Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
REVIEW

Oogonial stem cells as a model to study age-associated infertility in women

Neha Garg A B D and David A. Sinclair A B C
+ Author Affiliations
- Author Affiliations

A Glenn Laboratories for the Biological Mechanisms of Aging, Harvard Medical School, Boston, MA 02115, USA.

B Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.

C Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia.

D Corresponding author. Email: neha_garg@hms.harvard.edu

Reproduction, Fertility and Development 27(6) 969-974 https://doi.org/10.1071/RD14461
Submitted: 21 November 2014  Accepted: 14 March 2015   Published: 22 April 2015

Abstract

Fertility is the first biological process to break down during aging, thereby making it a useful tool to understand fundamental processes of aging. Reproductive aging in females is associated with a loss of ovarian function characterised by a reduction in the number and quality of oocytes. The central dogma, namely that females are born with a fixed pool of oocytes that progressively decline with increasing maternal age, has been challenged by evidence supporting postnatal oogenesis in mammals. Reports demonstrating formation of new oocytes from newly discovered germline stem cells, referred to as oogonial stem cells (OSCs), has opened new avenues for treatment of female infertility. In this review we discuss why the OSCs possibly lose their regenerative potential over time, and focus specifically on the aging process in germline stem cells as a possible mechanism for understanding female age-related infertility and how we can slow or delay ovarian aging.

Additional keywords: germline stem cells, meiosis, oocyte, postnatal oogenesis.


References

Anderson, E. L., Baltus, A. E., Roepers-Gajadien, H. L., Hassold, T. J., de Rooij, D. G., van Pelt, A. M., and Page, D. C. (2008). Stra8 and its inducer, retinoic acid, regulate meiotic initiation in both spermatogenesis and oogenesis in mice. Proc. Natl Acad. Sci. USA 105, 14 976–14 980.
Stra8 and its inducer, retinoic acid, regulate meiotic initiation in both spermatogenesis and oogenesis in mice.Crossref | GoogleScholarGoogle Scholar |

Angelo, G., and Van Gilst, M. R. (2009). Starvation protects germline stem cells and extends reproductive longevity in C. elegans. Science 326, 954–958.
Starvation protects germline stem cells and extends reproductive longevity in C. elegans.Crossref | GoogleScholarGoogle Scholar | 19713489PubMed |

Balasch, J. (2010). Ageing and infertility: an overview. Gynecol. Endocrinol. 26, 855–860.
Ageing and infertility: an overview.Crossref | GoogleScholarGoogle Scholar | 20642380PubMed |

Baltus, A. E., Menke, D. B., Hu, Y. C., Goodheart, M. L., Carpenter, A. E., de Rooij, D. G., and Page, D. C. (2006). In germ cells of mouse embryonic ovaries, the decision to enter meiosis precedes premeiotic DNA replication. Nat. Genet. 38, 1430–1434.
In germ cells of mouse embryonic ovaries, the decision to enter meiosis precedes premeiotic DNA replication.Crossref | GoogleScholarGoogle Scholar | 17115059PubMed |

Bentov, Y., Yavorska, T., Esfandiari, N., Jurisicova, A., and Casper, R. F. (2011). The contribution of mitochondrial function to reproductive aging. J. Assist. Reprod. Genet. 28, 773–783.
The contribution of mitochondrial function to reproductive aging.Crossref | GoogleScholarGoogle Scholar | 21617930PubMed |

Borum, K. (1961). Oogenesis in the mouse. A study of the meiotic prophase. Exp. Cell Res. 24, 495–507.
Oogenesis in the mouse. A study of the meiotic prophase.Crossref | GoogleScholarGoogle Scholar | 13871511PubMed |

Bowles, J., and Koopman, P. (2007). Retinoic acid, meiosis and germ cell fate in mammals. Development 134, 3401–3411.
Retinoic acid, meiosis and germ cell fate in mammals.Crossref | GoogleScholarGoogle Scholar | 17715177PubMed |

Channing, C. P., Schaerf, F. W., Anderson, L. D., and Tsafriri, A. (1980). Ovarian follicular and luteal physiology. Int. Rev. Physiol. 22, 117–201.
| 6248477PubMed |

De Felici, M., Klinger, F. G., Farini, D., Scaldaferri, M. L., Iona, S., and Lobascio, M. (2005). Establishment of oocyte population in the fetal ovary: primordial germ cell proliferation and oocyte programmed cell death. Reprod. Biomed. Online 10, 182–191.
Establishment of oocyte population in the fetal ovary: primordial germ cell proliferation and oocyte programmed cell death.Crossref | GoogleScholarGoogle Scholar | 15823221PubMed |

Dunlop, C. E., Telfer, E. E., and Anderson, R. A. (2014). Ovarian germline stem cells. Stem Cell Res. Ther. 5, 98.
Ovarian germline stem cells.Crossref | GoogleScholarGoogle Scholar | 25157949PubMed |

Gomes, A. P., Price, N. L., Ling, A. J., Moslehi, J. J., Montgomery, M. K., Rajman, L., White, J. P., Teodoro, J. S., Wrann, C. D., Hubbard, B. P., Mercken, E. M., Palmeira, C. M., de Cabo, R., Rolo, A. P., Turner, N., Bell, E. L., and Sinclair, D. A. (2013). Declining NAD(+) induces a pseudohypoxic state disrupting nuclear–mitochondrial communication during aging. Cell 155, 1624–1638.
Declining NAD(+) induces a pseudohypoxic state disrupting nuclear–mitochondrial communication during aging.Crossref | GoogleScholarGoogle Scholar | 24360282PubMed |

Govindaraj, V., Keralapura Basavaraju, R., and Rao, A. J. (2015). Changes in the expression of DNA double strand break repair genes in primordial follicles from immature and aged rats. Reprod. Biomed. Online 30, 303–310.
Changes in the expression of DNA double strand break repair genes in primordial follicles from immature and aged rats.Crossref | GoogleScholarGoogle Scholar | 25599822PubMed |

Greenfeld, C., and Flaws, J. A. (2004). Renewed debate over postnatal oogenesis in the mammalian ovary. Bioessays 26, 829–832.
Renewed debate over postnatal oogenesis in the mammalian ovary.Crossref | GoogleScholarGoogle Scholar | 15273985PubMed |

Guarente, L. (2013). Calorie restriction and sirtuins revisited. Genes Dev. 27, 2072–2085.
Calorie restriction and sirtuins revisited.Crossref | GoogleScholarGoogle Scholar | 24115767PubMed |

Guarente, L., and Picard, F. (2005). Calorie restriction: the SIR2 connection. Cell 120, 473–482.
Calorie restriction: the SIR2 connection.Crossref | GoogleScholarGoogle Scholar | 15734680PubMed |

Hassold, T., and Chiu, D. (1985). Maternal age-specific rates of numerical chromosome abnormalities with special reference to trisomy. Hum. Genet. 70, 11–17.
Maternal age-specific rates of numerical chromosome abnormalities with special reference to trisomy.Crossref | GoogleScholarGoogle Scholar | 3997148PubMed |

Holliday, R. (1989). Food, reproduction and longevity: is the extended lifespan of calorie-restricted animals an evolutionary adaptation? Bioessays 10, 125–127.
Food, reproduction and longevity: is the extended lifespan of calorie-restricted animals an evolutionary adaptation?Crossref | GoogleScholarGoogle Scholar | 2730632PubMed |

Howitz, K. T., Bitterman, K. J., Cohen, H. Y., Lamming, D. W., Lavu, S., Wood, J. G., Zipkin, R. E., Chung, P., Kisielewski, A., Zhang, L. L., Scherer, B., and Sinclair, D. A. (2003). Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature 425, 191–196.
Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan.Crossref | GoogleScholarGoogle Scholar | 12939617PubMed |

Hsu, H. J., and Drummond-Barbosa, D. (2009). Insulin levels control female germline stem cell maintenance via the niche in Drosophila. Proc. Natl Acad. Sci. USA 106, 1117–1121.
Insulin levels control female germline stem cell maintenance via the niche in Drosophila.Crossref | GoogleScholarGoogle Scholar | 19136634PubMed |

Huang, Y. H., Lin, M. H., Wang, P. C., Wu, Y. C., Chiang, H. L., Wang, Y. L., Chang, J. H., Huang, Y. K., Gu, S. Y., Ho, H. N., and Ling, T. Y. (2014). Hypoxia inducible factor 2alpha/insulin-like growth factor receptor signal loop supports the proliferation and Oct-4 maintenance of mouse germline stem cells. Mol. Hum. Reprod. 20, 526–537.
Hypoxia inducible factor 2alpha/insulin-like growth factor receptor signal loop supports the proliferation and Oct-4 maintenance of mouse germline stem cells.Crossref | GoogleScholarGoogle Scholar | 24598112PubMed |

Johnson, J., Canning, J., Kaneko, T., Pru, J. K., and Tilly, J. L. (2004). Germline stem cells and follicular renewal in the postnatal mammalian ovary. Nature 428, 145–150.
Germline stem cells and follicular renewal in the postnatal mammalian ovary.Crossref | GoogleScholarGoogle Scholar | 15014492PubMed |

Jones, D. L., and Rando, T. A. (2011). Emerging models and paradigms for stem cell ageing. Nat. Cell Biol. 13, 506–512.
Emerging models and paradigms for stem cell ageing.Crossref | GoogleScholarGoogle Scholar | 21540846PubMed |

Kezele, P., Nilsson, E., and Skinner, M. K. (2002). Cell–cell interactions in primordial follicle assembly and development. Front. Biosci. 7, d1990–d1996.
Cell–cell interactions in primordial follicle assembly and development.Crossref | GoogleScholarGoogle Scholar | 12161345PubMed |

Kirilly, D., and Xie, T. (2007). The Drosophila ovary: an active stem cell community. Cell Res. 17, 15–25.
The Drosophila ovary: an active stem cell community.Crossref | GoogleScholarGoogle Scholar | 17199109PubMed |

Kirkwood, T. B., and Holliday, R. (1979). The evolution of ageing and longevity. Proc. R. Soc. Lond. B Biol. Sci. 205, 531–546.
The evolution of ageing and longevity.Crossref | GoogleScholarGoogle Scholar |

LaFever, L., and Drummond-Barbosa, D. (2005). Direct control of germline stem cell division and cyst growth by neural insulin in Drosophila. Science 309, 1071–1073.
Direct control of germline stem cell division and cyst growth by neural insulin in Drosophila.Crossref | GoogleScholarGoogle Scholar | 16099985PubMed |

Lee, S. E., Sun, S. C., Choi, H. Y., Uhm, S. J., and Kim, N. H. (2012). mTOR is required for asymmetric division through small GTPases in mouse oocytes. Mol. Reprod. Dev. 79, 356–366.
mTOR is required for asymmetric division through small GTPases in mouse oocytes.Crossref | GoogleScholarGoogle Scholar | 22407942PubMed |

Lehmann, R. (2012). Germline stem cells: origin and destiny. Cell Stem Cell 10, 729–739.
Germline stem cells: origin and destiny.Crossref | GoogleScholarGoogle Scholar | 22704513PubMed |

Li, J., Kawamura, K., Cheng, Y., Liu, S., Klein, C., Liu, S., Duan, E. K., and Hsueh, A. J. (2010). Activation of dormant ovarian follicles to generate mature eggs. Proc. Natl Acad. Sci. USA 107, 10 280–10 284.
Activation of dormant ovarian follicles to generate mature eggs.Crossref | GoogleScholarGoogle Scholar |

Lin, S. J., and Guarente, L. (2003). Nicotinamide adenine dinucleotide, a metabolic regulator of transcription, longevity and disease. Curr. Opin. Cell Biol. 15, 241–246.
Nicotinamide adenine dinucleotide, a metabolic regulator of transcription, longevity and disease.Crossref | GoogleScholarGoogle Scholar | 12648681PubMed |

Liu, M., Yin, Y., Ye, X., Zeng, M., Zhao, Q., Keefe, D. L., and Liu, L. (2013). Resveratrol protects against age-associated infertility in mice. Hum. Reprod. 28, 707–717.
Resveratrol protects against age-associated infertility in mice.Crossref | GoogleScholarGoogle Scholar | 23293221PubMed |

Mair, W., McLeod, C. J., Wang, L., and Jones, D. L. (2010). Dietary restriction enhances germline stem cell maintenance. Aging Cell 9, 916–918.
Dietary restriction enhances germline stem cell maintenance.Crossref | GoogleScholarGoogle Scholar | 20569233PubMed |

Munné, S., and Cohen, J. (1998). Chromosome abnormalities in human embryos. Hum. Reprod. Update 4, 842–855.
Chromosome abnormalities in human embryos.Crossref | GoogleScholarGoogle Scholar | 10098475PubMed |

Nakamura, S., Kobayashi, K., Nishimura, T., Higashijima, S., and Tanaka, M. (2010). Identification of germline stem cells in the ovary of the teleost medaka. Science 328, 1561–1563.
Identification of germline stem cells in the ovary of the teleost medaka.Crossref | GoogleScholarGoogle Scholar | 20488987PubMed |

Nelson, J. F., Gosden, R. G., and Felicio, L. S. (1985). Effect of dietary restriction on estrous cyclicity and follicular reserves in aging C57BL/6J mice. Biol. Reprod. 32, 515–522.
Effect of dietary restriction on estrous cyclicity and follicular reserves in aging C57BL/6J mice.Crossref | GoogleScholarGoogle Scholar | 4039610PubMed |

Niikura, Y., Niikura, T., and Tilly, J. L. (2009). Aged mouse ovaries possess rare premeiotic germ cells that can generate oocytes following transplantation into a young host environment. Aging (Albany, NY) 1, 971–978.
| 20157580PubMed |

Oh, J., Lee, Y. D., and Wagers, A. J. (2014). Stem cell aging: mechanisms, regulators and therapeutic opportunities. Nat. Med. 20, 870–880.
Stem cell aging: mechanisms, regulators and therapeutic opportunities.Crossref | GoogleScholarGoogle Scholar | 25100532PubMed |

Pacchiarotti, J., Maki, C., Ramos, T., Marh, J., Howerton, K., Wong, J., Pham, J., Anorve, S., Chow, Y. C., and Izadyar, F. (2010). Differentiation potential of germ line stem cells derived from the postnatal mouse ovary. Differentiation 79, 159–170.
Differentiation potential of germ line stem cells derived from the postnatal mouse ovary.Crossref | GoogleScholarGoogle Scholar | 20138422PubMed |

Pepling, M. E. (2006). From primordial germ cell to primordial follicle: mammalian female germ cell development. Genesis 44, 622–632.
From primordial germ cell to primordial follicle: mammalian female germ cell development.Crossref | GoogleScholarGoogle Scholar | 17146778PubMed |

Pérez-López, F. R., Chedraui, P., Gilbert, J. J., and Pérez-Roncero, G. (2009). Cardiovascular risk in menopausal women and prevalent related co-morbid conditions: facing the post-Women’s Health Initiative era. Fertil. Steril. 92, 1171–1186.
Cardiovascular risk in menopausal women and prevalent related co-morbid conditions: facing the post-Women’s Health Initiative era.Crossref | GoogleScholarGoogle Scholar | 19700149PubMed |

Reddy, P., Liu, L., Adhikari, D., Jagarlamudi, K., Rajareddy, S., Shen, Y., Du, C., Tang, W., Hamalainen, T., Peng, S. L., Lan, Z. J., Cooney, A. J., Huhtaniemi, I., and Liu, K. (2008). Oocyte-specific deletion of Pten causes premature activation of the primordial follicle pool. Science 319, 611–613.
Oocyte-specific deletion of Pten causes premature activation of the primordial follicle pool.Crossref | GoogleScholarGoogle Scholar | 18239123PubMed |

Richardson, S. J., Senikas, V., and Nelson, J. F. (1987). Follicular depletion during the menopausal transition: evidence for accelerated loss and ultimate exhaustion. J. Clin. Endocrinol. Metab. 65, 1231–1237.
Follicular depletion during the menopausal transition: evidence for accelerated loss and ultimate exhaustion.Crossref | GoogleScholarGoogle Scholar | 3119654PubMed |

Selesniemi, K., Lee, H. J., and Tilly, J. L. (2008). Moderate caloric restriction initiated in rodents during adulthood sustains function of the female reproductive axis into advanced chronological age. Aging Cell 7, 622–629.
Moderate caloric restriction initiated in rodents during adulthood sustains function of the female reproductive axis into advanced chronological age.Crossref | GoogleScholarGoogle Scholar | 18549458PubMed |

Selesniemi, K., Lee, H. J., Niikura, T., and Tilly, J. L. (2009). Young adult donor bone marrow infusions into female mice postpone age-related reproductive failure and improve offspring survival. Aging (Albany, NY) 1, 49–57.

Selesniemi, K., Lee, H. J., Muhlhauser, A., and Tilly, J. L. (2011). Prevention of maternal aging-associated oocyte aneuploidy and meiotic spindle defects in mice by dietary and genetic strategies. Proc. Natl Acad. Sci. USA 108, 12 319–12 324.
Prevention of maternal aging-associated oocyte aneuploidy and meiotic spindle defects in mice by dietary and genetic strategies.Crossref | GoogleScholarGoogle Scholar |

Shanley, D. P., and Kirkwood, T. B. (2000). Calorie restriction and aging: a life-history analysis. Evolution 54, 740–750.
Calorie restriction and aging: a life-history analysis.Crossref | GoogleScholarGoogle Scholar | 10937249PubMed |

Sohal, R. S., and Weindruch, R. (1996). Oxidative stress, caloric restriction, and aging. Science 273, 59–63.
Oxidative stress, caloric restriction, and aging.Crossref | GoogleScholarGoogle Scholar | 8658196PubMed |

te Velde, E. R., and Pearson, P. L. (2002). The variability of female reproductive ageing. Hum. Reprod. Update 8, 141–154.
The variability of female reproductive ageing.Crossref | GoogleScholarGoogle Scholar | 12099629PubMed |

The American College of Obstetricians and Gynecologists Committee on Gynecologic Practice and The Practice Committee of the American Society for Reproductive Medicine (2014). Female age-related fertility decline. Committee opinion no. 589. Fertil. Steril. 101, 633–634.
Female age-related fertility decline. Committee opinion no. 589.Crossref | GoogleScholarGoogle Scholar | 24559617PubMed |

Tilly, J. L., and Sinclair, D. A. (2013). Germline energetics, aging, and female infertility. Cell Metab. 17, 838–850.
Germline energetics, aging, and female infertility.Crossref | GoogleScholarGoogle Scholar | 23747243PubMed |

Titus, S., Li, F., Stobezki, R., Akula, K., Unsal, E., Jeong, K., Dickler, M., Robson, M., Moy, F., Goswami, S., and Oktay, K. (2013). Impairment of BRCA1-related DNA double-strand break repair leads to ovarian aging in mice and humans. Sci. Transl. Med. 5, 172ra21.
Impairment of BRCA1-related DNA double-strand break repair leads to ovarian aging in mice and humans.Crossref | GoogleScholarGoogle Scholar | 23408054PubMed |

Valli, H., Phillips, B. T., Shetty, G., Byrne, J. A., Clark, A. T., Meistrich, M. L., and Orwig, K. E. (2014). Germline stem cells: toward the regeneration of spermatogenesis. Fertil. Steril. 101, 3–13.
Germline stem cells: toward the regeneration of spermatogenesis.Crossref | GoogleScholarGoogle Scholar | 24314923PubMed |

Wang, N., and Tilly, J. L. (2010). Epigenetic status determines germ cell meiotic commitment in embryonic and postnatal mammalian gonads. Cell Cycle 9, 339–349.
Epigenetic status determines germ cell meiotic commitment in embryonic and postnatal mammalian gonads.Crossref | GoogleScholarGoogle Scholar | 20009537PubMed |

White, Y. A., Woods, D. C., Takai, Y., Ishihara, O., Seki, H., and Tilly, J. L. (2012). Oocyte formation by mitotically active germ cells purified from ovaries of reproductive-age women. Nat. Med. 18, 413–421.
Oocyte formation by mitotically active germ cells purified from ovaries of reproductive-age women.Crossref | GoogleScholarGoogle Scholar | 22366948PubMed |

Wood, J. G., Rogina, B., Lavu, S., Howitz, K., Helfand, S. L., Tatar, M., and Sinclair, D. (2004). Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Nature 430, 686–689.
Sirtuin activators mimic caloric restriction and delay ageing in metazoans.Crossref | GoogleScholarGoogle Scholar | 15254550PubMed |

Woods, D. C., and Tilly, J. L. (2013). Isolation, characterization and propagation of mitotically active germ cells from adult mouse and human ovaries. Nat. Protoc. 8, 966–988.
Isolation, characterization and propagation of mitotically active germ cells from adult mouse and human ovaries.Crossref | GoogleScholarGoogle Scholar | 23598447PubMed |

Zhang, Y., Yang, Z., Yang, Y., Wang, S., Shi, L., Xie, W., Sun, K., Zou, K., Wang, L., Xiong, J., Xiang, J., and Wu, J. (2011). Production of transgenic mice by random recombination of targeted genes in female germline stem cells. J. Mol. Cell Biol. 3, 132–141.
Production of transgenic mice by random recombination of targeted genes in female germline stem cells.Crossref | GoogleScholarGoogle Scholar | 21149239PubMed |

Zhao, R., Xuan, Y., Li, X., and Xi, R. (2008). Age-related changes of germline stem cell activity, niche signaling activity and egg production in Drosophila. Aging Cell 7, 344–354.
Age-related changes of germline stem cell activity, niche signaling activity and egg production in Drosophila.Crossref | GoogleScholarGoogle Scholar | 18267001PubMed |

Zheng, W., Gorre, N., Shen, Y., Noda, T., Ogawa, W., Lundin, E., and Liu, K. (2010). Maternal phosphatidylinositol 3-kinase signalling is crucial for embryonic genome activation and preimplantation embryogenesis. EMBO Rep. 11, 890–895.
Maternal phosphatidylinositol 3-kinase signalling is crucial for embryonic genome activation and preimplantation embryogenesis.Crossref | GoogleScholarGoogle Scholar | 20930845PubMed |

Zou, K., Yuan, Z., Yang, Z., Luo, H., Sun, K., Zhou, L., Xiang, J., Shi, L., Yu, Q., Zhang, Y., Hou, R., and Wu, J. (2009). Production of offspring from a germline stem cell line derived from neonatal ovaries. Nat. Cell Biol. 11, 631–636.
Production of offspring from a germline stem cell line derived from neonatal ovaries.Crossref | GoogleScholarGoogle Scholar | 19363485PubMed |