You are on page 1of 6

CSIRO PUBLISHING

Reproduction, Fertility and Development


Review
http://dx.doi.org/10.1071/RD14461

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


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

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.

Received 21 November 2014, accepted 14 March 2015, published online 22 April 2015

Introduction fail with advancing age and is associated with changes in


Reproductive aging is characterised by a progressive decline in ovarian function (te Velde and Pearson 2002; Selesniemi et al.
fertility and fecundity with advancing age (te Velde and Pearson 2009). The primary functional unit of the mammalian ovary is
2002). In women, fertility decreases drastically beyond 37 years the follicle, consisting of an oocyte and supporting somatic cells
of age (The American College of Obstetricians and Gynecolo- that function in the growth and development of the oocyte
gists Committee on Gynecologic Practice and The Practice (Channing et al. 1980). Each oocyte, enclosed within a primor-
Committee of the American Society for Reproductive Medicine dial follicle, is arrested in the diplotene stage of meiotic
2014) and is associated with reduced ovarian function. This prophase I (Borum 1961). The pool of primordial follicles laid
decline is believed to contribute to the development of health down at birth represents the total population of follicles existing
complications, including osteoporosis, cardiovascular disease, in the ovary throughout the reproductive lifespan of a female
cancer and urinary problems (Pérez-López et al. 2009). Com- (Kezele et al. 2002). In mammals, several of the oocytes die
bined with increasing numbers of women delaying childbirth, during development or shortly after birth, and those remaining
immense efforts are being made to diagnose and counteract age- continue to decline with age (De Felici et al. 2005). This
linked female infertility. eventual exhaustion of the ovarian follicular reserve culminates
Although it is known that age is the single most important in a complete cessation of normal ovarian function, driving
determinant of female fertility (Balasch 2010), recent work has menopause and a decline in female physiology (Richardson
shown that the decline in fertility with age is far more malleable et al. 1987).
than originally thought. Studies in lower organisms such as A central dogma in the field of reproductive biology is that
Drosophila have demonstrated that, with age, there is a gradual ovaries of mammalian females are endowed with a finite non-
decline in the rate of germline stem cell division accompanied renewable pool of oocytes at the time of birth (Borum 1961).
by reduced egg production and increased incidence of cell death The foundation for this notion is the supposed absence of
of developing eggs, especially in oldest females (Zhao et al. mitotically active germ cells in postnatal mammalian ovaries
2008). In mammals, the female reproductive axis is the first to (Pepling 2006). In contrast, females of non-mammalian species,

Journal compilation Ó CSIRO 2015 www.publish.csiro.au/journals/rfd


B Reproduction, Fertility and Development N. Garg and D. A. Sinclair

including flies and fish, retain germline stem cells and continue (Prdm1), developmental pluripotency associated 3 (Dppa3),
producing new oocytes well into adult life (Kirilly and Xie 2007; interferon-induced transmembrane protein 3 (Ifitm3), telomerase
Nakamura et al. 2010). Germline stem cells are a unique stem reverse transcriptase (Tert), Ddx4 and deleted in azoospermia-
cell population involved in reproduction and transmitting genetic like gene family (Dazl), possess pluripotency markers octamer-
information (Lehmann 2012). In males, spermatogenesis con- binding transcription factor 4 (Oct4) and stage-specific mouse
tinues throughout adult life due to the presence of spermatogonial embryonic antigen (Ssea1) and have a moderate level of
stem cells (Valli et al. 2014). However, recent studies may have telomerase activity (White et al. 2012; Woods and Tilly
disproved the theory of fixed ovarian reserve in adult females by 2013). The OSCs maintain their germline profile even when
providing evidence for the presence of germline stem cells in the cultured for a long time in vitro. In culture, a subset of these
postnatal mammalian ovary that are capable of undergoing OSCs undergoes spontaneous meiosis to generate oocyte-like
differentiation into oocytes both in vitro and in vivo (Johnson cells that express classic oocyte markers, including Kit, newborn
et al. 2004; White et al. 2012). Nonetheless, this field is still in its ovary homeobox (Nobox), Y-box-binding protein 2 (Ybx2), LIM
infancy and lacks a complete understanding of how these putative homeobox 8 (Lhx8) and growth differentiation factor 9 (Gdf9),
germline stem cells and fertility respond during aging. Given that as well as zona pellucida glycoprotein (Zp)1, Zp2 and Zp3
oocyte numbers decline with age, it is possible that menopause in (White et al. 2012). In transplantation studies, mouse OSCs
females is not a result of depletion of the ovarian reserve, but with stable green fluorescent protein (GFP) expression have
rather a result of aging of germline stem cells (Dunlop et al. been shown to differentiate into follicle-enclosed GFP-positive
2014). In this review we discuss the connection between stem oocytes that, upon ovulation, are released as mature MII eggs.
cells, fertility and aging. These eggs are fertilisation competent and can produce viable
offspring (Zou et al. 2009; White et al. 2012). Like mouse OSCs,
human GFP-tagged OSCs also undergo neo-oogenesis, produc-
Postnatal oogenesis, germline stem cells and fertility
ing large, ovoid, GFP-positive oocytes in adult human ovarian
In mammals, oogenesis is believed to be primarily prenatal. cortical tissue in vivo (White et al. 2012).
During development in females, precursor germ cells known as
primordial germ cells arise extra-embryonically and migrate to
their final destination in the gonads, where they enter meiosis Germline stem cell aging
and differentiate into oocytes, thereby ending their stem cell Aging is characterised by a progressive decline of tissue and
potential (Bowles and Koopman 2007). The possibility of organ function. The human body is composed of a variety of
postnatal oogenesis in the mammalian ovary was a topic of adult stem cells that are capable of replenishing somatic cells
debate for a long time (Greenfeld and Flaws 2004). In 2004, and tissues as they get damaged, diseased or die to maintain
multiple lines of evidence were provided, for the first time, for tissue homeostasis (Jones and Rando 2011). However, during
the existence of germline stem cells in adult mouse ovaries aging, stem cells lose their regenerative and proliferative
capable of generating oocytes to form new follicles (Johnson potential and this loss of stem cell function over time is con-
et al. 2004). A small population of mitotically active germ cells sidered a major cause of age-related organ deterioration and
was detected in the surface epithelium of adult ovaries, which, disease (Jones and Rando 2011; Oh et al. 2014). To date, the
upon isolation, differentiated to produce oocyte-like cells in mechanisms of such stem cell aging are poorly understood.
culture (Johnson et al. 2004). By calculating the number of Comprehending the molecular pathways that regulate stem cell
follicles undergoing atresia, it was concluded that postnatal survival, self-renewal, proliferation and commitment to specific
production of follicles to the existing ovarian reserve was nec- differentiated cell lineages and age-dependent stem cell quies-
essary to reach the expected reproductive lifespan in mice cence is critical in determining age-associated stem cell dys-
(Johnson et al. 2004). Thus, the demonstration of the existence function. Furthermore, such knowledge will be essential for the
of putative postnatal germline stem cells changed the traditional development of therapeutic interventions that can delay age-
thinking that mammalian oogenesis is prenatal. related degenerative processes to enhance repair and maintain
Following the work of Johnson et al. (2004), several inde- healthy function in aging tissues.
pendent laboratories demonstrated evidence for the existence of In the context of regenerative medicine and treating age-
postnatal oogenesis, each having isolated germline stem cells related diseases, adult somatic stem cells and embryonic stem
from neonatal and adult mouse ovaries using different isolation cells have attracted much attention. However, germline stem
techniques (Zou et al. 2009; Pacchiarotti et al. 2010; Zhang et al. cells have been largely ignored. This was mainly because these
2011; White et al. 2012). The study by White et al. (2012) cells were not even known to exist. Not only have these cells
further went on to isolate these germline stem cells, henceforth now been identified, but they also populate atrophic ovaries
referred to as oogonial stem cells (OSCs), from adult human of reproductively aged mice despite complete depletion of
ovaries. The OSCs were isolated using a ATP-dependent oocytes (Niikura et al. 2009). This suggests that premeiotic
DEAD-box RNA helicase (DDX4) antibody-based immuno- germ cells or OSCs, although present in ovaries of aged mice,
magnetic sort or fluorescence-activated cell sorting (FACS; Zou exist in a dormant state and are apparently unable to proliferate
et al. 2009; White et al. 2012). These OSCs constitute a very and differentiate into oocytes during aging. When transplanted
small subset, approximately 0.01%, of the total ovarian cell into a young host, these germ cells resume differentiation to
population. Freshly isolated mouse and human OSCs express form oocytes, suggesting that ovarian aging may be reversible
germline markers, namely PR domain zinc finger protein 1 (Niikura et al. 2009). Thus, from a reproductive aging
Oogonial stem cells and aging Reproduction, Fertility and Development C

perspective, the main question is what causes the debility in mechanism by which CR may delay aging in lower organisms
transition of OSCs into oocytes. (Mair et al. 2010).
Evidence for ongoing oogenesis in adult postnatal ovaries In mice, restricting calories has been shown to prolong
comes from reports demonstrating the presence of the germ cell lifespan by almost 50% and retard the onset of age-related
specific gene stimulated by retinoic acid gene 8 (Stra8), a diseases (Sohal and Weindruch 1996). Although female mice
regulator of meiotic initiation and progression in both spermato- experience reduced fecundity while on the CR diet, age-associ-
genesis and oogenesis (Niikura et al. 2009). Stra8 is required for ated loss of oestrous cyclicity and follicular reserve are delayed
premeiotic DNA replication and progression into meiotic pro- and, upon resumption of ad libitum diet, mice remain fertile for
phase (Baltus et al. 2006; Anderson et al. 2008). Stra8/ germ an extended period of time (Nelson et al. 1985). In aging
cells do not undergo premeiotic DNA replication and do not rodents, CR has been shown to extend fertility (Selesniemi
enter meiotic prophase (Baltus et al. 2006). Induction of Stra8, et al. 2008). An adult onset of CR for a brief period of time
in both male and female germ cells, occurs in response to diminished age-related chromosomal abnormalities and mito-
retinoic acid (RA) signalling in the somatic cells (Anderson chondrial dysfunction in oocytes and improved postnatal sur-
et al. 2008). An important step in the regulation of mammalian vival of offspring delivered by aged female mice (Selesniemi
oogenesis is the sex-specific timing of RA-mediated Stra8 et al. 2008; Selesniemi et al. 2011). Although CR is an effective
transcription. Recently, it was shown that, in adult mammalian way to delay ovarian aging, little is known about the underlying
ovaries, RA-induced Stra8 expression is epigenetically regulat- molecular mechanisms by which CR delays aging and repro-
ed at the RA response elements (RARE) region of the Stra8 ductive senescence.
promoter (Wang and Tilly 2010). Based on the pivotal role of Of the several pathways mediating CR, the nutrient sensing
Stra8 in male spermatogenesis and some initial evidence of pathways of insulin/insulin-like growth factor-1 (IGF-1),
expression in the adult ovary (Niikura et al. 2009), Stra8 is a mammalian target of rapamycin (mTOR), sirtuins and AMP-
good marker for postnatal oogenesis and should be investigated activated kinase (AMPK) are well established (Tilly and
in OSCs. In addition, genes for meiosis, including Stra8, serve as Sinclair 2013). The metabolic and energy sensors sirtuin 1
good indicators of neo-oogenesis in postnatal mammalian (SIRT1) and AMPK operate as a synchronised network in
ovaries. It is noteworthy to mention that only a fraction of the response to environmental signals and interact with insulin/
OSCs in culture undergo meiosis to form oocytes (Woods and IGF-1 and mTOR to regulate cellular metabolism, growth and
Tilly 2013). Why only a few cells express Stra8 and undergo energy intake. The phosphatase and tensin homologue (PTEN)/
differentiation remains unknown. Thus, the identification of a phosphatidylinositol 3-kinase (PI3K) pathway has been
molecular clock regulating differentiation in OSCs is awaited. shown to be functional in regulating oocyte growth (Reddy
et al. 2008; Li et al. 2010) and the mTOR/PI3K/PTEN pathway
plays a role in meiotic progression and embryonic genome
Delaying reproductive aging
activation in embryos (Zheng et al. 2010; Lee et al. 2012).
According to the ‘disposable soma theory’ of aging, more Recently, a regulatory role for hypoxia-mediated IGF-I receptor
energy is invested by animals into reproduction than for the (IGF-IR)–PI3K/Akt–mTOR–hypoxia inducible factor (HIF)-2a
maintenance of the soma. This results in fewer resources signalling was demonstrated to mediate proliferation and main-
available for protecting the soma for indefinite survival, thereby tenance of pluripotency in mouse germline stem cells (Huang
driving aging (Kirkwood and Holliday 1979). Conversely, cal- et al. 2014).
orie restriction (CR), a dietary regimen that involves con- Some of the beneficial effects of CR are mediated in a SIRT1,
sumption of a diet approximately 30%–40% reduced in calories an NADþ dependent deacetylase and nutrient sensing protein,
compared with ad libitum diet, redirects energy required for dependent manner (Guarente and Picard 2005; Guarente 2013).
reproduction towards somatic maintenance, thus retarding aging Alternatively, increasing SIRT1 activity or raising intracellular
at the cost of reduced fertility. This temporary hiatus from fer- levels of NADþ has also been shown to mimic CR-like physio-
tility permits an individual to survive and preserve fertility for logy and protection from several age-associated diseases in mice
when resources become available (Shanley and Kirkwood (Howitz et al. 2003; Gomes et al. 2013). Moreover, increased
2000). SIRT1 activity, as depicted by use of small molecule activators
Of the several interventions studied to prevent aging, CR by of SIRT1, such as resveratrol, has been shown to increase the
far stands out to be the most promising in lifespan extension number of eggs laid per organism in C. elegans and Drosophila
(Holliday 1989) in diverse species and protects against age- (Wood et al. 2004) and both the number and quality of oocytes in
related pathologies. In lower organisms such as Drosophila and aged mice compared with age-matched control mice (Liu et al.
Caenorhabditis elegans, nutritional status determines the fate of 2013). Considering OSCs accumulate in the ovaries of aged
germline stem cells (LaFever and Drummond-Barbosa 2005; mice with an apparent block in differentiation, it is reasonable to
Angelo and Van Gilst 2009; Hsu and Drummond-Barbosa postulate that raising NADþ levels of these cells, thereby
2009). Most of the germline degenerates when resources are activating SIRT1, will promote differentiation. Thus, com-
scarce. However, a small pool of germline stem cells is main- pounds that raise NADþ levels may serve as molecules with
tained that retains its ability to repopulate and reproduce when the potential to delay ovarian aging and extend female repro-
normal feeding resumes (Angelo and Van Gilst 2009; Mair et al. ductive lifespan. Whether this occurs through SIRT1-mediated
2010). Thus, the maintenance of a germline stem cell pool regulation of Stra8 is yet to be investigated. Identification of
during CR to extend reproductive period is a potential the pathways by which SIRT1 regulates the transition of OSCs
D Reproduction, Fertility and Development N. Garg and D. A. Sinclair

into oocytes will help open new dimensions in the understanding mammalian oocytes (Titus et al. 2013; Govindaraj et al. 2015).
of reproductive aging. Elucidating the mechanisms that cause OSCs to age could lead
to new treatments that could delay ovarian aging and slow
Mitochondria and energetics infertility.
It is now well recognised that CR results in extended lifespan
The oocyte has the largest number of mitochondria and mito-
and rebound fertility. However, the role of SIRT1, as a mediator
chondrial (mt) DNA per cell necessary for maintaining energy
of the CR response, has yet to be linked to the longevity of germ
requirements during proper oocyte maturation, spindle forma-
cells. The cell autonomous function of SIRT1 in germline stem
tion, fertilisation and embryo development (Bentov et al. 2011;
cells and reproductive aging, if any, is yet to be determined. One
Tilly and Sinclair 2013). Mitochondrial function in oocytes
of the theories of reproductive aging that we think can explain
declines with age and, consistent with this, oocytes from older
the role of SIRT1 in germ cells, or more so in the germline stem
women have decreased ATP levels, low mtDNA copy number
cells, links to its epigenetic regulation of Stra8 expression. The
and intracellular clusters of aggregated mitochondria that are
OSCs are natural precursor cells for oocytes and thus any
increased in size and density (Bentov et al. 2011; Selesniemi
manipulation or testing done in these cells should, theoretically,
et al. 2011; Tilly and Sinclair 2013). An energy deficit in the
have a similar effect on oocytes. Moreover, unlike oocytes,
aging oocyte also disables the formation of the meiotic spindle, a
OSCs can be propagated and cultured in vitro for long periods of
high energy-consuming process, and results in chromosomal
time and in seemingly infinite numbers. Thus, they can be
misalignment and aneuploidy, which are closely linked to tri-
exploited as a platform to screen for compounds that, by raising
somic conceptions, implantation failures and miscarriages
NADþ levels, may potentially rescue the OSCs from their
(Hassold and Chiu 1985; Munné and Cohen 1998; Tilly and
dormant state and facilitate the production of more oocytes
Sinclair 2013). Because mitochondria are solely inherited from
(Tilly and Sinclair 2013).
the mother, the embryo also inherits a dysfunctional energy
The field of female germ cell biology is still in its infancy,
system, which then poses a threat to embryonic development
and many questions remain. If human OSCs can be proven to
(Tilly and Sinclair 2013). Conversely, increasing mitochondrial
generate fertilisable eggs and eventually offspring, they hold
function and energy production have a positive impact on the
considerable promise in counteracting the effects of chemother-
overall fertility outcome in women of advanced maternal age.
apeutic drugs and aging on fertility. The OSCs may allow for
Being natural oocyte progenitors, OSCs in culture can be used to
germline correction of inherited dominant human diseases and,
identify compounds that, by raising energy levels, facilitate the
eventually, it may be possible to differentiate OSCs ex vivo
formation and maintenance of meiotic spindles, thereby reduc-
using tissue explants or cell coculture, allowing them to be
ing the occurrence of oocyte aneuploidy. One such method of
fertilised. In this way, dozens of healthy oocytes could be
increasing energy levels is by raising NADþ, which has emerged
produced from an infertile woman and screened before implan-
as a potent mitochondrial booster (Lin and Guarente 2003;
tation, thereby revolutionising human fertility. There may be
Gomes et al. 2013). Thus, it is reasonable to believe that using
other applications as well that, at present, we can only see, such
mitochondrial activators as in vivo agents would benefit women
as tissue regeneration and organ replacement.
with energetically compromised eggs or embryos, especially
those undergoing IVF.
References
Perspective Anderson, E. L., Baltus, A. E., Roepers-Gajadien, H. L., Hassold, T. J.,
In the past few years a lot of interest has been generated in the de Rooij, D. G., van Pelt, A. M., and Page, D. C. (2008). Stra8 and
field of stem cell aging. Aging seems to be a consequence of its inducer, retinoic acid, regulate meiotic initiation in both spermato-
diminished stem cell function or a decline in the function genesis and oogenesis in mice. Proc. Natl Acad. Sci. USA 105, 14 976–
of other self-renewing cells, and understanding how age- 14 980. doi:10.1073/PNAS.0807297105
Angelo, G., and Van Gilst, M. R. (2009). Starvation protects germline stem
dependent changes in stem cell function contribute to human
cells and extends reproductive longevity in C. elegans. Science 326,
aging may aid in the development of new anti-aging therapies. 954–958. doi:10.1126/SCIENCE.1178343
An outstanding example of cellular aging, separate from the Balasch, J. (2010). Ageing and infertility: an overview. Gynecol. Endocri-
aging of the rest of the organism, is the aging of germ cells nol. 26, 855–860. doi:10.3109/09513590.2010.501889
within the ovaries of postmenopausal women (Niikura et al. Baltus, A. E., Menke, D. B., Hu, Y. C., Goodheart, M. L., Carpenter, A. E.,
2009). Women struggling with infertility, due to either advanced de Rooij, D. G., and Page, D. C. (2006). In germ cells of mouse
age or ovarian insufficiency, have little option but to rely on egg embryonic ovaries, the decision to enter meiosis precedes premeiotic
donation to achieve a successful pregnancy. However, in such a DNA replication. Nat. Genet. 38, 1430–1434. doi:10.1038/NG1919
case the child is not their biological child (Tilly and Sinclair Bentov, Y., Yavorska, T., Esfandiari, N., Jurisicova, A., and Casper, R. F.
2013). With the recent discovery of postnatal oogenesis and (2011). The contribution of mitochondrial function to reproductive
aging. J. Assist. Reprod. Genet. 28, 773–783. doi:10.1007/S10815-
mitotically active germ cells or OSCs, there may be a possibility
011-9588-7
of coaxing these cells into forming healthy oocytes in women of Borum, K. (1961). Oogenesis in the mouse. A study of the meiotic prophase.
advanced age. However, the reason OSCs fail to produce Exp. Cell Res. 24, 495–507. doi:10.1016/0014-4827(61)90449-9
functional oocytes in older women is not known. Alternatively, Bowles, J., and Koopman, P. (2007). Retinoic acid, meiosis and germ cell
it is possible that OSC aging is a result of impaired DNA double- fate in mammals. Development 134, 3401–3411. doi:10.1242/DEV.
strand break repair, a recently identified cause of aging in 001107
Oogonial stem cells and aging Reproduction, Fertility and Development E

Channing, C. P., Schaerf, F. W., Anderson, L. D., and Tsafriri, A. (1980). Lee, S. E., Sun, S. C., Choi, H. Y., Uhm, S. J., and Kim, N. H. (2012). mTOR
Ovarian follicular and luteal physiology. Int. Rev. Physiol. 22, 117–201. is required for asymmetric division through small GTPases in mouse
De Felici, M., Klinger, F. G., Farini, D., Scaldaferri, M. L., Iona, S., and oocytes. Mol. Reprod. Dev. 79, 356–366. doi:10.1002/MRD.22035
Lobascio, M. (2005). Establishment of oocyte population in the fetal Lehmann, R. (2012). Germline stem cells: origin and destiny. Cell Stem Cell
ovary: primordial germ cell proliferation and oocyte programmed cell 10, 729–739. doi:10.1016/J.STEM.2012.05.016
death. Reprod. Biomed. Online 10, 182–191. doi:10.1016/S1472-6483 Li, J., Kawamura, K., Cheng, Y., Liu, S., Klein, C., Liu, S., Duan, E. K., and
(10)60939-X Hsueh, A. J. (2010). Activation of dormant ovarian follicles to generate
Dunlop, C. E., Telfer, E. E., and Anderson, R. A. (2014). Ovarian germline mature eggs. Proc. Natl Acad. Sci. USA 107, 10 280–10 284.
stem cells. Stem Cell Res. Ther. 5, 98. doi:10.1186/SCRT487 doi:10.1073/PNAS.1001198107
Gomes, A. P., Price, N. L., Ling, A. J., Moslehi, J. J., Montgomery, M. K., Lin, S. J., and Guarente, L. (2003). Nicotinamide adenine dinucleotide, a
Rajman, L., White, J. P., Teodoro, J. S., Wrann, C. D., Hubbard, B. P., metabolic regulator of transcription, longevity and disease. Curr. Opin.
Mercken, E. M., Palmeira, C. M., de Cabo, R., Rolo, A. P., Turner, N., Cell Biol. 15, 241–246. doi:10.1016/S0955-0674(03)00006-1
Bell, E. L., and Sinclair, D. A. (2013). Declining NAD(þ) induces a Liu, M., Yin, Y., Ye, X., Zeng, M., Zhao, Q., Keefe, D. L., and Liu, L. (2013).
pseudohypoxic state disrupting nuclear–mitochondrial communication Resveratrol protects against age-associated infertility in mice. Hum.
during aging. Cell 155, 1624–1638. doi:10.1016/J.CELL.2013.11.037 Reprod. 28, 707–717. doi:10.1093/HUMREP/DES437
Govindaraj, V., Keralapura Basavaraju, R., and Rao, A. J. (2015). Changes Mair, W., McLeod, C. J., Wang, L., and Jones, D. L. (2010). Dietary
in the expression of DNA double strand break repair genes in primordial restriction enhances germline stem cell maintenance. Aging Cell 9,
follicles from immature and aged rats. Reprod. Biomed. Online 30, 916–918. doi:10.1111/J.1474-9726.2010.00602.X
303–310. doi:10.1016/J.RBMO.2014.11.010 Munné, S., and Cohen, J. (1998). Chromosome abnormalities in human
Greenfeld, C., and Flaws, J. A. (2004). Renewed debate over postnatal embryos. Hum. Reprod. Update 4, 842–855. doi:10.1093/HUMUPD/
oogenesis in the mammalian ovary. Bioessays 26, 829–832. doi:10.1002/ 4.6.842
BIES.20094 Nakamura, S., Kobayashi, K., Nishimura, T., Higashijima, S., and Tanaka,
Guarente, L. (2013). Calorie restriction and sirtuins revisited. Genes Dev. 27, M. (2010). Identification of germline stem cells in the ovary of the teleost
2072–2085. doi:10.1101/GAD.227439.113 medaka. Science 328, 1561–1563. doi:10.1126/SCIENCE.1185473
Guarente, L., and Picard, F. (2005). Calorie restriction: the SIR2 connection. Nelson, J. F., Gosden, R. G., and Felicio, L. S. (1985). Effect of dietary
Cell 120, 473–482. doi:10.1016/J.CELL.2005.01.029 restriction on estrous cyclicity and follicular reserves in aging C57BL/6J
Hassold, T., and Chiu, D. (1985). Maternal age-specific rates of numerical mice. Biol. Reprod. 32, 515–522. doi:10.1095/BIOLREPROD32.3.515
chromosome abnormalities with special reference to trisomy. Hum. Niikura, Y., Niikura, T., and Tilly, J. L. (2009). Aged mouse ovaries
Genet. 70, 11–17. doi:10.1007/BF00389450 possess rare premeiotic germ cells that can generate oocytes following
Holliday, R. (1989). Food, reproduction and longevity: is the extended transplantation into a young host environment. Aging (Albany, NY) 1,
lifespan of calorie-restricted animals an evolutionary adaptation? Bioes- 971–978.
says 10, 125–127. doi:10.1002/BIES.950100408 Oh, J., Lee, Y. D., and Wagers, A. J. (2014). Stem cell aging: mechanisms,
Howitz, K. T., Bitterman, K. J., Cohen, H. Y., Lamming, D. W., Lavu, S., regulators and therapeutic opportunities. Nat. Med. 20, 870–880.
Wood, J. G., Zipkin, R. E., Chung, P., Kisielewski, A., Zhang, L. L., doi:10.1038/NM.3651
Scherer, B., and Sinclair, D. A. (2003). Small molecule activators Pacchiarotti, J., Maki, C., Ramos, T., Marh, J., Howerton, K., Wong, J.,
of sirtuins extend Saccharomyces cerevisiae lifespan. Nature 425, Pham, J., Anorve, S., Chow, Y. C., and Izadyar, F. (2010). Differentia-
191–196. doi:10.1038/NATURE01960 tion potential of germ line stem cells derived from the postnatal mouse
Hsu, H. J., and Drummond-Barbosa, D. (2009). Insulin levels control female ovary. Differentiation 79, 159–170. doi:10.1016/J.DIFF.2010.01.001
germline stem cell maintenance via the niche in Drosophila. Proc. Natl Pepling, M. E. (2006). From primordial germ cell to primordial follicle:
Acad. Sci. USA 106, 1117–1121. doi:10.1073/PNAS.0809144106 mammalian female germ cell development. Genesis 44, 622–632.
Huang, Y. H., Lin, M. H., Wang, P. C., Wu, Y. C., Chiang, H. L., Wang, doi:10.1002/DVG.20258
Y. L., Chang, J. H., Huang, Y. K., Gu, S. Y., Ho, H. N., and Ling, T. Y. Pérez-López, F. R., Chedraui, P., Gilbert, J. J., and Pérez-Roncero, G.
(2014). Hypoxia inducible factor 2alpha/insulin-like growth factor (2009). Cardiovascular risk in menopausal women and prevalent related
receptor signal loop supports the proliferation and Oct-4 maintenance co-morbid conditions: facing the post-Women’s Health Initiative era.
of mouse germline stem cells. Mol. Hum. Reprod. 20, 526–537. Fertil. Steril. 92, 1171–1186. doi:10.1016/J.FERTNSTERT.2009.
doi:10.1093/MOLEHR/GAU016 06.032
Johnson, J., Canning, J., Kaneko, T., Pru, J. K., and Tilly, J. L. (2004). Reddy, P., Liu, L., Adhikari, D., Jagarlamudi, K., Rajareddy, S., Shen, Y.,
Germline stem cells and follicular renewal in the postnatal mammalian Du, C., Tang, W., Hamalainen, T., Peng, S. L., Lan, Z. J., Cooney, A. J.,
ovary. Nature 428, 145–150. doi:10.1038/NATURE02316 Huhtaniemi, I., and Liu, K. (2008). Oocyte-specific deletion of Pten
Jones, D. L., and Rando, T. A. (2011). Emerging models and paradigms causes premature activation of the primordial follicle pool. Science 319,
for stem cell ageing. Nat. Cell Biol. 13, 506–512. doi:10.1038/ 611–613. doi:10.1126/SCIENCE.1152257
NCB0511-506 Richardson, S. J., Senikas, V., and Nelson, J. F. (1987). Follicular depletion
Kezele, P., Nilsson, E., and Skinner, M. K. (2002). Cell–cell interactions during the menopausal transition: evidence for accelerated loss and
in primordial follicle assembly and development. Front. Biosci. 7, ultimate exhaustion. J. Clin. Endocrinol. Metab. 65, 1231–1237.
d1990–d1996. doi:10.2741/KEZELE doi:10.1210/JCEM-65-6-1231
Kirilly, D., and Xie, T. (2007). The Drosophila ovary: an active stem cell Selesniemi, K., Lee, H. J., and Tilly, J. L. (2008). Moderate caloric
community. Cell Res. 17, 15–25. doi:10.1038/SJ.CR.7310123 restriction initiated in rodents during adulthood sustains function of
Kirkwood, T. B., and Holliday, R. (1979). The evolution of ageing and the female reproductive axis into advanced chronological age. Aging
longevity. Proc. R. Soc. Lond. B Biol. Sci. 205, 531–546. doi:10.1098/ Cell 7, 622–629. doi:10.1111/J.1474-9726.2008.00409.X
RSPB.1979.0083 Selesniemi, K., Lee, H. J., Niikura, T., and Tilly, J. L. (2009). Young adult
LaFever, L., and Drummond-Barbosa, D. (2005). Direct control of germline donor bone marrow infusions into female mice postpone age-related
stem cell division and cyst growth by neural insulin in Drosophila. reproductive failure and improve offspring survival. Aging (Albany, NY)
Science 309, 1071–1073. doi:10.1126/SCIENCE.1111410 1, 49–57.
F Reproduction, Fertility and Development N. Garg and D. A. Sinclair

Selesniemi, K., Lee, H. J., Muhlhauser, A., and Tilly, J. L. (2011). Prevention White, Y. A., Woods, D. C., Takai, Y., Ishihara, O., Seki, H., and Tilly, J. L.
of maternal aging-associated oocyte aneuploidy and meiotic spindle (2012). Oocyte formation by mitotically active germ cells purified
defects in mice by dietary and genetic strategies. Proc. Natl Acad. Sci. from ovaries of reproductive-age women. Nat. Med. 18, 413–421.
USA 108, 12 319–12 324. doi:10.1073/PNAS.1018793108 doi:10.1038/NM.2669
Shanley, D. P., and Kirkwood, T. B. (2000). Calorie restriction and aging: a Wood, J. G., Rogina, B., Lavu, S., Howitz, K., Helfand, S. L., Tatar, M., and
life-history analysis. Evolution 54, 740–750. doi:10.1111/J.0014-3820. Sinclair, D. (2004). Sirtuin activators mimic caloric restriction and
2000.TB00076.X delay ageing in metazoans. Nature 430, 686–689. doi:10.1038/
Sohal, R. S., and Weindruch, R. (1996). Oxidative stress, caloric restriction, NATURE02789
and aging. Science 273, 59–63. doi:10.1126/SCIENCE.273.5271.59 Woods, D. C., and Tilly, J. L. (2013). Isolation, characterization and
te Velde, E. R., and Pearson, P. L. (2002). The variability of female propagation of mitotically active germ cells from adult mouse and
reproductive ageing. Hum. Reprod. Update 8, 141–154. doi:10.1093/ human ovaries. Nat. Protoc. 8, 966–988. doi:10.1038/NPROT.2013.047
HUMUPD/8.2.141 Zhang, Y., Yang, Z., Yang, Y., Wang, S., Shi, L., Xie, W., Sun, K., Zou, K.,
The American College of Obstetricians and Gynecologists Committee on Wang, L., Xiong, J., Xiang, J., and Wu, J. (2011). Production of
Gynecologic Practice and The Practice Committee of the American transgenic mice by random recombination of targeted genes in female
Society for Reproductive Medicine (2014). Female age-related fertility germline stem cells. J. Mol. Cell Biol. 3, 132–141. doi:10.1093/JMCB/
decline. Committee opinion no. 589. Fertil. Steril. 101, 633–634. MJQ043
doi:10.1016/J.FERTNSTERT.2013.12.032 Zhao, R., Xuan, Y., Li, X., and Xi, R. (2008). Age-related changes of
Tilly, J. L., and Sinclair, D. A. (2013). Germline energetics, aging, and germline stem cell activity, niche signaling activity and egg production
female infertility. Cell Metab. 17, 838–850. doi:10.1016/J.CMET.2013. in Drosophila. Aging Cell 7, 344–354. doi:10.1111/J.1474-9726.2008.
05.007 00379.X
Titus, S., Li, F., Stobezki, R., Akula, K., Unsal, E., Jeong, K., Dickler, M., Zheng, W., Gorre, N., Shen, Y., Noda, T., Ogawa, W., Lundin, E., and Liu,
Robson, M., Moy, F., Goswami, S., and Oktay, K. (2013). Impairment of K. (2010). Maternal phosphatidylinositol 3-kinase signalling is crucial
BRCA1-related DNA double-strand break repair leads to ovarian aging for embryonic genome activation and preimplantation embryogenesis.
in mice and humans. Sci. Transl. Med. 5, 172ra21. doi:10.1126/ EMBO Rep. 11, 890–895. doi:10.1038/EMBOR.2010.144
SCITRANSLMED.3004925 Zou, K., Yuan, Z., Yang, Z., Luo, H., Sun, K., Zhou, L., Xiang, J., Shi, L.,
Valli, H., Phillips, B. T., Shetty, G., Byrne, J. A., Clark, A. T., Meistrich, Yu, Q., Zhang, Y., Hou, R., and Wu, J. (2009). Production of offspring
M. L., and Orwig, K. E. (2014). Germline stem cells: toward the from a germline stem cell line derived from neonatal ovaries. Nat. Cell
regeneration of spermatogenesis. Fertil. Steril. 101, 3–13. Biol. 11, 631–636. doi:10.1038/NCB1869
doi:10.1016/J.FERTNSTERT.2013.10.052
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. doi:10.4161/CC.9.2.10447

www.publish.csiro.au/journals/rfd

You might also like