Skip to main content

Advertisement

Log in

Increased DNA damage and repair deficiency in granulosa cells are associated with ovarian aging in rhesus monkey

  • Reproductive Physiology and Disease
  • Published:
Journal of Assisted Reproduction and Genetics Aims and scope Submit manuscript

Abstract

Purpose

Ovarian aging is closely tied to the decline in ovarian follicular reserve and oocyte quality. During the prolonged reproductive lifespan of the female, granulosa cells connected with oocytes play critical roles in maintaining follicle reservoir, oocyte growth and follicular development. We tested whether double-strand breaks (DSBs) and repair in granulosa cells within the follicular reservoir are associated with ovarian aging.

Methods

Ovaries were sectioned and processed for epi-fluorescence microscopy, confocal microscopy, and immunohistochemistry. DNA damage was revealed by immunstaining of γH2AX foci and telomere damage by γH2AX foci co-localized with telomere associated protein TRF2. DNA repair was indicated by BRCA1 immunofluorescence.

Results

DSBs in granulosa cells increase and DSB repair ability, characterized by BRCA1 foci, decreases with advancing age. γH2AX foci increase in primordial, primary and secondary follicles with advancing age. Likewise, telomere damage increases with advancing age. In contrast, BRCA1 foci in granulosa cells of primordial, primary and secondary follicles decrease with monkey age. BRCA1 positive foci in the oocyte nuclei also decline with maternal age.

Conclusions

Increased DSBs and reduced DNA repair in granulosa cells may contribute to ovarian aging. Discovery of therapeutics that targets these pathways might help maintain follicle reserve and postpone ovarian dysfunction with age.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Faddy MJ. Follicle dynamics during ovarian ageing. Mol Cell Endocrinol. 2000;163:43–8.

    Article  CAS  PubMed  Google Scholar 

  2. te Velde ER, Scheffer GJ, Dorland M, Broekmans FJ, Fauser BC. Developmental and endocrine aspects of normal ovarian aging. Mol Cell Endocrinol. 1998;145:67–73.

    Article  Google Scholar 

  3. Faddy MJ, Gosden RG, Gougeon A, Richardson SJ, Nelson JF. Accelerated disappearance of ovarian follicles in mid-life: implications for forecasting menopause. Hum Reprod. 1992;7:1342–6.

    CAS  PubMed  Google Scholar 

  4. Zhang H, Zheng W, Shen Y, Adhikari D, Ueno H, Liu K. Experimental evidence showing that no mitotically active female germline progenitors exist in postnatal mouse ovaries. Proc Natl Acad Sci U S A. 2012;109:12580–5.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  5. Lei L., Spradling A.C. Female mice lack adult germ-line stem cells but sustain oogenesis using stable primordial follicles. Proc Natl Acad Sci U S A 2013;110:8585–90.

  6. Yuan J, Zhang D, Wang L, Liu M, Mao J, Yin Y, et al. No evidence for neo-oogenesis may link to ovarian senescence in adult monkey. Stem Cells. 2013;31:2538–50.

    Article  CAS  PubMed  Google Scholar 

  7. Handel MA, Eppig JJ, Schimenti JC. Applying “gold standards” to in-vitro-derived germ cells. Cell. 2014;157:1257–61.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  8. Ashwood-Smith MJ, Edwards RG. DNA repair by oocytes. Mol Hum Reprod. 1996;2:46–51.

    Article  CAS  PubMed  Google Scholar 

  9. Guli CL, Smyth DR. Lack of effect of maternal age on UV-induced DNA repair in mouse oocytes. Mutat Res. 1989;210:323–8.

    Article  CAS  PubMed  Google Scholar 

  10. Oktem O, Oktay K. A novel ovarian xenografting model to characterize the impact of chemotherapy agents on human primordial follicle reserve. Cancer Res. 2007;67:10159–62.

    Article  CAS  PubMed  Google Scholar 

  11. Soleimani R, Heytens E, Darzynkiewicz Z, Oktay K. Mechanisms of chemotherapy-induced human ovarian aging: double strand DNA breaks and microvascular compromise. Aging (Albany NY). 2011;3:782–93.

    Google Scholar 

  12. Titus S, Li F, Stobezki R, Akula K, Unsal E, Jeong K, et al. Impairment of BRCA1-related DNA double-strand break repair leads to ovarian aging in mice and humans. Sci Transl Med. 2013;5:172ra121.

    Google Scholar 

  13. Nagaoka SI, Hassold TJ, Hunt PA. Human aneuploidy: mechanisms and new insights into an age-old problem. Nat Rev Genet. 2012;13:493–504.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  14. Finch CE. The evolution of ovarian oocyte decline with aging and possible relationships to Down syndrome and Alzheimer disease. Exp Gerontol. 1994;29:299–304.

    Article  CAS  PubMed  Google Scholar 

  15. Wang ZB, Schatten H, Sun QY. Why is chromosome segregation error in oocytes increased with maternal aging? Physiology (Bethesda). 2011;26:314–25.

    Article  CAS  Google Scholar 

  16. Ghosh S, Feingold E, Chakraborty S, Dey SK. Telomere length is associated with types of chromosome 21 nondisjunction: a new insight into the maternal age effect on Down syndrome birth. Hum Genet. 2010;127:403–9.

    Article  PubMed  Google Scholar 

  17. Yamada-Fukunaga T, Yamada M, Hamatani T, Chikazawa N, Ogawa S, Akutsu H, et al. Age-associated telomere shortening in mouse oocytes. Reprod Biol Endocrinol. 2013;11:108.

    Article  PubMed Central  PubMed  Google Scholar 

  18. Liu L, Franco S, Spyropoulos B, Moens PB, Blasco MA, Keefe DL. Irregular telomeres impair meiotic synapsis and recombination in mice. Proc Natl Acad Sci U S A. 2004;101:6496–501.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  19. Keefe DL, Liu L. Telomeres and reproductive aging. Reprod Fertil Dev. 2009;21:10–4.

    Article  CAS  PubMed  Google Scholar 

  20. Treff NR, Su J, Taylor D, Scott Jr RT. Telomere DNA deficiency is associated with development of human embryonic aneuploidy. PLoS Genet. 2011;7:e1002161.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  21. Eppig JJ, Chesnel F, Hirao Y, O’Brien MJ, Pendola FL, Watanabe S, et al. Oocyte control of granulosa cell development: how and why. Hum Reprod. 1997;12:127–32.

    CAS  PubMed  Google Scholar 

  22. Su YQ, Sugiura K, Eppig JJ. Mouse oocyte control of granulosa cell development and function: paracrine regulation of cumulus cell metabolism. Semin Reprod Med. 2009;27:32–42.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  23. Li Q, McKenzie LJ, Matzuk MM. Revisiting oocyte-somatic cell interactions: in search of novel intrafollicular predictors and regulators of oocyte developmental competence. Mol Hum Reprod. 2008;14:673–8.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  24. Tatone C, Amicarelli F. The aging ovary–the poor granulosa cells. Fertil Steril. 2013;99:12–7.

    Article  CAS  PubMed  Google Scholar 

  25. Cheng EH, Chen SU, Lee TH, Pai YP, Huang LS, Huang CC, et al. Evaluation of telomere length in cumulus cells as a potential biomarker of oocyte and embryo quality. Hum Reprod. 2013;28:929–36.

    Article  CAS  PubMed  Google Scholar 

  26. Dzafic E, Stimpfel M, Virant-Klun I. Plasticity of granulosa cells: on the crossroad of stemness and transdifferentiation potential. J Assist Reprod Genet. 2013;30:1255–61.

    Article  PubMed Central  PubMed  Google Scholar 

  27. Bayne S, Li H, Jones ME, Pinto AR, van Sinderen M, Drummond A, et al. Estrogen deficiency reversibly induces telomere shortening in mouse granulosa cells and ovarian aging in vivo. Protein Cell. 2011;2:333–46.

    Article  CAS  PubMed  Google Scholar 

  28. Ozturk S, Sozen B, Demir N. Telomere length and telomerase activity during oocyte maturation and early embryo development in mammalian species. Mol Hum Reprod. 2014;20:15–30.

    Article  CAS  PubMed  Google Scholar 

  29. Blasco MA. Telomere length, stem cells and aging. Nat Chem Biol. 2007;3:640–9.

    Article  CAS  PubMed  Google Scholar 

  30. Collado M, Blasco MA, Serrano M. Cellular senescence in cancer and aging. Cell. 2007;130:223–33.

    Article  CAS  PubMed  Google Scholar 

  31. Mermershtain I, Glover JN. Structural mechanisms underlying signaling in the cellular response to DNA double strand breaks. Mutat Res. 2013;750:15–22.

    Article  CAS  PubMed  Google Scholar 

  32. Caestecker KW, Van de Walle GR. The role of BRCA1 in DNA double-strand repair: past and present. Exp Cell Res. 2013;319:575–87.

    Article  CAS  PubMed  Google Scholar 

  33. Rosen EM. BRCA1 in the DNA damage response and at telomeres. Front Genet. 2013;4:85.

    Article  PubMed Central  PubMed  Google Scholar 

  34. Robson M, Gilewski T, Haas B, Levin D, Borgen P, Rajan P, et al. BRCA-associated breast cancer in young women. J Clin Oncol. 1998;16:1642–9.

    CAS  PubMed  Google Scholar 

  35. Kauff ND, Satagopan JM, Robson ME, Scheuer L, Hensley M, Hudis CA, et al. Risk-reducing salpingo-oophorectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med. 2002;346:1609–15.

    Article  PubMed  Google Scholar 

  36. Rzepka-Gorska I, Tarnowski B, Chudecka-Glaz A, Gorski B, Zielinska D, Toloczko-Grabarek A. Premature menopause in patients with BRCA1 gene mutation. Breast Cancer Res Treat. 2006;100:59–63.

    Article  CAS  PubMed  Google Scholar 

  37. Johnson J, Keefe DL. Ovarian aging: breaking up is hard to fix. Sci Transl Med. 2013;5:172fs175.

    Google Scholar 

  38. Martinez-Delgado B, Yanowsky K, Inglada-Perez L, de la Hoya M, Caldes T, Vega A, et al. Shorter telomere length is associated with increased ovarian cancer risk in both familial and sporadic cases. J Med Genet. 2012;49:341–4.

    Article  PubMed  Google Scholar 

  39. Cabuy E, Newton C, Slijepcevic P. BRCA1 knock-down causes telomere dysfunction in mammary epithelial cells. Cytogenet Genome Res. 2008;122:336–42.

    Article  CAS  PubMed  Google Scholar 

  40. McPherson JP, Hande MP, Poonepalli A, Lemmers B, Zablocki E, Migon E, et al. A role for Brca1 in chromosome end maintenance. Hum Mol Genet. 2006;15:831–8.

    Article  CAS  PubMed  Google Scholar 

  41. Wang X, Liu L, Montagna C, Ried T, Deng CX. Haploinsufficiency of Parp1 accelerates Brca1-associated centrosome amplification, telomere shortening, genetic instability, apoptosis, and embryonic lethality. Cell Death Differ. 2007;14:924–31.

    Article  CAS  PubMed  Google Scholar 

  42. Nichols SM, Bavister BD, Brenner CA, Didier PJ, Harrison RM, Kubisch HM. Ovarian senescence in the rhesus monkey (Macaca mulatta). Hum Reprod. 2005;20:79–83.

    Article  CAS  PubMed  Google Scholar 

  43. Gilardi KV, Shideler SE, Valverde CR, Roberts JA, Lasley BL. Characterization of the onset of menopause in the rhesus macaque. Biol Reprod. 1997;57:335–40.

    Article  CAS  PubMed  Google Scholar 

  44. Shideler SE, Gee NA, Chen J, Lasley BL. Estrogen and progesterone metabolites and follicle-stimulating hormone in the aged macaque female. Biol Reprod. 2001;65:1718–25.

    Article  CAS  PubMed  Google Scholar 

  45. Liu M, Yin Y, Ye X, Zeng M, Zhao Q, Keefe DL, et al. Resveratrol protects against age-associated infertility in mice. Hum Reprod. 2013;28:707–17.

    Article  CAS  PubMed  Google Scholar 

  46. Cawthon RM. Telomere measurement by quantitative PCR. Nucleic Acids Res. 2002;30:e47.

    Article  PubMed Central  PubMed  Google Scholar 

  47. Lowndes NF, Toh GW. DNA repair: the importance of phosphorylating histone H2AX. Curr Biol. 2005;15:R99–102.

    Article  CAS  PubMed  Google Scholar 

  48. Takai H, Smogorzewska A, de Lange T. DNA damage foci at dysfunctional telomeres. Curr Biol. 2003;13:1549–56.

    Article  CAS  PubMed  Google Scholar 

  49. Feng L, Fong KW, Wang J, Wang W, Chen J. RIF1 counteracts BRCA1-mediated end resection during DNA repair. J Biol Chem. 2013;288:11135–43.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  50. Beerman I, Seita J, Inlay MA, Weissman IL, Rossi DJ. Quiescent hematopoietic stem cells accumulate DNA damage during aging that is repaired upon entry into cell cycle. Cell Stem Cell. 2014;15:37–50.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  51. Stoop H, Honecker F, Cools M, de Krijger R, Bokemeyer C, Looijenga LH. Differentiation and development of human female germ cells during prenatal gonadogenesis: an immunohistochemical study. Hum Reprod. 2005;20:1466–76.

    Article  CAS  PubMed  Google Scholar 

  52. Starborg M, Gell K, Brundell E, Hoog C. The murine Ki-67 cell proliferation antigen accumulates in the nucleolar and heterochromatic regions of interphase cells and at the periphery of the mitotic chromosomes in a process essential for cell cycle progression. J Cell Sci. 1996;109(Pt 1):143–53.

    CAS  PubMed  Google Scholar 

  53. Funayama Y, Sasano H, Suzuki T, Tamura M, Fukaya T, Yajima A. Cell turnover in normal cycling human ovary. J Clin Endocrinol Metab. 1996;81:828–34.

    CAS  PubMed  Google Scholar 

  54. Scalercio S.R., Brito A.B..., Domingues S.F., Santos R.R., Amorim C.A. Immunolocalization of growth, inhibitory, and proliferative factors involved in initial ovarian folliculogenesis from adult common squirrel monkey (Saimiri collinsi). Reprod Sci 2015;22:68–74.

  55. Oktay K, Kim JY, Barad D, Babayev SN. Association of BRCA1 mutations with occult primary ovarian insufficiency: a possible explanation for the link between infertility and breast/ovarian cancer risks. J Clin Oncol. 2010;28:240–4.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  56. Leng M, Li G, Zhong L, Hou H, Yu D, Shi Q. Abnormal synapses and recombination in an azoospermic male carrier of a reciprocal translocation t(1;21). Fertil Steril. 2009;91:1293 e1217–1222.

    Article  Google Scholar 

  57. Ferguson KA, Chow V, Ma S. Silencing of unpaired meiotic chromosomes and altered recombination patterns in an azoospermic carrier of a t(8;13) reciprocal translocation. Hum Reprod. 2008;23:988–95.

    Article  CAS  PubMed  Google Scholar 

  58. Herbig U, Ferreira M, Condel L, Carey D, Sedivy JM. Cellular senescence in aging primates. Science. 2006;311:1257.

    Article  CAS  PubMed  Google Scholar 

  59. Agarwal A, Gupta S, Sharma R. Oxidative stress and its implications in female infertility - a clinician’s perspective. Reprod BioMed Online. 2005;11:641–50.

    Article  CAS  PubMed  Google Scholar 

  60. Liu L, Trimarchi JR, Navarro P, Blasco MA, Keefe DL. Oxidative stress contributes to arsenic-induced telomere attrition, chromosome instability, and apoptosis. J Biol Chem. 2003;278:31998–2004.

    Article  CAS  PubMed  Google Scholar 

  61. Liu J, Liu M, Ye X, Liu K, Huang J, Wang L, et al. Delay in oocyte aging in mice by the antioxidant N-acetyl-L-cysteine (NAC). Hum Reprod. 2012;27:1411–20.

    Article  CAS  PubMed  Google Scholar 

  62. Wang ET, Pisarska MD, Bresee C, Ida Chen YD, Lester J, Afshar Y, et al. BRCA1 germline mutations may be associated with reduced ovarian reserve. Fertil Steril. 2014;102:1723–8.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported by MOST of China National Basic Research Program (2010CB94500), National Natural Science Foundation of China (31430052), and PCSIRT (No. IRT13023). The authors have no competing interests to declare.

Conflict of interest

The authors declare no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to David L. Keefe or Lin Liu.

Additional information

Capsule Increased DSBs and reduced DNA repair in granulosa cells may contribute to ovarian aging.

Electronic supplementary material

Below is the link to the electronic supplementary material.

ESM 1

(DOCX 2818 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, D., Zhang, X., Zeng, M. et al. Increased DNA damage and repair deficiency in granulosa cells are associated with ovarian aging in rhesus monkey. J Assist Reprod Genet 32, 1069–1078 (2015). https://doi.org/10.1007/s10815-015-0483-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10815-015-0483-5

Keywords

Navigation