Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

K. Reynaud, M.A.

Driancourt * INRA-URA CNRS 1291,


PRMD, 37380 Nouzilly, France

Molecular and Cellular Endocrinology 163 (2000) 101–108

www.elsevier.com/locate/mce
Oocyte attrition

Received 25 June 1999; received in revised form 6 August 1999; accepted 29 September 1999

Abstract

During oogenesis, germ cell numbers sharply decrease when meiosis is initiated. There is solid evidence (DNA ladders, in situ detection)
that this loss is through apoptosis. Oocyte apoptosis appears to hit mitotic primordial germ cells (PGC), pachytene oocytes and early
primordial follicles. The control of oocyte apoptosis is not fully understood, although survival factors (LIF, kit ligand and FGF), as well as
death inducing factors (fas ligand, TGFb), have been identified. Fas ligand binding on oocytic fas may result in caspase 8 activation. Two
pathways inducing oocyte apoptosis may then be operating. In the first one, activated caspase 8 will induce activation of executioner
caspases. In the second one, activated caspase 8 will trigger the cleavage of the bcl2 family member Bid, which will act on mitochondria,
resulting in cytochrome c release, caspase 9 activation and finally, activation of all executioner caspases. As a consequence of caspase
activation, alterations in the cell nucleus (DNAse activation, PARP fragmentation), in the cell cytoskeleton (lamin) and cell metabolism will
occur, producing cell death. During folliculogenesis, germ cell loss, owing to oocyte apoptosis, has been postulated within primordial and
preantral follicles. Its regulatory mechanisms may be even more complex than those operating in foetal oocytes since additional control
factors include EGF/TGFa and bcl2 (survival) and activin (death inducer). In contrast, oocytes from antral follicles appear to be very
unsensitive to death inducing stimuli. © 2000 Elsevier Science Ireland Ltd. All rights reserved.

Keywords: Oocyte; Apoptosis; Follicle


Driancourt).
section of this review will attempt to clarify how and why
germ cell loss occurs throughout oogenesis. Ewes and
1. Introduction women ovulate every 17 and 28 days, respectively. This
infrequent occurrence of ovulation contrasts with the
Females of all domestic species and primates have a continuous exit from the reserve of primordial follicles (2
finite store of germ cells, which is established during
and 10–30 follicles initiate growth) every day in sheep and
oogenesis and used later for folliculogenesis. Formation of
in young women, respectively, according to Driancourt et
this store (i.e. the pool of primordial follicles) gener ally
al. (1985), Faddy and Gosden (1995). This suggests that a
occurs during foetal life as a result of a number of
overlapping events, migration of the primordial germ cells very strong tuning of the number of growing follicles
(PGC) to the gonadal ridge, proliferation of these PGC, occurs throughout folliculogenesis. The respective
initiation of meiotic prophase followed by its block to the contribution of oocyte apoptosis versus apoptosis of
diplotene stage, and finally, formation of the primordial somatic cells to generate a single ovulatory follicle from a
follicles, when cells originating from the rete ovarii continuous flux of growing follicles will be detailed in the
surround the oocytes. Germ cell loss appears however to second section of this review. For clarity, only three species
occur at each of these steps. The first which differ in the timings of their phases of oogenesis
(mouse versus sheep and humans) or in their strategies to
use their pool of primordial follicles for folliculogen esis
(mouse and sheep versus humans who display menopause)
* Corresponding author. Tel.: +33-2-41228265; fax: +33-2- 41228251. will be considered.
E-mail address: marcantone.driancourt@intervet.okzonobel.nl (M.A.
0303-7207/00/$ - see front matter © 2000 Elsevier Science Ireland Ltd. All rights reserved. PII:
S0303-7207(99)00246-4
102 K. Reynaud, M.A. Driancourt / Molecular and Cellular Endocrinology 163 (2000) 101–108
2.2. During folliculogenesis
2. Cell death in the ovary
In all species, the store of primordial follicles de creases
2.1. During oogenesis with time. In women, menopause is the conse quence of the
exhaustion of the pool of such follicles. Gougeon et al.
In all species, peak numbers of germ cells are ob served (1994), using mathematical modeling of the decrease in the
around the time of the mitotic to meiotic transi tion size of the primordial pool with time, have demonstrated
(Gondos, 1978). At this stage, the mean germ cell store of a that in women younger than 38-years-old, this decrease is
mouse, cow or woman ovary is 2.5×105, 2.1×106 and produced by growth initia tion and oocyte loss while, in
6.8×106 germ cells, respectively (Baker, 1963; Erickson, older women, it is solely generated by growth initiation
occurring at a faster pace.
1966; Tam and Snow, 1981). In vivo results (i.e.
visualization of ‘atretic divisions’, Baker, 1963) suggest In sheep, comparison of the population of primordial
follicles in ovaries of 2 and 8-year-old ewes has demon
that the rapid proliferation of oogonia may be associated
strated that eight primordial follicles disappear from the
with germ cell loss. Furthermore, strong evidence for germ
reserve pool every day (Driancourt et al., 1985). This
cell loss amongst the popula tion of proliferative oogonia
contrasts with the estimated rate of initiation of growth
has been obtained in vitro (Coucouvanis et al., 1993; Pesce
from the pool (two and three follicles per day) and strongly
et al., 1993). Distinc tive features of the degenerating cells
suggests that five to six follicles die every day within the
are condensed nuclei with clumps of dense chromatin
primordial pool. Since granulosa cell apoptosis is never
located at the nuclear periphery. From their peak number
visualized in such follicles, it may be assumed that oocyte
observed at E13, E80–110 and fifth month of pregnancy
death is the cause of this high death rate within primordial
(mouse, cattle and women, respectively), the number of
follicles (Fig. 1). A similar reasoning demonstrates that
germ cells sharply decreases with two main periods of high
50% of the preantral folli cles disappear through the
germ cell loss, the pachytene stage of meiosis in oocytes
preantral stage (Driancourt et al., 1985; Fig. 1).
and the formation of the primordial follicles (Gondos,
1978). As a consequence, the number of germ cells Although these mathematical considerations strongly
support the idea that oocyte death occurs within pri mordial
enclosed in primordial follicles at birth is less than 20%
and preantral follicles until recently, there were few data
(human, Baker, 1963) or less than 5% (cow, Erickson,
firmly demonstrating it. This is mainly because pycnotic
1966) of its peak number. This is solid evidence demon
bodies, the most common marker used to
strating that the normal fate for a female germ cell during
oogenesis is death.

Fig. 1. Importance of germ cell loss within the pool of primordial follicles and the preantral follicles. These data derive from the counts of follicles in sheep
ovaries at two ages (Driancourt et al., 1985).
K. Reynaud, M.A. Driancourt / Molecular and Cellular Endocrinology 163 (2000) 101–108 103
which cell type is apoptotic. This is why in situ
identify a dying cell, are seldom visualized in oocytes. In identification of apoptotic cells was achieved in recent
addition, because the process of oocyte death ap pears to be studies (De Pol et al., 1997; Driancourt et al., 1997) using
fast (see below), the chances of picking up a small fraction terminal deoxynucleotidyl transferase (TdT) me diated
of dying oocytes within several thou sand and several dUTP nick end labeling (TUNEL) method. This procedure
hundred of primordial and preantral follicles, respectively, uses TdT to catalyze addition of residues of
are rather limited. However, re cent experiments describing digoxigenin-nucleotide to 3% OH ends of fragmented DNA
the consequences on the ovary of targeted expression of generated by internucleosomal cleavage. The apoptotic
bcl2 in mouse oocytes (increased number of healthy cells are then visualized by an anti-digoxi genin antibody
preantral follicles and decreased number of atretic preantral
labeled with peroxydase.
follicles, Morita et al., 1999a), have provided the first solid
Using this approach, De Pol et al. (1997) have shown
evidence demonstrating this event. Furthermore, the
that apoptosis in human foetuses 18-weeks-old is re stricted
observation that bax -/- mice have increased numbers of
primary/ preantral follicles when mature (Perez et al., to germ cells, and that around 8% of them are apoptotic at
1999) also demonstrates that apoptosis is a key regulatory this stage. Furthermore, Driancourt et al. (1997), using
process of primary/preantral numbers. observations of TUNEL stained sections of foetal mouse
Interestingly, once an antrum has appeared within a ovaries (E15.5, E17.5 and birth), have characterized
follicle, there is a clear shift in the respective sensitivi ties changes in apoptosis throughout foetal life in mice and
to cell death of germ cells and somatic cells. This coincides demonstrated a steady decrease in the frequency of
with the disappearance of DNAse I in the oocyte (Boone apoptotic oocytes from 3.691.0 at E15.5, to 2.490.7 and
and Tsang, 1997). Hence, the final tun ing of the number of 1.790.6% at E17.5 and at birth, respectively.
growing follicles to a single ovulatory follicle (cow, Since Morita and Tilly (1999) have shown that germ cell
women) is primarily produced by somatic cell death apoptosis is absent in E13.5 foetal gonads, the above
(reviews, Hsueh et al., 1994; Tilly, 1996). At the large changes in the frequency of apoptotic germ cells suggest
antral stage, it is quite puzzling to note that, at least in that the highest risk of death for female germ cells is
cattle, the oocyte is very resistant to death inducing factors throughout the meiotic prophase (E13–E16, according to
as demonstrated by the ability of oocytes contained in Borum, 1961), with a more limited risk occurring at the
atretic follicles to develop to the blastocyst stage following
time of primordial follicle formation (E18 to birth,
in vitro maturation, fertiliza tion and culture
according to Borum, 1961).
(IVM/IVF/IVC, Blondin and Sirard, 1995).
The rather limited frequency of apoptotic oocytes
estimated in these studies sharply contrasts with the
3. Oocyte apoptosis during oogenesis magnitude of the germ cell loss observed during oogen esis
(see above). Two hypotheses can be put forward to
3.1. E6idence that germ cell loss is through apoptosis reconcile these two findings. Either germ cell apoptosis is
not the only way contributing to the decrease in the number
Solid evidence supporting this claim has been ob tained of germ cells. Some authors (Gondos, 1978) have claimed
in mice using three approaches. Firstly, Coucou vanis et al. that germ cells may leave the ovary by traveling through the
(1993) used flow cytometry to identify (by their small size ovarian epithelium. Alternatively, germ cell apoptosis may
and reduced DNA content) a popula tion of apoptotic cells be a very rapid process, with the TUNEL positive oocytes
within foetal mouse ovaries. Some apoptotic cells were being rapidly destroyed. At least two lines of evidence
identified at E13 (4.3% of the total cells) and their support this last hypothe sis. Firstly, induction of apoptosis
frequency increased to reach 9– 11% of the total cells at by oxidative stress in vitro results in the appearance of an
E15 and E17. Secondly, in a more recent study (Ratts et al., increased propor tion of TUNEL positive oocytes as soon
1995), detection of DNA ladders was achieved on mouse
as 6 h follow ing treatment (Reynaud and Driancourt,
foetal ovaries, starting at E13.5. Apoptosis is associated
unpublished). Secondly, the pioneering studies of Morita et
with activa tion of endonuclease activity with specifically
al. (1999a,b) have demonstrated that in vitro culture of
cleaves cellular DNA between regularly spaced
nucleosomal units (180–200 bp). Such oligonucleosomal foetal ovaries in the absence of specific growth factors
fragments, when separated by agarose gel electrophoresis, results in massive oocyte loss with a 50% drop in oocyte
form a distinctive DNA ladder pattern. While absent in numbers at 48 h and a 90% drop at 72 h of culture. Finally,
E13.5 ovaries, obvious DNA ladders, assumed to be it has been reported (Gumienny et al., 1999) that germ cells
produced by oocyte apoptosis were visualized at E15.5, of Coenorhabditis elegans die by apop tosis and disappear
E18.5 and birth (Ratts et al., 1995). A major drawback of within 1 h.
this
method is that it does not allow the in situ determina tion of
104 K. Reynaud, M.A. Driancourt / Molecular and Cellular Endocrinology 163 (2000) 101–108
Gonocytes
Table 1
What can be learned from survival of PGC and gonocytes PGC Sur6i6al factors
SCF, acts on survival (Dolci et SCF, survival factor al., 1991;
Godin et al., 1991; (Yoshinaga et al., 1991) Pesce et al., Support for a positive role of bclXL is suggested by the
1993) and its
receptor c-kit is present
observations showing that transfec tion of PGC with a bclXL
LIF, increases survival (Dolci et LIF, survival factor (De al., containing construction pro motes their survival (Watanabe
1993; Pesce et al., 1993) Miguel et al., 1996) and its receptor et al., 1997). Since bax KO mice (Perez et al., 1999) and
is present bcl2 KO mice (Ratts et al., 1995) have been generated, the
(Cheng et al., 1994)
observation of their gonads, when at the PGC stage, will
FGF2 FGF mitogenic and/or survival , survival factor (Van factor
(Resnick et al., 1992) Dissel-Emiliani et al., 1996) and FGF
provide informa tion on the effect of these two compounds
receptors are on PGC apoptosis. This has yet to be performed.
present on PGC’s (Resnick et
al., 1998) 3.3. Candidates for the control of apoptosis throughout
IL4, survival factor (Cooke et late oogenesis (E14 to birth)
al., 1996)
Death factors The same approaches as before (in vitro culture with
? Fas/fas L, death inducing factors (Lee et al., 1997) compounds known to be present at this stage, gene
? TGFb1 and TGFb2, death inducing factors (Olaso et al.,
knock-out) have been used to identify relevant candi dates.
1998)
Extensive information suggesting a role of the kit–kit ligand
interaction is available. With the excep tion of E16 when
neither the mRNA nor the kit protein are present, numerous
data (Horie et al., 1991; Keshet et al., 1991; Manova et al.,
3.2. Candidates for the control of oocyte apoptosis in 1993; Yoshida et al., 1997) demonstrate that the kit–kit
oogonia/PGC ligand interaction may be functional. In addition, the recent
results of Morita et al. (1999b), who cultured E13 foetal
Two approaches have been used to provide informa tion ovaries for 3 days, have provided evidence that kit ligand,
in this field. Most of the data reported below have been in the presence of LIF, is a survival factor for oocytes.
obtained in vitro after addition of compounds present in Positive effects of LIF, retinoic acid and Insulin like growth
young foetal gonads to cultured PGC. These data were factor-1 have been reported on oocyte survival by the same
partly extended by observing the extent of apoptosis in group (Morita and Tilly, 1999; Morita et al., 1999a,b), but
foetuses harboring specific gene manipulations. these data should be considered preliminary since it is
Extensive data on apoptosis of PGC/mitotic oogonia unclear whether the concentrations used in vitro are
(mostly obtained in vitro) and some useful information on physiologically relevant. For example, it is unknown
apoptosis of testicular gonocytes are available (Table 1). whether foetal ovaries have the ability to produce IGF
Strong evidence identifying three families (kit/kit ligand; binding proteins regulating IGF-1 bioavailability. On the
LIF and its receptor; FGF and its receptor) as survival same line, negative effects of TGFb on oocyte survival have
factors have been obtained both from the studies on PGC been reported (Morita et al., 1999b) but it is unknown
and testicular gonocytes. Whether expression of the whether activation of TGFb from its latent form may occur
receptor binding a specific compound and its ligand occur in foetal gonads. As regards the fas/fas ligand interaction,
on the same cell (in tracrine regulation) or involves we (Reynaud and Driancourt, un published) have recently
interactions between cells (juxtacrine regulation) is not yet immunolocalized fas and fas ligand in foetal gonads
known. Further more, the partial additivity of LIF and kit between E15 and birth, the proportion of fas ligand positive
ligand effects on young PGC cultured for 3 days (Dolci et cells being similar to the proportion of TUNEL positive
al., 1993) may indicate that survival signals may differ from cells. Furthermore, addition of 50 ng/ml fas ligand to
cell-to-cell. cultured E16 ovaries produced an increased proportion of
Interestingly, testicular gonocytes have also provided apoptotic germ cells.
information on death inducing compounds (Table 1). Both The intracellular mechanisms modulating life and death
TGFb and fas ligand, through the interaction with their have been partly unraveled through the findings obtained
respective receptors, appear to be inimical to gonocyte following gene knock-out. For example, cas pase 2
survival (Lee et al., 1997; Olaso et al., 1998). knock-out results in increased numbers of pri mordial
It is generally agreed that, within a specific cell, the follicles present at birth (Bergeron et al., 1998). Caspase 3
balance between members of two families present within is also present in some oocytes throughout
mitochondria (survival, bcl2+bclXL; death, bax+bclXS) is a
key intracellular modulator of death or survival factors.
K. Reynaud, M.A. Driancourt / Molecular and Cellular Endocrinology 163 (2000) 101–108 105
that bax knock-out does not affect the size of the primordial
late oogenesis (Reynaud and Driancourt, unpublished), was
follicle pool at birth (Perez et al., 1999). Bcl2
although evidence that these oocytes are TUNEL posi tive
is still missing. Reports on the detection of other caspases undetectable by immuno
histochemistry on foetal gonads,
in foetal ovaries are not yet available. Only limited data while the presence of both forms of bclX (bclXL and bclXS) is
support a possible modulating effect of bcl2 family members not yet documented.
on oocyte apoptosis. Bax positive oocytes can be detected
in foetal gonads during late oogenesis (Reynaud and 3.4. Oocyte apoptosis during late oogenesis: which
Driancourt, unpublished) but it is quite puzzling to observe
mechanisms are operating? cytoskeleton proteins and (3) disruption of cell energy
metabolism. Heat shock proteins, particularly hsp90 which
Since apoptosis appears to be a well conserved mech is present in large amounts in oocytes (Ohsako et al., 1995),
anism (Gumienny et al., 1999), the data summarized above, through their chaperone effects, may be involved at this
together with data available on fas-mediated apopotosis (Li stage to control the magnitude of the proteolytic effects of
et al., 1998; Luo et al., 1998), have been used to generate a caspases.
working hypothesis (Fig. 2). Apopto sis in oocytes would This summary figure should be considered prelimi nary
occur as an interaction between fas and fas ligand. This since there is no information at present on Bid or caspase 8
interaction produces activation of caspase 8, which is the in foetal oocytes. The identification of the site(s) of action
apical caspase in the pathway. Active caspase 8 results in of survival factors (block in caspase activation, increased
activation of Bid, a bcl2 family member, and its amounts of mitochondrial bclXL…) also requires further
translocation from the cytosol to the mitochondria. Active investigations.
Bid has the ability to induce complete release of
cytochrome c from mito chondria. In addition, Bid may
inactivate anti apop 4. Oocyte apoptosis during folliculogenesis
totic members of the bcl2 family (particularly bclXL) by
dimerization. The release of mitochondrial cytochrome c 4.1. Follicle/oocyte function in primordial, primary and
may produce caspase 9 activation, followed by cas pase 3 preantral follicles
activation, an event resulting in morphological and
functional changes typical of apoptosis. This in cludes: (1) Changes in gene expression associated with initiation of
alterations within the nucleus occurring as a consequence follicular growth up to the preantral stage have been
of DNAse I activation, PARP fragmenta tion and U1-70 k summarized recently in an excellent review (McNatty et
inhibition; (2) disassembly of cell structure by cleavage of

Fig. 2. Schematic drawing of the apoptotic cascade in foetal oocytes. For details, see text.
106 K. Reynaud, M.A. Driancourt / Molecular and Cellular Endocrinology 163 (2000) 101–108
1999) and TGFb and its receptor in primordial to preantral
al., 1999). Amongst the compounds appearing in pri follicles is available. Furthermore, fas mediated oocyte
mordial oocytes, around growth initiation, GDF-9, fol apoptosis is also likely to occur since there are reports of
listatin and activin A can be listed. In addition, at least in fas ligand (Hakuno et al., 1996) and possibly fas (Kondo et
hamsters and pig, EGF/TGFa has been localized to oocytes al., 1996) expression in oocytes (by
of primordial/primary follicles (McNatty et al., 1999). immunohistochemistry). Furthermore, Lpr mice that
Inhibin A becomes detectable in oocytes at the preantral express little fas antigen due to a mutation have increased
stage. numbers of secondary follicles (Sakamaki et al., 1997; Xu
Most of the compounds which were already operat ing in et al., 1998). Com ponents of the TNFa signaling pathway
foetal oocytes are still present in the postnatal ovary. Solid also appear to be present in oocytes of preantral follicles
evidence demonstrating the presence of kit and kit ligand (Cheng et al., 1993). Caspase 3 appears in contrast to be
(Manova et al., 1993), FGF and its receptor (McNatty et al., present in oocytes of immature gonadotrophin treated rats
(Boone and Tsang, 1998) lowing in vivo injection of an antibody blocking the
There is also solid evidence that members of the bcl2 interaction of these two compounds. However, using the
family (bcl2, bclXL, bax…) play a key role in oocyte culture system of Smitz and Cortvrindt (1998), we were not
apoptosis in primordial to preantral follicles. Such a claim able to demonstrate such an effect on follicle survival after
is supported by the increased numbers of such follicles addition of another blocking antibody to preantral follicles
following bax knock-out (Perez et al., 1999) and by the maintained in vitro (Reynaud and Driancourt, unpublished
decreased numbers of primordial follicles in bcl2 knock-out observations). A common lim itation to all the above
mice (Ratts et al., 1995). Caspase 1 and bclXS may not be studies has to be stressed. They all assess follicle survival
important as their expression was not detectable in fully and not oocyte survival (al though the two parameters are
grown oocytes (Jurisicova et al., 1998). tightly correlated (see above)). At last useful information
was generated by the in vitro models. The demonstration of
4.2. Which of these new compounds are rele6ant to identical survival rates of follicles which contained no
oocyte sur6i6al/death? theca cells and those which had some when dissected
strongly suggests that theca cell secretions do not play a
At least two main strategies (i.e. knock-out of specific key role in follicle/oocyte survival.
genes and in vitro culture) have been fruitful to identify Overall, it is likely that the mechanisms operating to
relevant compounds amongst those identified above. control apoptosis in primordial, primary and preantral
Firstly, the generation of transgenic mice which dis follicles may be even more complex than those operat ing
played defects/blocks in follicular growth demonstrated that in foetal oocytes (Fig. 2). The former oocytes may benefit
GDF-9 (Dong et al., 1996) and follistatin (Guo et al., 1998) from the survival effects of EGF/TGFa and of bcl2 in
were likely to be involved in the control of survival of mitochondria but may suffer from the death inducing effect
primary/preantral follicles or in the control of cell of activin.
proliferation in such follicles. In vitro culture systems were
used to check which of these two hypothe ses was valid.
They demonstrated that the primary role of GDF-9 was 5. Conclusions
mostly on cell proliferation (Hayashi et al., 1999). As
regards follistatin, there are no in vitro data analyzing its The process of oocyte apoptosis, despite its numerical
effect on preantral follicles. But one of the proteins bound importance, is still poorly documented, compared to the
by follistatin, activin, has detri mental effects on follicle large amounts of data available on follicle apoptosis (Hsueh
survival in vitro (Smitz et al., 1998). et al., 1994; Tilly, 1997). The present review has attempted
Secondly, the development of culture systems for to gather evidence underlining its relevance and define a
preantral follicles of a range of species (Eppig and O’Brien, few working hypotheses to study its con trol. It is obvious
1996; Wandji et al., 1996; Smitz and Cortvrindt, 1998) has that germ cell apoptosis, both during the prenatal period
been useful to check which growth factors/proteins present and from birth to adulthood, is more likely to be regulated
in preantral follicles could modulate their survival. The by a complex combination of compounds with positive or
most comprehensive study (Wandji et al., 1996) established negative effects rather than by a single signal.
that EGF and FGF were survival factors while TGFb had a A key question which still has no answer at present is
marked detrimental effect on survival. Good evidence why there is so much oocyte apoptosis. In the long term, a
support ing the relevance of the kit–kit ligand interaction better understanding of its mechanisms could have useful
for the survival of the preantral follicles has been reported consequence on: (1) the protection of oocytes of cancer
by Yoshida et al. (1997) who demonstrated a large and patients; (2) devise of efficient culture
quick drop in the population of preantral follicles fol
K. Reynaud, M.A. Driancourt / Molecular and Cellular Endocrinology 163 (2000) 101–108 107
human ovaries. Proc. R. Soc. B 158, 417–433.
systems for primordial, preantral follicles from primates Bergeron, L., Perez, G.I., Macdonald, G., Shi, L., Sun, Y., Jurisi cova, A.,
Varmuza, S., Latham, K.E., Flaws, J.A., Salter, J.C., Hara, H.,
and domestic animals; and possibly (3) development of new Moskowitz, M.A., Li, E., Greenberg, A., Tilly, J.L., Yuan, J., 1998.
strategies to manage the pool of primordial folli cles to Defects in regulation of apoptosis in caspase 2 deficient mice. Genes
postpone menopause. Dev. 12, 1304–1314.
Blondin, P., Sirard, M.A., 1995. Oocyte and follicular morphology as
determining characteristics for developmental competence in bovine
Acknowledgements oocytes. Mol. Reprod. Dev. 1, 54–62.
Boone, D.L., Tsang, B.K., 1997. Identification and localization of
deoxyribonuclease I in the rat ovary. Biol. Reprod. 57, 813–821. Boone,
We kindly acknowledge J. Smitz and R. Cortvrindt for D.L., Tsang, B.K., 1998. Caspase 3 in the rat ovary: localiza tion and
exciting discussions and efficient collaboration and J.J. possible role in follicular atresia and luteal regression. Biol. Reprod. 58,
Panthier and F. Bernex for provision of the WlacZ/ +mice. 1533–1539.
Borum, K., 1961. Oogenesis in the mouse: a study of the meiotic
prophase. Exp. Cell Res. 24, 495–507.
Cheng, H.L., Marcinkiewicz, J.L., Sancho-Tello, M., Hunt, J.S.,
References
Terranova, P.F., 1993. Tumor necrosis factor a gene expression in
mouse oocytes and follicular cells. Biol. Reprod. 48, 707–714.
Baker, T.G., 1963. A quantitative and cytological study of germ cells in
Cheng, L., Gearing, D.P., White, L.S., Compton, D.L., Schooley, K.,
Donovan, P.J., 1994. Role of leukemia inhibitory factor and its Ecochard, R., Thalabard, J.C., 1994. Age related changes in the population
receptor in mouse primordial germ cell growth. Development 120, of human ovarian follicles: increase in the disappearance rate of
3145–3153. non-growing and early growing follicles in aging women. Biol. Reprod.
Cooke, J.E., Heasman, J., Wylie, C.C., 1996. The role of interleukin-4 in 50, 653–663.
the regulation of mouse primordial germ cell numbers. Dev. Biol. 174, Gumienny, T.L., Lambie, E., Hartwieg, E., Horvitz, H.R., Hengart ner,
14–21. M.O., 1999. Genetic control of programmed cell death in the
Coucouvanis, E.C., Sherwood, S.W., Carswell-Crumpton, C., Spack, E.G., Caenorhabditis elegans hermaphrodite germline. Development 126,
John, P.P., 1993. Evidence that the mechanism of prenatal germ cell 1011–1022.
death in the mouse is apoptosis. Exp. Cell Res. 209, 238–247. Guo, Q., Kumar, T., Woodruff, T., Hadswell, L., de Mayo, F., Matzuk, M.,
De Miguel, M.P., De Boer-Brouwer, M., Paniagna, R., Van den Hurk, R., 1998. Overexpression of mouse follistatin causes reproductive defects
De Rooij, D.G., Van Dissel-Emiliani, F.M.F., 1996. Leukemia in transgenic mice. Mol. Endocrinol. 12, 96–106.
inhibitory factor and ciliary neurotropic factor promote the survival of Hakuno, N., Koji, T., Yano, T., Kobayashi, N., Tsutsumi, O., Taketani, Y.,
Sertoli cells and gonocytes in a coculture system. Endocrinology 137, Nakane, P.K., 1996. Fas/APO-1/CD95 system as a mediator of
1885–1893. granulosa cell apoptosis in ovarian follicle atresia. Endocrinology 137,
De Pol, A., Vaccina, F., Forabosco, A., Cavazzuti, E., Marzona, L., 1997. 1938–1948.
Apoptosis of germ cells during human prenatal oogenesis. Hum. Hayashi, M., Mc Gee, E.A., Min, G., Klein, G., Rose, U.M., Van Duin,
Reprod. 12, 2235–2241. M., Hsueh, A.J., 1999. Recombinant growth differentiation factor 9
Dolci, S., Williams, D.E., Ernst, M.K., Resnick, J.L., Brannan, C.I., Loak, (GDF-9) enhances growth and differentiation of cultured early ovarian
L.F., Lyman, S.D., Boswell, H.S., Donovan, P.J., 1991. Requirement follicles. Endocrinology 140, 1236–1244.
for mast cell growth factor for primordial germ cell survival in culture. Horie, K., Takakura, K., Taii, S., Narimoto, K., Noda, Y., Nishikawa, S.,
Nature 352, 809–811. Nakayama, H., Fujita, J., Mori, T., 1991. The expression of c-kit
Dolci, S., Pesce, M., De Felici, M., 1993. Combined action of stem cell protein during oogenesis and early embryonic development. Biol.
factor, leukemia inhibitory factor and cAMP on in vitro proliferation of Reprod. 45, 547–552.
mouse primordial germ cells. Mol. Reprod. Dev. 35, 134–139. Hsueh, A.J.W., Billig, H., Tsafriri, A., 1994. Ovarian follicle atresia: a
Dong, J., Albertini, D.F., Nishimori, K., Kumar, T., Lu, N., Matzuk, M., hormonally controlled apoptotic process. Endocr. Rev. 15, 707–724.
1996. Growth differentiation factor-9 is required during early ovarian Jurisicova, A., Latham, K.E., Casper, R.F., Varmuza, S.L., 1998.
folliculogenesis. Nature 383, 531–535. Expression and regulation of genes associated with cell death during
Driancourt, M.A., Cahill, L.P., Bindon, B.M., 1985. Ovarian follicu lar murine preimplantation embryo development. Mol. Re prod. Dev. 51,
populations and preovulatory enlargement in Booroola and control 243–253.
Merino ewes. J. Reprod. Fertil. 73, 93–103. Keshet, E., Lyman, S.D., Williams, D.E., Anderson, D.M., Jenkins, N.A.,
Driancourt, M.A., Reynaud, K., Panthier, J.J., Bernex, F., 1997. Copeland, N.G., Parada, L.F., 1991. Embryonic RNA expression
Mechanisms involved in germ cell apoptosis in prenatal and young patterns of the c-kit receptor and its cognate ligand suggest multiple
mice. J. Reprod. Fertil. Abst. Series 20, 9. functional roles in mouse development. EMBO J. 10, 2425–2435.
Eppig, J.J., O’Brien, M.J., 1996. Development in vitro of mouse oocytes Kondo, H., Maruo, T., Peng, X., Mochizuki, M., 1996. Immunolog ical
from primordial oocytes. Biol. Reprod. 54, 197–207. Erickson, B.H., evidence for the expression of the fas antigen in the infant and adult
1966. Development and radio response of the prena tal bovine ovary. J. human ovary during follicular regression and atresia. J. Clin. Endocr.
Reprod. Fertil. 11, 97–105. Metab. 81, 2702–2710.
Faddy, M.J., Gosden, R.G., 1995. A mathematical model of follicle Lee, J., Richburg, J.H., Younkin, S.G., Boekelheide, F., 1997. The fas
dynamics in the human. Hum. Reprod. 10, 770–775. Godin, I., Deed, R., system is a key regulator of germ cell apoptosis in the testis.
Cooke, J., Zsebo, K., Dexter, M., Wylie, C.C., 1991. Effects of the steel Endocrinology 138, 2081–2088.
gene product on mouse primordial germ cells in culture. Nature 352, Li, H., Zhu, H., Xu, C.J., Yuan, J., 1998. Cleavage of BID by caspase 8
807–809. mediates the mitochondrial damage in the fas pathway of apoptosis.
Gondos, B., 1978. Oogonia and oocytes in mammals. In: Jones, R.E. Cell 94, 491–501.
(Ed.), The Vertebrate Ovary. Plenum Press, New York. Gougeon, A.,
108 K. Reynaud, M.A. Driancourt / Molecular and Cellular Endocrinology 163 (2000) 101–108
Ohsako, S., Bunick, D., Hayashi, Y., 1995. Immunocytochemical
Luo, X., Budihardjo, I., Zou, H., Slaughter, C., Wang, X., 1998. Bid, a observation of the 90 kD heat shock protein (hsp 90): high expression
Bcl2 interacting protein, mediates cytochrome c release from in primordial and pre-meiotic germ cells of male and female rat
mitochondria in response to activation of cell surface death receptors. gonads. J. Histochem. Cytochem. 43, 67–76.
Cell 94, 481–490. Olaso, R., Pairault, C., Boulogne, B., Durand, P., Habert, R., 1998.
Manova, K., Huang, E.J., Angeles, M., de Leon, V., Sanchez, S., Transforming growth factor b1 and b2 reduce the number of gonocytes
Pronovost, S.M., Besmer, P., Bachvarova, R.F., 1993. The expres sion by increasing apoptosis. Endocrinology 139, 733–740.
pattern of the c-kit ligand in gonads of mice supports a role for the Perez, G.I., Robles, R., Knudson, C.M., Flaws, J.A., Korsmeyer, S.J.,
c-kit receptor in oocyte growth and in proliferation of spermatogonia. Tilly, J.L., 1999. Prolongation of ovarian lifespan into ad vanced
Dev. Biol. 157, 85–99. chronological age by bax deficiency. Nat. Genet. 21, 200–203.
Mc Natty, K.P., Heath, D.A., Lundy, T., Fidler, A.E., Quirke, L.D., Stent, Pesce, M., Farrace, M.G., Piacentini, M., Dolci, S., De Felici, M., 1993.
V.C., O’Connell, A., Smith, P.R., Groome, N., Tisdall, D.J., 1999. Stem cell factor and leukemia inhibitory factor promote primordial
Control of early ovarian follicular development. J. Reprod. Fertil. germ cell survival by suppressing programmed cell death (apoptosis).
Suppl. 54, 3–16. Development 118, 1089–1094.
Morita, Y., Tilly, J.L., 1999. Segregation of retinoic acid effects on fetal Ratts, V.S., Flaws, J.A., Kolp, R., Sorenson, C.M., Tilly, J.M., 1995.
ovarian germ cell mitosis versus apoptosis by requirement for new Ablation of bcl2 gene expression decreases the number of oocytes and
macromolecular synthesis. Endocrinology 140, 2696– 2703. primordial follicles established in the postnatal female mouse gonad.
Morita, Y., Perez, G.I., Maravei, D.V., Tilly, K.I., Tilly, J.L., 1999a. Endocrinology 136, 3665–3668.
Targeted expression of Bcl-2 in mouse oocytes inhibits ovarian follicle Resnick, J.L., Bixler, L.S., Cheng, L., Donovan, P.J., 1992. Long term
atresia and prevents spontaneous and chemotherapy-in duced oocyte proliferation of mouse primordial germ cells in culture. Nature 359,
apoptosis in vitro. Mol. Endocrinol. 13, 841–850. 550–551.
Morita, Y., Manganaro, T.F., Tao, X.-J., Martimbeau, S., Donahoe, P.K., Resnick, J.L., Ortiz, M., Keller, J.R., Donovan, P.J., 1998. Role of
Tilly, J.L., 1999b. Requirement for phosphatidylinositol 3% kinase in fibroblast growth factors and their receptors in mouse primordial germ
cytokine mediated germ cell survival during fetal oogen esis in the cell growth. Biol. Reprod. 59, 1224–1229.
mouse. Endocrinology 140, 941–949. Sakamaki, K., Yoshida, H., Nishimura, Y., Nishikawa, S.I., Manabe, N.,
Yonehara, S., 1997. Involvement of fas antigen in ovarian follicular
atresia and luteolysis. Mol. Reprod. Dev. 47, 11–18. affect the growth and endocrine function in vitro of granulosa cells of
Smitz, J., Cortvrindt, R., 1998. Inhibin A and B secretion in mouse bovine preantral follicles. Theriogenology 45, 817–832.
preantral follicle culture. Hum. Reprod. 13, 927–935. Smitz, J., Cortvrindt, Watanabe, M., Shirayoshi, Y., Koshimizu, U., Hashimoto, S., Yone hara,
R., Hu, Y., Vanderstichele, H., 1998. Effects of recombinant activin A on S., Eguchi, Y., Tsujimoto, Y., Nakatsuji, N., 1997. Gene transfection of
in vitro culture of mouse preantral follicles. Mol. Reprod. Dev. 50, mouse primordial germ cells in vitro and analysis of their survival and
294–304. growth control. Exp. Cell. Res. 230, 76–83.
Tam, P.P., Snow, M.H., 1981. Proliferation and migration of primor dial Xu, J.P., Li, X., Mori, E., Guo, M.W., Mori, T., 1998. Aberrant expression
germ cells during compensatory growth in mouse embryos. J. and dysfunction of fas antigen in MRL/MpJ-lpr/lpr murine ovary.
Embryol. Exp. Morphol. 64, 133–147. Zygote 6, 359–367.
Tilly, J.L., 1996. Apoptosis and ovarian function. Rev. Reprod. 1, Yoshida, H., Takakura, N., Karaoka, H., Kunisada, T., Okamura, H.,
162–172. Nishikawa, S.I., 1997. Stepwise requirement of c-kit tyrosine kinase in
Tilly, J.L., 1997. Apoptosis and the ovary: a fashionable trend or food for mouse ovarian follicle development. Dev. Biol. 184, 122–137.
thought. Fertil. Steril. 67, 226–228. Yoshinaga, K., Nishikawa, S., Ogawa, M., Hayashi, S.I., Kunisada, T.,
Van Dissel-Emiliani, F.M.F., De Boer-Brouwer, M., De Rooij, D.G., 1996. Fujimoto, T., Nishikawa, S.I., 1991. Role of c-kit in mouse
Effect of fibroblast growth factor 2 on Sertoli cells and gonocytes in spermatogenesis: identification of spermatogonia as a specific site of
coculture during the prenatal period. Endocrinology 137, 647–654. c-kit expression and function. Development 113, 689–699.
Wandji, S.A., Eppig, J.J., Fortune, J.E., 1996. FSH and growth factors

. .

You might also like