Mitochondrial peptides modulate mitochondrial function during cellular senescence

Su-Jeong Kim, Hemal H Mehta, Junxiang Wan, Chisaka Kuehnemann, Jingcheng Chen, Ji-Fan Hu, Andrew R Hoffman, Pinchas Cohen, Su-Jeong Kim, Hemal H Mehta, Junxiang Wan, Chisaka Kuehnemann, Jingcheng Chen, Ji-Fan Hu, Andrew R Hoffman, Pinchas Cohen

Abstract

Cellular senescence is a complex cell fate response that is thought to underlie several age-related pathologies. Despite a loss of proliferative potential, senescent cells are metabolically active and produce energy-consuming effectors, including senescence-associated secretory phenotypes (SASPs). Mitochondria play crucial roles in energy production and cellular signaling, but the key features of mitochondrial physiology and particularly of mitochondria-derived peptides (MDPs), remain underexplored in senescence responses. Here, we used primary human fibroblasts made senescent by replicative exhaustion, doxorubicin or hydrogen peroxide treatment, and examined the number of mitochondria and the levels of mitochondrial respiration, mitochondrial DNA methylation and the mitochondria-encoded peptides humanin, MOTS-c, SHLP2 and SHLP6. Senescent cells showed increased numbers of mitochondria and higher levels of mitochondrial respiration, variable changes in mitochondrial DNA methylation, and elevated levels of humanin and MOTS-c. Humanin and MOTS-c administration modestly increased mitochondrial respiration and selected components of the SASP in doxorubicin-induced senescent cells partially via JAK pathway. Targeting metabolism in senescence cells is an important strategy to reduce SASP production for eliminating the deleterious effects of senescence. These results provide insight into the role of MDPs in mitochondrial energetics and the production of SASP components by senescent cells.

Keywords: SASP (senescence-associated secretory phenotype); mitochondria; mitochondrial energetics; mitochondrial-derived peptides; mtDNA methylation; senescence.

Conflict of interest statement

CONFLICTS OF INTEREST: Pinchas Cohen is a consultant and stockholder of CohBar Inc.

Figures

Figure 1
Figure 1
Mitochondria mass and energetics are altered during doxorubicin-induced senescence. (A) mitochondrial DNA (mtDNA) copy number in non-senescent (quiescent) and senescent cells. (B) Representative images of Tom20 (green; mitochondria) and Hoechst 33258 (blue; nucleus) immunostaining in non-senescent (quiescent) and senescent cells. Scale bar, 20 μm. The area of Tom20 staining per cells were measured using image J. (C) Cellular ATP levels in non-senescent (quiescent) and senescent cells. (D) Cellular oxygen consumption rate (OCR) in non-senescent and senescent cells. The basal respiration, spare respiratory capacity, and ATP production are calculated based on the sequential compound injection according to the manufacture’s instruction. (E) The extracellular acidification rate (ECAR) in non-senescent (quiescent) and senescent cells. Glycolysis, glycolytic capacity, and glycolytic reserve are calculated based on the sequential compound injection according to the manufacture’s instruction. Data are reported as mean ± SEM of three to eight independent experiments. Significant differences were determined by Student’s t-tests. *p<0.05, ***p<0.001. Abbreviations: NS, Non-senescent cells (quiescent); SEN, Senescent cells.
Figure 2
Figure 2
Mitochondrial fuel usage is altered during doxorubicin-induced senescence. (A) Glucose uptake rate was measured by 2-NBDG, a fluorescently labeled deoxyglucose analog. Quantification and representative western blots of (B) carnitine palmitoyltransferase I (CPT1A) and (C) glutaminase (GLS1), both showing beta-actin as a loading control. Data are reported as mean ± SEM of three to six independent experiments. Significant differences were determined by Student’s t-tests. *p<0.05. Abbreviations: NS, Non-senescent cells (quiescent); SEN, Senescent cells.
Figure 3
Figure 3
Mitochondrial respiration was not altered, but glycolysis was enhanced in replicative senescence. (A) Cellular oxygen consumption rate (OCR) in non-senescence (quiescent) and senescent cells. The basal respiration, spare respiratory capacity, and ATP production are calculated based on the sequential compound injection according to the manufacture’s instruction. (B) The extracellular acidification rate (ECAR) in non-senescent (quiescent) and senescent cells. (C) Glucose uptake rate were measured by 2-NBDG, a fluorescently labeled deoxyglucose analog. Data are reported as mean ± SEM of three to eight independent experiments. Significant differences were determined with Student’s t-tests. *p<0.05, **p<0.01, ***p<0.001. Abbreviations: NS, Non-senescent cells (quiescent); SEN, Senescent cells.
Figure 4
Figure 4
Mitochondria DNA methylation changes during doxorubicin-induced senescence. (A) Schematic diagram of mitochondrial genes and CpG sites in the mtDNA. Quantification and representative agarose gel images of mtDNA methylation levels at the site of (B) CpG1 and (C) CpG4. (D) Quantification and representative western blots of COX1 (MT-CO1) in non-senescent and senescent cells. Reduced lamin B1 was used as a senescence marker. Data are reported as mean ± SEM of three to four independent experiments. Significant differences were determined by Student’s t-tests. *p<0.05, ***p<0.001. Abbreviations: NS, Non-senescent cells (quiescent); SEN, Senescent cells.
Figure 5
Figure 5
Expression levels of MDPs are differentially regulated during cellular senescence. Humanin and MOTS-c levels were examined in (A) doxorubicin-induced senescence and (B) replicative senescence. Data are reported as mean ± SEM of three independent experiments. Significant differences were determined by Student’s t-tests. *p<0.05.
Figure 6
Figure 6
Humanin and MOTS-c modulate the mitochondrial respiration. (A) Cellular oxygen consumption rate (OCR; pmole/min/total DNA) in non-senescence (quiescent) and senescent cells in the absence or presence of either HNG (a potent analogue of humanin with a glycine substitution, S14G) or MOTS-c. The basal respiration, spare respiratory capacity, and ATP production are calculated based on the sequential compound injection according to the manufacture’s instruction. (B) Representative western blots of carnitine palmitoyltransferase I (CPT1A) in non-senescence and senescent cells in the absence or presence of either HNG (a potent analogue of humanin with a glycine substitution, S14G) or MOTS-c. Quantification of carnitine palmitoyltransferase I (CPT1A) expression. Data are reported as mean ± SEM of three independent experiments. Significant differences were determined with one-way ANOVA followed by Tukey’s post hoc test. *p<0.05.

References

    1. Liu B, Du Q, Chen L, Fu G, Li S, Fu L, Zhang X, Ma C, Bin C. CpG methylation patterns of human mitochondrial DNA. Sci Rep. 2016; 6:23421. 10.1038/srep23421
    1. Iacobazzi V, Castegna A, Infantino V, Andria G. Mitochondrial DNA methylation as a next-generation biomarker and diagnostic tool. Mol Genet Metab. 2013; 110:25–34. 10.1016/j.ymgme.2013.07.012
    1. Shock LS, Thakkar PV, Peterson EJ, Moran RG, Taylor SM. DNA methyltransferase 1, cytosine methylation, and cytosine hydroxymethylation in mammalian mitochondria. Proc Natl Acad Sci USA. 2011; 108:3630–35. 10.1073/pnas.1012311108
    1. Saini SK, Mangalhara KC, Prakasam G, Bamezai RN. DNA Methyltransferase1 (DNMT1) Isoform3 methylates mitochondrial genome and modulates its biology. Sci Rep. 2017; 7:1525. 10.1038/s41598-017-01743-y
    1. Mawlood SK, Dennany L, Watson N, Dempster J, Pickard BS. Quantification of global mitochondrial DNA methylation levels and inverse correlation with age at two CpG sites. Aging (Albany NY). 2016; 8:636–41. 10.18632/aging.100892
    1. Ikonen M, Liu B, Hashimoto Y, Ma L, Lee KW, Niikura T, Nishimoto I, Cohen P. Interaction between the Alzheimer’s survival peptide humanin and insulin-like growth factor-binding protein 3 regulates cell survival and apoptosis. Proc Natl Acad Sci USA. 2003; 100:13042–47. 10.1073/pnas.2135111100
    1. Cobb LJ, Lee C, Xiao J, Yen K, Wong RG, Nakamura HK, Mehta HH, Gao Q, Ashur C, Huffman DM, Wan J, Muzumdar R, Barzilai N, Cohen P. Naturally occurring mitochondrial-derived peptides are age-dependent regulators of apoptosis, insulin sensitivity, and inflammatory markers. Aging (Albany NY). 2016; 8:796–809. 10.18632/aging.100943
    1. Lee C, Zeng J, Drew BG, Sallam T, Martin-Montalvo A, Wan J, Kim SJ, Mehta H, Hevener AL, de Cabo R, Cohen P. The mitochondrial-derived peptide MOTS-c promotes metabolic homeostasis and reduces obesity and insulin resistance. Cell Metab. 2015; 21:443–54. 10.1016/j.cmet.2015.02.009
    1. Muzumdar RH, Huffman DM, Atzmon G, Buettner C, Cobb LJ, Fishman S, Budagov T, Cui L, Einstein FH, Poduval A, Hwang D, Barzilai N, Cohen P. Humanin: a novel central regulator of peripheral insulin action. PLoS One. 2009; 4:e6334. 10.1371/journal.pone.0006334
    1. Gong Z, Tas E, Muzumdar R. Humanin and age-related diseases: a new link? Front Endocrinol (Lausanne). 2014; 5:210. 10.3389/fendo.2014.00210
    1. Bachar AR, Scheffer L, Schroeder AS, Nakamura HK, Cobb LJ, Oh YK, Lerman LO, Pagano RE, Cohen P, Lerman A. Humanin is expressed in human vascular walls and has a cytoprotective effect against oxidized LDL-induced oxidative stress. Cardiovasc Res. 2010; 88:360–66. 10.1093/cvr/cvq191
    1. Oh YK, Bachar AR, Zacharias DG, Kim SG, Wan J, Cobb LJ, Lerman LO, Cohen P, Lerman A. Humanin preserves endothelial function and prevents atherosclerotic plaque progression in hypercholesterolemic ApoE deficient mice. Atherosclerosis. 2011; 219:65–73. 10.1016/j.atherosclerosis.2011.06.038
    1. Niikura T, Sidahmed E, Hirata-Fukae C, Aisen PS, Matsuoka Y. A humanin derivative reduces amyloid beta accumulation and ameliorates memory deficit in triple transgenic mice. PLoS One. 2011; 6:e16259. 10.1371/journal.pone.0016259
    1. Hashimoto Y, Niikura T, Tajima H, Yasukawa T, Sudo H, Ito Y, Kita Y, Kawasumi M, Kouyama K, Doyu M, Sobue G, Koide T, Tsuji S, et al.. A rescue factor abolishing neuronal cell death by a wide spectrum of familial Alzheimer’s disease genes and Abeta. Proc Natl Acad Sci USA. 2001; 98:6336–41. 10.1073/pnas.101133498
    1. Kim SJ, Guerrero N, Wassef G, Xiao J, Mehta HH, Cohen P, Yen K. The mitochondrial-derived peptide humanin activates the ERK1/2, AKT, and STAT3 signaling pathways and has age-dependent signaling differences in the hippocampus. Oncotarget. 2016; 7:46899–912. 10.18632/oncotarget.10380
    1. Hashimoto Y, Suzuki H, Aiso S, Niikura T, Nishimoto I, Matsuoka M. Involvement of tyrosine kinases and STAT3 in Humanin-mediated neuroprotection. Life Sci. 2005; 77:3092–104. 10.1016/j.lfs.2005.03.031
    1. Hashimoto Y, Kurita M, Aiso S, Nishimoto I, Matsuoka M. Humanin inhibits neuronal cell death by interacting with a cytokine receptor complex or complexes involving CNTF receptor alpha/WSX-1/gp130. Mol Biol Cell. 2009; 20:2864–73. 10.1091/mbc.e09-02-0168
    1. Guo B, Zhai D, Cabezas E, Welsh K, Nouraini S, Satterthwait AC, Reed JC. Humanin peptide suppresses apoptosis by interfering with Bax activation. Nature. 2003; 423:456–61. 10.1038/nature01627
    1. Klein LE, Cui L, Gong Z, Su K, Muzumdar R. A humanin analog decreases oxidative stress and preserves mitochondrial integrity in cardiac myoblasts. Biochem Biophys Res Commun. 2013; 440:197–203. 10.1016/j.bbrc.2013.08.055
    1. Sreekumar PG, Ishikawa K, Spee C, Mehta HH, Wan J, Yen K, Cohen P, Kannan R, Hinton DR. The Mitochondrial-Derived Peptide Humanin Protects RPE Cells From Oxidative Stress, Senescence, and Mitochondrial Dysfunction. Invest Ophthalmol Vis Sci. 2016; 57:1238–53. 10.1167/iovs.15-17053
    1. Zhao ST, Zhao L, Li JH. Neuroprotective Peptide humanin inhibits inflammatory response in astrocytes induced by lipopolysaccharide. Neurochem Res. 2013; 38:581–88. 10.1007/s11064-012-0951-6
    1. Zapała B, Kaczyński Ł, Kieć-Wilk B, Staszel T, Knapp A, Thoresen GH, Wybrańska I, Dembińska-Kieć A. Humanins, the neuroprotective and cytoprotective peptides with antiapoptotic and anti-inflammatory properties. Pharmacol Rep. 2010; 62:767–77. 10.1016/S1734-1140(10)70337-6
    1. Ming W, Lu G, Xin S, Huanyu L, Yinghao J, Xiaoying L, Chengming X, Banjun R, Li W, Zifan L. Mitochondria related peptide MOTS-c suppresses ovariectomy-induced bone loss via AMPK activation. Biochem Biophys Res Commun. 2016; 476:412–19. 10.1016/j.bbrc.2016.05.135
    1. Hodes RJ, Sierra F, Austad SN, Epel E, Neigh GN, Erlandson KM, Schafer MJ, LeBrasseur NK, Wiley C, Campisi J, Sehl ME, Scalia R, Eguchi S, et al.. Disease drivers of aging. Ann N Y Acad Sci. 2016; 1386:45–68. 10.1111/nyas.13299
    1. Demaria M, Ohtani N, Youssef SA, Rodier F, Toussaint W, Mitchell JR, Laberge RM, Vijg J, Van Steeg H, Dollé ME, Hoeijmakers JH, de Bruin A, Hara E, Campisi J. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev Cell. 2014; 31:722–33. 10.1016/j.devcel.2014.11.012
    1. Hartley AV, Martin M, Lu T. Aging: Cancer - an unlikely couple. Aging (Albany NY). 2017; 9:1949–50. 10.18632/aging.101295
    1. Burd CE, Sorrentino JA, Clark KS, Darr DB, Krishnamurthy J, Deal AM, Bardeesy N, Castrillon DH, Beach DH, Sharpless NE. Monitoring tumorigenesis and senescence in vivo with a p16(INK4a)-luciferase model. Cell. 2013; 152:340–51. 10.1016/j.cell.2012.12.010
    1. Kuilman T, Peeper DS. Senescence-messaging secretome: SMS-ing cellular stress. Nat Rev Cancer. 2009; 9:81–94. 10.1038/nrc2560
    1. Özcan S, Alessio N, Acar MB, Mert E, Omerli F, Peluso G, Galderisi U. Unbiased analysis of senescence associated secretory phenotype (SASP) to identify common components following different genotoxic stresses. Aging (Albany NY). 2016; 8:1316–29. 10.18632/aging.100971
    1. Kang TW, Yevsa T, Woller N, Hoenicke L, Wuestefeld T, Dauch D, Hohmeyer A, Gereke M, Rudalska R, Potapova A, Iken M, Vucur M, Weiss S, et al.. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature. 2011; 479:547–51. 10.1038/nature10599
    1. Freund A, Orjalo AV, Desprez PY, Campisi J. Inflammatory networks during cellular senescence: causes and consequences. Trends Mol Med. 2010; 16:238–46. 10.1016/j.molmed.2010.03.003
    1. Fougère B, Boulanger E, Nourhashémi F, Guyonnet S, Cesari M. Chronic Inflammation: Accelerator of Biological Aging. J Gerontol A Biol Sci Med Sci. 2017; 72:1218–25. 10.1093/gerona/glw240
    1. Chang J, Wang Y, Shao L, Laberge RM, Demaria M, Campisi J, Janakiraman K, Sharpless NE, Ding S, Feng W, Luo Y, Wang X, Aykin-Burns N, et al.. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat Med. 2016; 22:78–83. 10.1038/nm.4010
    1. Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, Kirkland JL, van Deursen JM. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011; 479:232–36. 10.1038/nature10600
    1. Jeon OH, Kim C, Laberge RM, Demaria M, Rathod S, Vasserot AP, Chung JW, Kim DH, Poon Y, David N, Baker DJ, van Deursen JM, Campisi J, Elisseeff JH. Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat Med. 2017; 23:775–81. 10.1038/nm.4324
    1. Ogrodnik M, Miwa S, Tchkonia T, Tiniakos D, Wilson CL, Lahat A, Day CP, Burt A, Palmer A, Anstee QM, Grellscheid SN, Hoeijmakers JH, Barnhoorn S, et al.. Cellular senescence drives age-dependent hepatic steatosis. Nat Commun. 2017; 8:15691. 10.1038/ncomms15691
    1. Xu M, Palmer AK, Ding H, Weivoda MM, Pirtskhalava T, White TA, Sepe A, Johnson KO, Stout MB, Giorgadze N, Jensen MD, LeBrasseur NK, Tchkonia T, Kirkland JL. Targeting senescent cells enhances adipogenesis and metabolic function in old age. eLife. 2015; 4:e12997. 10.7554/eLife.12997
    1. Zhu Y, Tchkonia T, Fuhrmann-Stroissnigg H, Dai HM, Ling YY, Stout MB, Pirtskhalava T, Giorgadze N, Johnson KO, Giles CB, Wren JD, Niedernhofer LJ, Robbins PD, Kirkland JL. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell. 2016; 15:428–35. 10.1111/acel.12445
    1. Baar MP, Brandt RM, Putavet DA, Klein JD, Derks KW, Bourgeois BR, Stryeck S, Rijksen Y, van Willigenburg H, Feijtel DA, van der Pluijm I, Essers J, van Cappellen WA, et al.. Targeted Apoptosis of Senescent Cells Restores Tissue Homeostasis in Response to Chemotoxicity and Aging. Cell. 2017; 169:132–147.e16. 10.1016/j.cell.2017.02.031
    1. Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, Palmer AK, Ikeno Y, Hubbard GB, Lenburg M, O’Hara SP, LaRusso NF, Miller JD, et al.. The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell. 2015; 14:644–58. 10.1111/acel.12344
    1. Wiley CD, Campisi J. From Ancient Pathways to Aging Cells-Connecting Metabolism and Cellular Senescence. Cell Metab. 2016; 23:1013–21. 10.1016/j.cmet.2016.05.010
    1. Bittles AH, Harper N. Increased glycolysis in ageing cultured human diploid fibroblasts. Biosci Rep. 1984; 4:751–56. 10.1007/BF01128816
    1. Quijano C, Cao L, Fergusson MM, Romero H, Liu J, Gutkind S, Rovira II, Mohney RP, Karoly ED, Finkel T. Oncogene-induced senescence results in marked metabolic and bioenergetic alterations. Cell Cycle. 2012; 11:1383–92. 10.4161/cc.19800
    1. Wiley CD, Velarde MC, Lecot P, Liu S, Sarnoski EA, Freund A, Shirakawa K, Lim HW, Davis SS, Ramanathan A, Gerencser AA, Verdin E, Campisi J. Mitochondrial Dysfunction Induces Senescence with a Distinct Secretory Phenotype. Cell Metab. 2016; 23:303–14. 10.1016/j.cmet.2015.11.011
    1. Stolzing A, Coleman N, Scutt A. Glucose-induced replicative senescence in mesenchymal stem cells. Rejuvenation Res. 2006; 9:31–35. 10.1089/rej.2006.9.31
    1. Laberge RM, Sun Y, Orjalo AV, Patil CK, Freund A, Zhou L, Curran SC, Davalos AR, Wilson-Edell KA, Liu S, Limbad C, Demaria M, Li P, et al.. MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat Cell Biol. 2015; 17:1049–61. 10.1038/ncb3195
    1. Gao C, Ning B, Sang C, Zhang Y. Rapamycin prevents the intervertebral disc degeneration via inhibiting differentiation and senescence of annulus fibrosus cells. Aging (Albany NY). 2018; 10:131–43. 10.18632/aging.101364
    1. Pascal T, Debacq-Chainiaux F, Chrétien A, Bastin C, Dabée AF, Bertholet V, Remacle J, Toussaint O. Comparison of replicative senescence and stress-induced premature senescence combining differential display and low-density DNA arrays. FEBS Lett. 2005; 579:3651–59. 10.1016/j.febslet.2005.05.056
    1. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009; 324:1029–33. 10.1126/science.1160809
    1. Correia-Melo C, Marques FD, Anderson R, Hewitt G, Hewitt R, Cole J, Carroll BM, Miwa S, Birch J, Merz A, Rushton MD, Charles M, Jurk D, et al.. Mitochondria are required for pro-ageing features of the senescent phenotype. EMBO J. 2016; 35:724–42. 10.15252/embj.201592862
    1. Strigun A, Wahrheit J, Niklas J, Heinzle E, Noor F. Doxorubicin increases oxidative metabolism in HL-1 cardiomyocytes as shown by 13C metabolic flux analysis. Toxicol Sci. 2012; 125:595–606. 10.1093/toxsci/kfr298
    1. Berthiaume JM, Wallace KB. Persistent alterations to the gene expression profile of the heart subsequent to chronic Doxorubicin treatment. Cardiovasc Toxicol. 2007; 7:178–91. 10.1007/s12012-007-0026-0
    1. Tokarska-Schlattner M, Lucchinetti E, Zaugg M, Kay L, Gratia S, Guzun R, Saks V, Schlattner U. Early effects of doxorubicin in perfused heart: transcriptional profiling reveals inhibition of cellular stress response genes. Am J Physiol Regul Integr Comp Physiol. 2010; 298:R1075–88. 10.1152/ajpregu.00360.2009
    1. Yu D, Du Z, Pian L, Li T, Wen X, Li W, Kim SJ, Xiao J, Cohen P, Cui J, Hoffman AR, Hu JF. Mitochondrial DNA Hypomethylation Is a Biomarker Associated with Induced Senescence in Human Fetal Heart Mesenchymal Stem Cells. Stem Cells Int. 2017; 2017:1764549–12. 10.1155/2017/1764549
    1. Lee C, Kim KH, Cohen P. MOTS-c: A novel mitochondrial-derived peptide regulating muscle and fat metabolism. Free Radic Biol Med. 2016; 100:182–87. 10.1016/j.freeradbiomed.2016.05.015
    1. Heinrich PC, Behrmann I, Müller-Newen G, Schaper F, Graeve L. Interleukin-6-type cytokine signalling through the gp130/Jak/STAT pathway. Biochem J. 1998; 334:297–314. 10.1042/bj3340297
    1. Xu M, Tchkonia T, Ding H, Ogrodnik M, Lubbers ER, Pirtskhalava T, White TA, Johnson KO, Stout MB, Mezera V, Giorgadze N, Jensen MD, LeBrasseur NK, Kirkland JL. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc Natl Acad Sci USA. 2015; 112:E6301–10. 10.1073/pnas.1515386112
    1. Wang Z, Wei D, Xiao H. Methods of cellular senescence induction using oxidative stress. Methods Mol Biol. 2013; 1048:135–44. 10.1007/978-1-62703-556-9_11
    1. Demaria M, O’Leary MN, Chang J, Shao L, Liu S, Alimirah F, Koenig K, Le C, Mitin N, Deal AM, Alston S, Academia EC, Kilmarx S, et al.. Cellular Senescence Promotes Adverse Effects of Chemotherapy and Cancer Relapse. Cancer Discov. 2017; 7:165–76. 10.1158/-16-0241
    1. Khan A, Gillis K, Clor J, Tyagarajan K. Simplified evaluation of apoptosis using the Muse cell analyzer. Postepy Biochem. 2012; 58:492–96.
    1. Chin YP, Keni J, Wan J, Mehta H, Anene F, Jia Y, Lue YH, Swerdloff R, Cobb LJ, Wang C, Cohen P. Pharmacokinetics and tissue distribution of humanin and its analogues in male rodents. Endocrinology. 2013; 154:3739–44. 10.1210/en.2012-2004

Source: PubMed

3
Prenumerera