Nutr Res Pract.  2007 Jun;1(2):105-112.

Ascorbic acid extends replicative life span of human embryonic fibroblast by reducing DNA and mitochondrial damages

Affiliations
  • 1Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Gangwon 200-702, Korea. jyolee@hallym.ac.kr

Abstract

Ascorbic acid has been reported to extend replicative life span of human embryonic fibroblast (HEF). Since the detailed molecular mechanism of this phenomenon has not been investigated, we attempted to elucidate. Continuous treatment of HEF cells with ascorbic acid (at 200 micrometer) from 40 population doubling (PD) increased maximum PD numbers by 18% and lowered SA-beta-gal positive staining, an aging marker, by 2.3 folds, indicating that ascorbic acid extends replicative life span of HEF cells. Ascorbic acid treatment lowered DCFH by about 7 folds and Rho123 by about 70%, suggesting that ascorbic acid dramatically decreased ROS formation. Ascorbic acid also increased aconitase activity, a marker of mitochondrial aging, by 41%, indicating that ascorbic acid treatment restores age-related decline of mitochondrial function. Cell cycle analysis by flow cytometry revealed that ascorbic acid treatment decreased G1 population up to 12%. Further western blot analysis showed that ascorbic acid treatment decreased levels of p53, phospho-p53 at ser 15, and p21, indicating that ascorbic acid relieved senescence-related G1 arrest. Analysis of AP (apurinic/apyrimidinic) sites showed that ascorbic acid treatment decreased AP site formation by 35%. We also tested the effect of hydrogen peroxide treatment, as an additional oxidative stress. Continuous treatment of 20 micrometer of hydrogen peroxide from PD 40 of HEF cells resulted in premature senescence due to increased ROS level, and increased AP sites. Taken together, the results suggest that ascorbic acid extends replicative life span of HEF cells by reducing mitochondrial and DNA damages through lowering cellular ROS.

Keyword

Ascorbic acid, human embryonic fibroblasts, replicative life span, mitochondrial damage, DNA damage; reactive oxygen species

MeSH Terms

Aconitate Hydratase
Aging
Ascorbic Acid*
Blotting, Western
Cell Cycle
DNA Damage
DNA*
Fibroblasts*
Flow Cytometry
Humans*
Hydrogen Peroxide
Oxidative Stress
Reactive Oxygen Species
Aconitate Hydratase
Ascorbic Acid
DNA
Hydrogen Peroxide
Reactive Oxygen Species

Figure

  • Fig. 1 The effect of ascorbic acid on growth and replicative life span of human embryonic fibroblast (HEF) cells. HEF cells were cultivated continuously from PD 40 with ascorbic acid and growth curve was prepared as described in "Materials and Methods". (A) The effect of ascorbic acid on replicative life span of HEF cells. The treatments of ascorbic acid are as follows; untreated (♦), 2 µM ascorbic acid (▾), 20 µM ascorbic acid (▴), 200 µM ascorbic acid (▪). (B) The effect of ascorbic acid and hydrogen peroxide on replicative life span of HEF cells. HEF cells were treated continuously with 20 µM hydrogen peroxide from PD 40. The treatments are as follows; 20 µM H2O2 (♦), 2 µM ascorbic acid and 20µM H2O2 (▾), 20 µM ascorbic acid and 20 µM H2O2 (▴), 200 µM ascorbic acid and 20 µM H2O2 (▪).

  • Fig. 2 The effect of ascorbic acid on SA-β-gal staining in HEF cells. (A) HEF cells were seeded at an initial density of 0.5 × 106/dish at PD 40 and cultivated continuously with ascorbic acid. SA-β-gal staining was performed (a) untreated old HEF cells (PD 64) (b) untreated young HEF cells (PD 30) (c) HEF cells treated with 200 µM ascorbic acid (PD 64) (d) HEF cells treated with 20 µM hydrogen peroxide (PD 56). (B) The percentage of SA-β-gal positive cells. SA-β-gal staining was performed on ascorbic acid-treated HEF cells (PD 64), untreated old HEF cells (PD 64), and H2O2-treated HEF cells (PD 58). The diagram was prepared from the staining result.

  • Fig. 3 Analysis of DCFH oxidation for measuring cellular ROS level in ascorbic acid-treated HEF cells. (A) Analysis of DCFH oxidation in ascorbic acid-treated HEF cells. HEF cells were treated with ascorbic acid beginning from PD 40. Oxidation of DCFH was determined at PD 64. (B) Analysis of DCFH oxidation in ascorbic acid/ H2O2-treated HEF cells. HEF cells were co-treated with 20 µM hydrogen peroxide and ascorbic acid beginning from PD 40.

  • Fig. 4 Analyses of Rho123 accumulation and aconitase activity as markers of mitochondrial function in ascorbic acid-treated HEF cells. HEF cells were treated with ascorbic acid beginning from PD 40. Ascorbic acidtreated HEF cells were harvested at PD 64 and used for analysis. (A) Analysis of Rho123 accumulation in ascorbic acid-treated HEF cells. Accumulation of Rho123 was determined as described in "Materials and Methods". (B) Analysis of Rho123 accumulation in ascorbic acid/ H2O2-treated HEF cells. HEF cells were co-treated with 20 µM hydrogen peroxide and ascorbic acid. Analysis of Rho123 accumulation was performed in old untreated HEF cells (PD 64) and H2O2-treated HEF cells (PD 58). (C) Analysis of aconitase activity in ascorbic acid-treated HEF cells. Cell extracts were promptly assayed for aconitase activity as described in "Materials and Methods".

  • Fig. 5 Cell cycle analysis of ascorbic acid-treated HEF cells. Ascorbic acid-treated HEF cells were stained with propidium iodide and cell cycle was determined by FACS analysis. Upper panel represents the results of FACS analysis and lower panel shows a plot prepared from above results. (A) Untreated old HEF cells (PD 62), (B) H2O2-treated old HEF cells (PD 56), (C) ascorbic acid-treated HEF cells (2 µM, PD 62), (D) ascorbic acid-treated HEF cells (20 µM, PD 62), (E) ascorbic acid-treated HEF cells (200 µM, PD 62).

  • Fig. 6 Western blot analysis of cell cycle marker proteins. Ascorbic acid-treated HEF cells were harvested and cell extracts were prepared. Western blot analysis was performed for cell extracts using specific antibodies against p21, p53, and phospho-p53 at ser 15. β-actin was used as a protein loading control. Lane 1; untreated old HEF cells (PD 54), lane 2; ascorbic acid-treated HEF cells (200 µM, PD 54), lane 3; ascorbic acid-treated HEF cells (200 µM, PD 64), lane 4; untreated old HEF cells (PD 64), lane 5; ascorbic acid-treated HEF cells (200 µM, PD 72).

  • Fig. 7 Analysis of AP sites in ascorbic acid-treated HEF cells. AP site were determined in ascorbic acid-treated HEF cells as described under "Materials and Methods". Samples were as follows; untreated-young HEF cells (PD 30), untreated-old HEF cells (PD 64), ascorbic acid-treated old HEF cells (2, 20, and 200 µM ascorbic acid, PD 64), and H2O2-treated HEF cells (PD 58).


Reference

1. Amer MA. Modulation of age-related biochemical changes and oxidative stress by ascorbic acid and glutathione supplementation in old rats. Ann Nutr Metab. 2002. 46:165–168.
Article
2. Barja G, Herrero A. Oxidative damage to mitochondrial DNA is inversely related to maximum life span in the heart and brain of mammals. Faseb J. 2000. 14:312–318.
Article
3. Campisi J. From cells to organisms: Can we learn about aging from cells in culture. Exp Gerontol. 2001. 36:607–618.
Article
4. Carty JL, Bevan R, Waller H, Mistry N, Cooke M, Lunec J, Griffiths HR. The effects of ascorbic acid supplementation on protein oxidation in healthy volunteers. Biochem Biophys Res Commun. 2000. 273:729–735.
Article
5. Chen LH, Boissonneault GA, Glauert HP. Ascorbic acid, vitamin E and cancer. Anticancer Res. 1988. 8:739–748.
6. Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, Mederano EE, Rubely I, Pereira-Smith O, Peacocke M, Campisi J. A biomarker that identifies senescence human cells in culture and in aging skin in vivo. Proc Natl Acad Sci USA. 1995. 92:9363–9367.
Article
7. Fagagna F, Reaper PM, Clay-Farrace L, Fiegler H, Carr P, Von Zglinicki T. A DNA damage checkpoint response in telomere-initiated senescence. Nature. 2003. 426:194–198.
Article
8. Farriol M, Mourelle M, Schwartz S. Effect of ascorbic acid and vitamin E analog on aged fibroblasts. Rev Esp Fisiol. 1994. 50:253–257.
9. Fumeron C, Nguyen-Khoa T, Saltiel C, Kebede M, Buisson C, Drüeke TB, Lacour B, Massy ZA. Effects of oral vitamin C supplementation on oxidative stress and inflammation status in haemodialysis patients. Nephrol Dial Transplant. 2005. 20:1874–1879.
Article
10. Gorenne I, Kavurma M, Scott S, Bennett M. Vascular smooth muscle cell senescence in atherosclerosis. Cardiovasc Res. 2006. 72:9–17.
Article
11. Hamilton ML, Van Remmen H, Drake JA, Yang H, Guo ZM, Kewitt K, Walter CA, Richardson A. Does oxidative damage to DNA increase with age? Proc Natl Acad Sci USA. 2001. 98:10469–10474.
Article
12. Hart RW, Setlow RB. Correlation between deoxyribonucleic acid excision-repair and life-span in a number of mammalian species. Proc Natl Acad Sci USA. 1974. 71:2169–2173.
Article
13. Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961. 25:585–621.
Article
14. Holloszy JO. Longevity of exercising male rats: effect of an antioxidant supplemented diet. Mech Ageing Dev. 1998. 100:211–219.
Article
15. Huang HY, Appel LJ, Croft KD, Miller ER 3rd, Mori TA, Puddey IB. Effects of ascorbic acid and vitamin E on in vivo lipid peroxidation: results of a randomized controlled trial. Am J Clin Nutr. 2002. 76:549–555.
Article
16. Kashino G, Kodama S, Nakayama Y, Suzuki K, Fukase K, Goto M, Watanabe M. Relief of oxidative stress by ascorbic acid delays cellular senescence of normal human and Werner syndrome fibroblast cells. Free Radic Biol Med. 2003. 35:438–443.
Article
17. Kim HS, Yeo EJ, Park SH, Park JI, Park SC, Shin JY, Kim MJ, Oh SJ, Won MH, Kang TC, Park JB, Kim J, Kim JI, Lee HY, Lee JY. p21WAF/CIP1/SDI1 is upregulated due to increased mRNA stability during hydroxyurea-induced senescence of human fibroblasts. Mech Ageing Dev. 2005. 126:1255–1261.
Article
18. Lenton KJ, Therriault H, Fülöp T, Payette H, Wagner JR. Glutathione and ascorbate are negatively correlated with oxidative DNA damage in human lymphocytes. Carcinogenesis. 1999. 20:607–613.
Article
19. Levine AJ. p.53, the cellular gatekeeper for growth and division. Cell. 1997. 88:323–331.
Article
20. Levine RL. Carbonyl modified proteins in cellular regulation, aging, and disease. Free Radic Biol Med. 2002. 32:790–796.
21. Li M, Chiu JF, Mossman BT, Fukagawa NK. Down-regulation of manganese-superoxide dismutase through phosphorylation of FOXO3a by Akt in explanted vascular smooth muscle cells from old rats. J Biol Chem. 2006. 281:40429–40439.
Article
22. Liang JY, Rodeny LL, Rajindar SS. Oxidative damage during aging targets mitochondrial aconitase. Proc Natl Acad Sci USA. 1997. 94:11168–11172.
23. Lombard DB, Chua KF, Mostoslavsky R, Franco S, Gostissa M, Alt FW. DNA repair, genome stability, and aging. Cell. 2005. 120:497–512.
Article
24. Massie HR, Aiello VR, Doherty TJ. Dietary ascorbic acid improves the survival of mice. Gerontology. 1984. 30:371–375.
25. Mostoslavsky R, Chua KF, Lombard DB, Pang WW, Fischer MR, Gellon L, Liu P, Mostoslavsky G, Franco S, Murphy MM, Mills KD, Patel P, Hsu JT, Hong AL, Ford E, Cheng HL, Kennedy C, Nunez N, Bronson R, Frendewey D, Auerbach W, Valenzuela D, Karow M, Hottiger MO, Hursting S, Barrett JC, Guarente L, Mulligan R, Demple B, Yancopoulos GD, Alt FW. Genomic Instability and Aging-like Phenotype in the Absence of Mammalian SIRT6. Cell. 2006. 124:315–329.
Article
26. Nakamura J, Walker VE, Upton PB, Chiang S, Kow YW, Swenberg JA. Highly sensitive apurinic/apyrimidinic site assay can detect spontaneous and chemically induced depurinaton under physiological conditions. Cancer Res. 1998. 58:222–225.
27. Pamplona R, Portero-Otin M, Ruiz C, Gredilla R, Herrero A, Barja G. Double bond content of phospholipids and lipid peroxidation negatively correlate with maximum longevity in the heart of mammals. Mech Ageing Dev. 2000. 112:169–183.
Article
28. Parrinello S, Samper E, Krtolica A, Goldstein J, Melov S, Campisi J. Oxygen sensitivity severely limits the replicative lifespan of murine fibroblasts. Nat Cell Biol. 2003. 5:741–747.
Article
29. Petersen S, Saretzki G, von Zglinicki T. Preferential accumulation of single-stranded regions in telomeres of human fibroblasts. Exp Cell Res. 1998. 239:152–160.
Article
30. Selman C, McLaren JS, Meyer C, Duncan JS, Redman P, Collins AR, Duthie GG, Speakman JR. Life-long ascorbic acid supplementation in combination with cold exposure does not affect oxidative damage or lifespan in mice, but decreases expression of antioxidant protection genes. Mech Ageing Dev. 2006. 127:897–904.
Article
31. Serrano M, Blasco MA. Putting the stress on senescence. Curr Opin Cell Biol. 2001. 13:748–753.
Article
32. Sharma GG, Gupta A, Wang H, Scherthan H, Dhar S, Gandhi V. hTERT associates with human telomeres and enhances genomic stability and DNA repair. Oncogene. 2003. 22:131–146.
Article
33. Smogorzewska A, de Lange T. Different telomere damage signaling pathways in human and mouse cells. EMBO J. 2002. 21:4338–4348.
Article
34. Veurink G, Liu D, Taddei K, Perry G, Smith MA, Robertson TA, Hone E, Groth DM, Atwood CS, Martins RN. Reduction of inclusion body pathology in ApoE-deficient mice fed a combination of antioxidants. Free Radic Biol Med. 2003. 34:1070–1077.
Article
35. Vogel DS, Lim G, Karsenty M, Finegold , Hasty P. Deletion of Ku86 causes early onset of senescence in mice. Proc Natl Acad Sci USA. 1999. 96:10770–10775.
Article
Full Text Links
  • NRP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr