell-induced B cell generation. J Exp Med. 1993;177:191–194.
    1. Kirkland JL, Tchkonia T. Cellular Senescence: A Translational Perspective. EBioMedicine. 2017
    1. Zhu Y, et al. The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell. 2015;14:644–658.
    1. Stern JH, Rutkowski JM, Scherer PE. Adiponectin, Leptin, and Fatty Acids in the Maintenance of Metabolic Homeostasis through Adipose Tissue Crosstalk. Cell Metab. 2016;23:770–784.
    1. Farmer SR. Transcriptional control of adipocyte formation. Cell Metab. 2006;4:263–273.
    1. Reuben DB, Judd-Hamilton L, Harris TB, Seeman TE. The associations between physical activity and inflammatory markers in high-functioning older persons: MacArthur Studies of Successful Aging. Journal of the American Geriatrics Society. 2003;51:1125–1130.
    1. Cohen HJ, Pieper CF, Harris T, Rao KM, Currie MS. The association of plasma IL-6 levels with functional disability in community-dwelling elderly. J Gerontol A Biol Sci Med Sci. 1997;52:M201–208.
    1. Beyer I, et al. Inflammation-related muscle weakness and fatigue in geriatric patients. Exp Gerontol. 2012;47:52–59.
    1. Lu Y, et al. Inflammatory and immune markers associated with physical frailty syndrome: findings from Singapore longitudinal aging studies. Oncotarget. 2016;7:28783–28795.
    1. Kao TW, et al. Examining how p16(INK4a) expression levels are linked to handgrip strength in the elderly. Sci Rep. 2016;6:31905.
    1. Justice JN, 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. 2017
    1. Christopher LJ, et al. Metabolism and disposition of dasatinib after oral administration to humans. Drug metabolism and disposition: the biological fate of chemicals. 2008;36:1357–1364.
    1. Graefe EU, et al. Pharmacokinetics and bioavailability of quercetin glycosides in humans. Journal of clinical pharmacology. 2001;41:492–499.
    1. Roos CM, et al. Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell. 2016
    1. Farr JN, et al. Targeting cellular senescence prevents age-related bone loss in mice. Nature medicine. 2017;23:1072–1079.
    1. Ogrodnik M, et al. Cellular senescence drives age-dependent hepatic steatosis. Nature communications. 2017;8:15691.
    1. Baker DJ, et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011;479:232–236.
    1. Srinivas-Shankar U, Wu FC. Frailty and muscle function: role for testosterone? Front Horm Res. 2009;37:133–149.
    1. Kane AE, et al. Animal models of frailty: current applications in clinical research. Clin Interv Aging. 2016;11:1519–1529.
    1. Hall BM, et al. p16(Ink4a) and senescence-associated beta-galactosidase can be induced in macrophages as part of a reversible response to physiological stimuli. Aging (Albany NY) 2017
    1. Helman A, et al. p16-induced senescence of pancreatic beta cells enhances insulin secretion. Nature medicine. 2016
    1. Costa LG, Garrick JM, Roque PJ, Pellacani C. Mechanisms of Neuroprotection by Quercetin: Counteracting Oxidative Stress and More. Oxid Med Cell Longev. 2016;2016:2986796.
    1. Montani D, et al. Pulmonary arterial hypertension in patients treated by dasatinib. Circulation. 2012;125:2128–2137.
    1. Kirkland JL, Tchkonia T, Zhu Y, Niedernhofer LJ, Robbins PD. The Clinical Potential of Senolytic Drugs. Journal of the American Geriatrics Society. 2017
    1. Ness KK, et al. Frailty in childhood cancer survivors. Cancer. 2015;121:1540–1547.
Methods-Only References
    1. Pajvani UB, et al. Fat apoptosis through targeted activation of caspase 8: a new mouse model of inducible and reversible lipoatrophy. Nature medicine. 2005;11:797–803.
    1. Miller RA, et al. An Aging Interventions Testing Program: study design and interim report. Aging Cell. 2007;6:565–575.
    1. Ikeno Y, et al. Housing density does not influence the longevity effect of calorie restriction. The journals of gerontology. Series A, Biological sciences and medical sciences. 2005;60:1510–1517.
    1. Tchkonia T, et al. Increased TNFalpha and CCAAT/enhancer-binding protein homologous protein with aging predispose preadipocytes to resist adipogenesis. Am J Physiol Endocrinol Metab. 2007;293:E1810–1819.
    1. Jurk D, et al. Chronic inflammation induces telomere dysfunction and accelerates ageing in mice. Nature communications. 2014;2:4172.
    1. Xu J. Preparation, culture, and immortalization of mouse embryonic fibroblasts. Curr Protoc Mol Biol. 2005:21. Chapter 28, Unit 28.

Source: PubMed

3
Tilaa