You are on page 1of 27

C H A P T E R

2
Regulation of Follicle Formation and
Development by Ovarian Signaling Pathways
Rexxi D. Prasasya

Kelly E. Mayo

Introduction demonstrate the diversity of signaling pathways used


in intrafollicular cell communication.
In mammals, ovarian follicles make up the basic func- During embryonic development, primordial germ cells
tional unit of the female gonad, the ovary. Within the fol- (PGCs) arise at the proximal epiblast and proceed to pro-
licle, the female germ cell, the oocyte, interacts with the liferate, migrate, and eventually colonize the bipotential
surrounding granulosa cells, and they in turn with thecal gonad [1]. While mechanisms of sex determination are
cells, through both physical contact and secreted signals. beyond the scope of this chapter, it is now known that
Bidirectional communication between the oocyte and the the process is somatic cell driven, and ovarian fate is pro-
follicular somatic cells is necessary to ensure fertility moted in the absence of the Y chromosome and Sry gene
through the eventual ovulation of a fertilizable oocyte expression. In the XY individual, the presence of SRY sup-
as well as production of hormones by the somatic cells presses Wnt4/β-catenin pathway signaling (reviewed in
that are important for overall physiology. The endocrine Ref. [2]). Conversely, in the XX individual, the lack of
control of follicular growth and ovulation by the pituitary SRY allows for initiation of pregranulosa cell differentia-
gonadotropins follicle-stimulating hormone (FSH) and tion as well as continued suppression of the Sertoli cell fate
luteinizing hormone (LH) is well recognized from classic by an increase in Wnt4/β-catenin signaling (reviewed in
physiology experiments. More recently, the advent of Ref. [2]). At around embryonic day 13.5 (E13.5) in the
modern molecular biology techniques and genetic mouse mouse or 10 weeks of gestation in the human, a rise in reti-
models has allowed investigations into the cellular noic acid (RA) levels within the embryonic ovary induces
signaling pathways and gene regulatory events that are the expression of stimulated by retinoic acid gene 8 (Stra8),
responsible for follicle formation and development. As which triggers germ cells, or oocytes, to enter meiosis [3,4].
will become clear early in this chapter, a myriad of over- RA in the fetal gonad originates from the mesonephros,
lapping and often interacting pathways govern each and is actively degraded by the P450 cytochrome enzyme
specific stage of follicular development. It is perhaps by Cyp26b1 in the embryonic testis, thus preventing early
design that a built-in redundancy exists in the molecular entry into meiosis in spermatogonia [5]. In contrast,
control of a physiological process that ultimately deter- Cyp26b1 expression is downregulated by E12.5 in fetal
mines the continuation of the species. We begin by focus- mouse ovaries, thus allowing the rise in RA and initiation
ing our discussion on emerging signaling pathways that of meiosis [4,5].
contribute to the least understood stages of follicular Interestingly, meiosis, while necessary for fertility,
development, the formation and activation of primordial does not appear to be required for oocyte differentiation
follicles. Subsequently, we review gonadotropin- or the ability of oocytes to interact with granulosa cells to
independent and -dependent follicular growth by focus- form follicles. While there is an increased rate of germ
ing on several types of dominant local factors that cells loss in Stra8-deficient ovaries, leading to complete

The Ovary 23 © 2019 Elsevier Inc. All rights reserved.


https://doi.org/10.1016/B978-0-12-813209-8.00002-9
24 2. REGULATION OF FOLLICLE FORMATION AND DEVELOPMENT BY OVARIAN SIGNALING PATHWAYS

germ cell depletion by 6–8 weeks of age, the surviving two different sources of pregranulosa cells, one migrating
germ cells are contained within follicular structures, from the mesonephros and one migrating from the sur-
which can be ovulated through exogenous hormone stim- face epithelium [11]. Moreover, migration and the subse-
ulation [6]. Moreover, these 2n oocyte-like cells are able to quent differentiation and association with germ cell nests
produce a zona pellucida (ZP) matrix as would bona fide by these two distinct types of pregranulosa cells occur
oocytes [6]. Meiosis will continue to the diplotene stage of sequentially, resulting in what is now accepted to
meiotic prophase I, at which it arrests and will not be be two waves of primordial follicle formation [10,11].
resumed until postovulation. Following meiotic arrest, Both studies demonstrate that following birth, primor-
germ cell clusters, which arise through incomplete cytoki- dial follicles with embryonically labeled granulosa cells
nesis of rapidly dividing germ cells, begin interacting with (presumably part of the first wave of formation) are
surrounding pregranulosa cells to form a structure termed immediately recruited into the growing follicle pool,
a germ cell syncytia, germ cell cyst, or germ cell nest, which some reaching the early antral stage by PND23 [10,11].
serves as a precursor to primordial follicles [7]. Pregranu- Furthermore, these first wave follicles appear to contrib-
losa cells are thought to arise from at least three distinct ute to fertility in early adulthood, as labeled corpora lutea
origins: somatic cells within the bipotential gonad, the (CLs) are detected in PND90 ovaries [10]. These studies
ovarian surface epithelium, and a structure at the ovary- demonstrate that early events such as follicular formation
mesonephros border termed the rete ovarii [2,8,9]. and activation have direct consequences on fertility dur-
The number of primordial follicles formed following ing adulthood.
completion of germ cell nest breakdown contributes to
establishing the reproductive life span of the individual.
The dynamics of primordial follicle formation and activa- CELLULAR SIGNALS REGULATING GERM
tion were recently studied by two independent groups CELL NEST BREAKDOWN AND
using highly sensitive and temporally inducible reporter PRIMORDIAL FOLLICLE FORMATION
lines to label granulosa cells [10,11]. These studies show
that while some primordial follicles are recruited into A mammalian female is born with a finite number of
the growing population immediately following their for- oocytes, and as such, the subsequent number of primor-
mation, the majority of primordial follicles remain quies- dial follicles formed will in part determine the fecundity
cent for varying periods of time [10,11]. Mork and and reproductive life span of the individual. Fig. 1 illus-
colleagues confirmed that in the mouse, there are at least trates the process of follicle formation and development,

FIG. 1 Formation and development of the ovarian follicle. During gestation, migrating primordial germ cells form connected clusters through
incomplete cytokinesis and aggregation. Following colonization of the embryonic ovary, somatic pregranulosa cells will interact with these germ
cell clusters to form germ cell nests. Primordial follicles are formed when pregranulosa cells invade the germ cell clusters to encapsulate single
oocytes within a follicle. This process is accompanied by programmed oocyte death, leaving only about 30% surviving oocytes at the completion
of nest breakdown. Promoters of nest breakdown include paracrine and juxtacrine signaling through Activin, KITL, NGF, and Notch pathway sig-
naling. Conversely, nest breakdown is inhibited by steroid hormones such as estrogen and progesterone. Primordial follicles are gradually recruited
into the growing pool throughout the reproductive lifetime. The extracellular cues that initiate primordial follicle activation remain unclear; however,
PI3K/AKT and mTORC signaling within the oocyte are known to govern its exit from quiescence. The growing follicle population, in turn, provides
negative feedback to the primordial follicle pool through production of AMH. Oocyte activation is also regulated by a network of oocyte-specific
transcription factors that includes NOBOX, LHX8, SOHLH1, and SOHLH2. These transcription factors regulate expression of oocyte-specific genes
including those involved in ZP matrix deposition and Gdf9 and Bmp15, which encode secreted factors that are necessary for continued follicle growth.
As the follicle reaches the multilayer secondary stage, the final somatic cell layer, the theca layer, is specified through activation of Hh signaling. Up to
this stage, follicle development is independent of pituitary gonadotropins. Following formation of the fluid-filled antral cavity, FSH becomes dom-
inant in regulating proliferation and differentiation of granulosa cells toward the preovulatory phenotype working in concert with local paracrine
signals.

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


CELLULAR SIGNALS REGULATING GERM CELL NEST BREAKDOWN AND PRIMORDIAL FOLLICLE FORMATION 25
accompanied by major factors that are involved in each primordial follicle formation. For example, an ovary that
developmental stage. In the XX individual, PGCs lacks expression of the oocyte-restricted Factor in germ
undergo rapid mitotic division as they migrate and colo- line alpha (Figla), a basic helix-loop-helix transcription
nize the gonad until the initiation of meiosis [12]. This factor that is required for ZP formation [25], does not
mitotic division is accompanied by incomplete cytokine- form follicles, leading to eventual demise of oocytes
sis [13–15], resulting in structures termed nests or syncy- within disorganized germ cell nests [24]. In the following
tia where multiple cells are connected by cytoplasmic sections, we discuss the proposed mechanisms for pro-
bridges, similar to those found in invertebrates [16,17]. grammed germ cell death during the period of nest
There is evidence that organelles, including centrosomes, breakdown, as well as emerging signaling pathways that
Golgi bodies, and mitochondria, are transferred between contribute to the formation of primordial follicles.
sister oocytes through these cytoplasmic bridges, produc-
ing a cloud of organelles termed the Balbiani body within
oocytes that are more likely to survive following cyst res-
olution [18,19]. While there is a strong correlation
Molecular Control of Neonatal Oocyte Survival
between the presence of the Balbiani body and oocyte An experimental tool that has been extensively used to
survival [18], the cytoplasmic bridges do not appear to study signaling pathways during germ cell nest break-
be absolutely necessary for female fertility. This is dem- down is the ex vivo culture of neonatal rodent ovaries
onstrated by a knockout model of Tex14, a component [26]. This organotypic culture system, along with genetic
of the cytoplasmic bridges [20]. While Tex14/ mice mouse models, has revealed many candidate factors and
are endowed with reduced numbers of oocytes as com- signaling pathways that contribute to bidirectional com-
pared to littermate controls following completion of germ munication between somatic pregranulosa cells and
cell nest breakdown, their primordial follicle pool is able oocytes during nest breakdown. Several investigations
to support a normal reproductive life span and full have explored whether modulation of oocyte survival
fertility [20]. can alter the primordial follicle endowment, and thus
Primordial follicles are formed when pregranulosa the reproductive life span. Of interest is the B cell
cells send projections toward the germ cell nests to encap- lymphoma-2 (BCL-2) family of proteins, whose members
sulate an individual oocyte within a follicle [7]. In the are key regulators of cellular apoptosis. These proteins
mouse, germ cell nest breakdown begins shortly before promote or inhibit apoptosis by controlling mitochon-
birth and concludes at about PND6, when more than drial outer membrane permeabilization. Deletion of
80% of germ cells are contained within primordial folli- Bax, a proapoptotic member of the Bcl-2 family, prolongs
cles [7,21]. Only about 30% of oocytes from germ cell reproductive life span by 10–12 months in the mouse due
nests survive to become primordial follicles, as a wave to a decreased rate of follicular atresia [27]. While it was
of oocyte death occurs concurrently with the progression initially thought that the primordial follicle pool is not
of germ cell nest breakdown [7]. It is hypothesized that altered in Bax/ ovaries, a second study shows an
this massive loss of oocytes allows for the optimization increase in the absolute number of germ cells, and of pri-
of the number of available pregranulosa cells to the num- mordial follicles formed, in embryonic and neonatal
ber of surviving oocytes, and also for the remaining Bax/ ovaries [28]. Surprisingly, a wave of oocyte death
oocytes to acquire organelles from those that are lost. during the period of germ cell nest breakdown is still
Alternatively, oocyte death has also been suggested to observed in Bax/ ovaries, suggesting that oocyte death
directly facilitate the resolution of germ cell syncytia into during nest breakdown is Bax-independent.
primordial follicles, by creating smaller syncytia follow- Additional evidence that suggests alternative mecha-
ing selective oocyte lost [7], or as a means to eliminate nisms of apoptotic pathway activation in lieu of BCL-2
oocytes with chromosomal aberrations [22]. family proteins in neonatal oocytes comes from knock-
There is no consensus on whether germ cell nest break- outs of Puma and Bcl-2-modifying factor (Bmf), both proa-
down is driven by the germ cells or by the somatic cells. It poptotic proteins [29,30]. Similar to the Bax/ ovary,
is apparent that interactions between properly specified both Puma/ and Bmf/ ovaries contain more oocytes
somatic pregranulosa cells and oocytes are important at embryonic and neonatal ages [29,30]. In both models,
for successful nest breakdown and establishment of the however, a wave of germ cell death occurs during the
primordial follicle pool. Foxl2, a gene encoding a period of nest breakdown. In the case of Bmf/ ovaries,
winged-helix forkhead gene transcription factor, is one the greater number of germ cells does not result in a
of the earliest markers identified to be required for gran- larger endowment of primordial follicles at the conclu-
ulosa cell specification [23]. Foxl2/ mice are infertile due sion of germ cell nest breakdown [30]. This normalization
to incompetency of pregranulosa cells to interact with in the number of follicles is also observed in an overex-
germ cells clusters to form follicles [24]. In turn, develop- pression model of the antiapoptotic gene Bcl2 where
mental competency of the oocyte is also important in the increased numbers of primordial follicles formed

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


26 2. REGULATION OF FOLLICLE FORMATION AND DEVELOPMENT BY OVARIAN SIGNALING PATHWAYS

are eventually lost, with normalization to that of litter- of steroid hormones to which the newborns or fetuses
mate controls by early adulthood [31]. While the mecha- are exposed [33–37], leading to the hypothesis that estro-
nisms behind the initiation of programmed oocyte death gen and progesterone act as negative regulators of pri-
during nest breakdown remain unresolved, it is generally mordial follicle assembly (Fig. 2). Using ex vivo cultures
accepted that oocyte death occurs via apoptosis following of neonatal rat and mouse ovaries, estradiol and proges-
activation of CASPASE-2. Activation of caspases or terone were found to inhibit primordial follicle forma-
cysteine-aspartic proteases leads to cleavage of cellular tion, resulting in the retention of germ cell nests [38,39].
proteins and triggers apoptosis. At PND4, Caspase2/ The effect of progesterone on nest breakdown does not
ovaries contain greater number of primordial follicles, appear to be due to its conversion to estradiol, since expo-
and thus oocytes, compared to ovaries of littermate con- sure of neonatal ovaries to a nonhydrolyzable form of
trols [32]. Moreover, Caspase2/ oocytes are shown to progesterone, promegestone, also results in retention of
better survive assault from exposure to the chemothera- nests [39]. Estrogen receptor-α (Esr1) is expressed in preg-
peutic agent doxorubicin [32]. While genetic mouse ranulosa cells, and estrogen receptor-β (Esr2) is expressed
models have allowed for identification of necessary as in both pregranulosa cells and oocytes at birth in mouse
well as dispensable molecules in programmed oocyte ovaries [40,41]. Using selective agonists and antagonists
death, the mechanisms by which surviving oocytes are for ER-α, ER-β, and the nongenomic, membrane-bound
able to escape apoptosis remain elusive. form of estrogen receptor, Chen and colleague show that
inhibition of germ cell nest breakdown by estrogen can be
carried out by all three forms of estrogen receptor [40].
The Role of Steroid Hormones in Nest
It is thought that there is a critical window within
Breakdown which nest breakdown must occur, after which pregranu-
In rodents, cattle, and macaques, the period of nest losa cell invasion ceases regardless of whether all oocytes
breakdown coincides with a drastic drop in the levels have been contained in a follicle. Therefore, it is

FIG. 2 Select signals that promote germ cell nest breakdown and primordial follicle formation. The process of germ cell nest breakdown occurs
during midgestation in human and during the neonatal period in the mouse. Experimental evidence has shown that high levels of estrogen and
progesterone inhibit primordial follicle formation. Several signaling pathways that are initiated by growth factors have been shown to promote
primordial follicle formation. Activin stimulates proliferation of pregranulosa cells and promotes Notch signaling in the granulosa cells, which
enhances primordial follicle formation. The Ntrk receptors are localized to both oocytes and pregranulosa cells during the period of nest breakdown
and their ligands are expressed by the pregranulosa cells. Signaling through Ntrks promotes nest breakdown, potentially through upregulation of
Jag1 expression in the oocyte. The juxtacrine signal Notch, which is mediated by the ligand JAG1 in the oocyte and the receptor NOTCH2 in preg-
ranulosa cells, is emerging as an important promoter of nest breakdown and granulosa cell proliferation in the early stages of follicle development.
Signaling through the c-KIT receptor in the oocyte and its ligand KITL in the granulosa cells promotes follicle assembly through its contribution to
programmed germ cell death.

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


CELLULAR SIGNALS REGULATING GERM CELL NEST BREAKDOWN AND PRIMORDIAL FOLLICLE FORMATION 27
postulated that remnants of germ cell nests from delayed formation and maintenance of the primordial follicle pool
breakdown will ultimately lead to the formation of multi- through its influence on granulosa cell proliferation and
oocytic follicles (MOFs). The in vitro findings of the neg- likely, germ cell survival.
ative effects of estrogen on nest breakdown are consistent The importance of receptor tyrosine kinase (RTK) sig-
with in vivo studies where neonatal exposure to the syn- naling in promoting germ cell nest breakdown is begin-
thetic estrogen diethylstilbestrol (DES) [42,43] or the soy ning to be elucidated. Kit ligand (KITL) and its
phytoestrogen genistein [44,45] results in delayed germ tyrosine-kinase receptor c-KIT are localized to pregranu-
cell nest break down and a higher incidence of MOFs losa cells and oocytes, respectively, during ovarian
as compared to control mice. The consequence of MOFs embryonic development (Fig. 2) and have been shown
for fertility remains unclear. Iguchi et al. report reduced to be necessary for PGC migration [54]. Their expression
fertilization capacity of oocytes obtained from ovulated remains during the period of nest breakdown and expo-
MOFs in a mouse model, but the confounding effects of sure of cultured neonatal mouse ovaries to a c-KIT block-
early exposure to DES in these oocytes cannot be elimi- ing antibody leads to the retention of germ cell nests,
nated [46]. However, at least one report suggests that which is hypothesized to result from inhibition of oocyte
using oocytes from MOFs does not lead to any adverse death [55]. Furthermore, exposure of neonatal rats to the
outcomes in human in vitro fertilization [47]. small molecule tyrosine kinase inhibitor imatinib, which
targets several RTKs including c-KIT, leads to retention of
germ cell nests and an increased incident of MOFs [56].
The Roles of Growth Factors and Somatic Cell- Expression of neurotrophic tropomyosin-related
Cycle Progression During Germ Cell Nest kinase (Ntrk) receptors, a family of RTKs, and their solu-
ble neurotrophin ligands is dynamically regulated during
Breakdown
the period of germ cell nest breakdown in neonatal rat
Several growth factors have been implicated in germ ovaries [57]. Knockout mouse models of the ligand nerve
cell nest breakdown. Activin is a member of the TGF-β growth factor (Ngf) or the receptors Ntrk1 and Ntrk2 lead
family of proteins that is expressed in granulosa cells of to a delay in germ cell nest breakdown and reduction in
growing follicles [48]. Treatment of neonatal mice with the number of primordial follicles formed [58,59]. Further
activin leads to a 30% increase in the number of primor- evidence for the involvement of Ntrk family of receptors
dial follicles, and this is associated with increased prolif- in nest breakdown is demonstrated by the enhancement
eration of both granulosa cells, and, to a limited degree, of primordial follicle formation observed in neonatal
the germ cells population (Fig. 2) [21]. Similar to several mouse ovaries that are cultured with connective tissue
genetic models of BCL-2 family protein modulation growth factor (CTGF), whose expression is upregulated
described earlier, the expanded primordial follicle pool during the neonatal period [60]. While the receptor by
that is observed in neonatally activin-exposed ovaries is which CTGF exerts its effects during germ cell nests
not maintained into reproductive maturity, a paradigm breakdown has not been specified, there is evidence of
that is described as an inherent quorum-sensing mecha- its ability to activate NTRK1 in human mesenchymal
nism by the ovary [21]. Follistatin (FST) is expressed in cell [61].
the gonad and functions to bind activin and neutralize Additional evidence for an association between regu-
its action [49]. Germ cell nest breakdown is significantly lations of germ cell nest breakdown and cell cycle pro-
delayed in a mouse model that selectively expresses a gression comes from a knockout model of the cell-cycle
high-affinity isoform of FST (FST-288) due to an increase inhibitor p27KipI. A member of the Cip/Kip subfamily,
in somatic cell proliferation and a decrease in germ cell p27KipI, acts as a cyclin-dependent kinase inhibitor.
death during the neonatal period [50]. Whether this delay p27KipI is localized to somatic cells of primordial folli-
in germ cell nest breakdown leads to MOFs was not char- cles, and p27KipI/ ovaries show an acceleration in germ
acterized [51]; however, the primordial follicle pool is cell nest breakdown, reaching completion by PND2 [62].
expanded in the FST-288 mouse [50]. Despite this, the While proliferation was not directly measured, this result
FST-288 mouse is subfertile due to accelerated depletion suggests that the loss of inhibition of somatic cell prolif-
of the primordial follicle pool in adulthood [52]. Consis- eration facilitates the formation of primordial follicles.
tent with this genetic mouse model, the addition of FST- This is consistent with the correlation between granulosa
288 to ex vivo ovarian culture system during the period of cell proliferation and germ cell nest breakdown that is
germ cell nest breakdown leads to a delay in primordial observed following modulation of activin signaling.
follicle formation [53]. FST-288 treatment leads to The number of primordial follicles formed in p27KipI/
reduced granulosa cell proliferation and suppressed ovaries is double that of litter-mate controls; however,
Notch signaling, a pathway that is known to promote the number of follicles eventually normalizes by early
germ cell nest breakdown, in granulosa cells [53]. These adulthood following massive follicular attrition, consis-
studies illustrate the importance of activin signaling in tent with the quorum-sensing model proposed to govern

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


28 2. REGULATION OF FOLLICLE FORMATION AND DEVELOPMENT BY OVARIAN SIGNALING PATHWAYS

follicular dynamic in the ovary [63]. p27KipI also appears granulosa cells as follicles are activated and mature.
to control primordial follicle maintenance, which we will The oocyte expresses predominantly the ligand JAG1 at
discuss in more detail in the subsequent section, as this time.
p27KipI/ mice are infertile due to a premature depletion The importance of functional Notch signaling was
of the primordial follicle pool [62]. demonstrated using an ex vivo ovarian culture system.
Following a culture period that represents the period of
germ cell nest breakdown, treatment of neonatal mouse
Regulation of Germ Cell Nest Breakdown by ovaries with the γ-secretase inhibitor, DAPT, leads to
the retention of germ cell nests and formation of fewer
Notch Signaling primordial follicles as compared to the vehicle controls
To form primordial follicles, pregranulosa cells must [75,76]. DAPT treatment also leads to suppression of
invade germ cells clusters and form physical contacts oocyte-specific transcription factors that are known to
with the oocyte that will eventually be surrounded and be important for early follicular development such as
survive. While direct contact between the first layer of newborn ovary homeobox protein (Nobox), Figla, sper-
granulosa cells and the oocyte is initially unimpeded, matogenesis and oogenesis specific basic helix-loop-helix
ZP matrix deposition following primordial follicle activa- 2 (Sohlh2), and LIM/homeobox protein 8 (Lhx8) [77].
tion will eventually limit these continuous cell-to-cell con- A disintegrin and metalloproteinase 10 (ADAM-10) is
tacts. To maintain physical contact with the oocyte, an integral component of the proteolytic cleavage
granulosa cells are able to send cytonemes that traverse sequence that leads to the liberation of NICD [78].
the ZP and form both adherens and gap junctions with In vitro treatment of embryonic ovaries with an ADAM-
the oocyte plasma membrane, termed transzonal projec- 10 inhibitor or conditional deletion of ADAM-10 in
tions (TZPs) [64]. While initially thought to exclusively somatic pregranulosa cells leads to suppression of Notch
function as a means for granulosa cells to shuttle nutri- signaling (as identified by reduced level of NICD and
ents in response to the metabolic demands of the oocyte decreased expression of Notch target genes Hey2 and
during development, the presence of TZPs provides a Hes1) and retention of germ cell nests [79].
rationale for investigations into the potential role of In other mouse models, conditional deletion, using the
cell-contact-dependent signaling during folliculogenesis. Cre-loxP system, of the most abundantly expressed Notch
Among these, the highly conserved Notch juxtacrine sig- receptor, Notch2, in the pregranulosa cells (N2KO) leads
naling pathway has emerged as a key regulator of germ to decreased numbers of primordial follicles and forma-
cell nest breakdown. In mammals, Notch signaling tion of MOFs [74,80]. The rate of apoptosing oocytes is
occurs when one of the five-membrane-bound ligands significantly decreased at PND1 in N2KO ovaries, which
(JAGGED1-2, DLL1,3,4) bind to one of the four- is associated with delayed germ cells nests breakdown
membrane-bound receptors (NOTCH1-4) on an oppos- [80]. A null mutation in the Notch receptor modifier Luna-
ing cell [65]. Ligand binding leads to conformational tic fringe (Lfng) also reveals the presence of MOFs and
changes of the Notch receptor, initiating a proteolytic cas- infertility [81]. Overall, these studies demonstrate the
cade that will ultimately lead to a γ-secretase-dependent importance of canonical Notch signaling in the somatic
cleavage and liberation of the Notch receptor intercellular pregranulosa cells for proper follicular formation.
domain (NICD) [66]. NICD will translocate into the As expected, conditional deletion of the most abun-
nucleus and function as a transcriptional coactivator of dantly expressed ligand Jag1, from the oocytes (J1KO)
Notch target genes upon binding with the obligate cofac- phenocopies, in a slightly more robust manner, the
tor RBPJ [67]. Notch signaling is known to regulate many N2KO line [74]. Jag1 expression within the oocyte was
cell-fate decisions during development [68], and to con- recently described to be controlled by RAC1, a small
trol the proliferation and differentiation of somatic follicle GTPase switch that affects gene transcription via the
cells (analogous to somatic pregranulosa cells in mam- STAT3 pathway (Fig. 2) [82]. Inhibition of RAC1 leads
mals) during ovariole development in the Drosophila mel- to decreased numbers of primordial follicles formed
anogaster ovary [69]. and reduced expression of several oocyte specific factors,
Notch receptors and ligands are expressed in neonatal including growth differentiation factor-9 (Gdf9), bone
and adult ovaries of mice [70–72]. Using a transgenic morphogenic protein-15 (Bmp15), Nobox, and Jag1 [82].
Notch reporter mouse line in which the green fluorescent MOFs in N2KO and J1KO mice can be extremely large,
protein (Gfp) is expressed in an NICD/RBPJ-dependent containing many oocytes, suggesting that they are indeed
manner [73], Notch active cells are detected as early as remnants of incompletely broken down germ cells nests
E15.5 in the mouse ovary [74]. By E18.5, Notch activity [74]. Both the N2KO [80] and J1KO [74] lines are subfer-
is detected in granulosa cells that are actively reorganiz- tile, indicating that disruption of critical signals during
ing to encapsulate germ cells clusters [74]. Reporter activ- follicular formation may lead to adverse fertility out-
ity and Notch receptor expression continue in somatic comes. Together, these genetic mouse lines support the

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


REGULATION OF PRIMORDIAL FOLLICLE ACTIVATION 29
model that physical contact between oocyte and pregra- Amh/ ovaries compared to wild-type littermates [92].
nulosa cells facilitates initiation of Notch signaling, which By 4 months of age, the number of primordial follicles
in turn acts as a critical mean of bidirectional communi- is significantly reduced in Amh/ ovaries, becoming
cation between the two cell types during follicular assem- almost completely depleted by 13 months of age [93].
bly (Fig. 2). Despite the increase in the absolute number of growing
It is interesting to note that many of the previously follicles, Amh-/- ovaries do not contain an increased pro-
discussed candidates for regulators of germ cell nest portion of preovulatory follicles due to an enhancement
breakdown appear to function upstream of Notch signal- in the rate of atresia of small preantral follicles [94]. Con-
ing. Progesterone exposure of embryonic and neonatal versely, addition of AMH to ex vivo cultured ovaries leads
ovaries, which is shown to suppress follicular assembly, to a decrease in the growing follicle population by about
inhibits expression of Jag2, Notch2, and a known Notch 40% [93]. These studies suggest that the growing follicle
target gene Hey2 [83]. Jag1, Hes1, and Hey2 were identi- population plays a role in regulating the dynamics of acti-
fied to be significantly downregulated in neonatal vation of the primordial follicle pool through production
Ntrk2/ ovaries, in which reductions in follicular assem- of AMH, which acts as a maintenance signal.
bly and granulosa cell proliferation are observed [84]. Despite unresolved questions regarding the extracellu-
Additionally, lentiviral-mediated targeted expression of lar signals initiating activation, studies involving the
Jag1 in oocytes of Ntrk2/ ovaries was shown to rescue intracellular pathways regulation of follicle activation
the defects in granulosa cell proliferation [84]. Fig. 2 sum- have been significantly advanced, largely through the
marizes known as well as proposed interactions between use of mouse genetic models. Fig. 3 summarizes pheno-
different signaling pathways that regulate germ cell nest types of relevant mouse genetic models that are discussed
breakdown. It will require further exploration to under- in the proceeding sections. In addition to discussing path-
stand whether Notch signaling acts as a potential integra- ways intrinsic to the oocyte and their role in primordial
tor of signals that promote germ cell nest breakdown in follicle activation, we review below the networks of tran-
the mammalian ovary. scriptional control associated with oocyte and granulosa
cell differentiation during the transition from the primor-
dial to the primary follicle stage.
REGULATION OF PRIMORDIAL FOLLICLE
ACTIVATION
Intracellular Signaling Pathways That Regulate
Throughout the reproductive life span, growth is initi- Primordial Follicle Activation
ated in a portion of primordial follicles, and they transi-
tion to the activated primary follicle stage. Regulation of Oocyte Activation by the PI3K/AKT
Morphologically, this is characterized by the transition Pathway
of the single layer of squamous granulosa cells into cuboi- Perhaps the most intensely investigated pathway in
dal cells, as well as deposition of a ZP matrix by the primordial follicle activation is PI3K/AKT signaling
oocyte [85,86]. In turn, a cohort of these small activated within the oocyte. A major kinase cascade that can be acti-
follicles will be recruited to undergo further growth dur- vated by RTKs, the PI3K/AKT pathway (Fig. 4) functions
ing each reproductive cycle, with their final fates being to regulate metabolism, growth, proliferation, and sur-
either atresia or ovulation. Perhaps one of the least under- vival through activation of gene transcription or protein
stood aspects of the mammalian ovary is the physiolog- synthesis. Following ligand binding, some RTKs trans-
ical cues that are responsible for initiating the duce signals through production of the plasma mem-
activation of some, but not all, primordial follicles at brane lipid phosphatidylinositol 3,4,5-triphosphate
any given moment. (PIP3) by phosphoinositide 3-kinase (PI3K). PIP3 acts as
One candidate for a signal regulating primordial folli- an anchor for phosphoinositide-dependent kinase
cle activation is the Anti-M€ ullerian hormone (AMH). I (PDK1), which will in turn phosphorylate and activate
A member of the TGF-β superfamily of proteins, AMH the serine/threonine kinase AKT. Direct targets of AKT
is produced by granulosa cells of growing follicles in are numerous and include transcription factors or repres-
the ovary [87,88]. Clinically, AMH is used as a marker sors such as Foxo3 [95]. An important antagonist of
of ovarian follicular reserve due to the well-characterized PI3K/AKT signaling is the phosphatase and tensin
positive correlation between its serum levels and antral homolog (PTEN), which functions to dephosphorylate
follicle counts [89]. Serum levels of AMH decline with PIP3 to phophatydylinositol 4,5-biphosphate (PIP2), thus
age, and AMH levels are often found to be low or unde- abrogating the anchoring of PDK1 to the plasma
tectable in clinical cases of primary ovarian insufficiency membrane.
[90,91]. Female mice with a null mutation of Amh are fer- FOXO3 is a transcriptional repressor that is now
tile; however, more growing follicles are detected in adult accepted to function as a brake for primordial follicle

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


30 2. REGULATION OF FOLLICLE FORMATION AND DEVELOPMENT BY OVARIAN SIGNALING PATHWAYS

FIG. 3 The balance of activating and maintenance cues


in the primordial follicle pool. (A) PI3K/AKT and mTORC
pathway activation, which promotes the exit of primordial
follicles from quiescence, is restrained by molecules such as
Pten and the Tsc1/2 complex. The transcriptional repressor
FOXO3 also serves as a molecular brake for primordial fol-
licle recruitment. Growing follicles secrete AMH that acts
as a maintenance signal for the primordial follicle pool,
most likely acting within the granulosa cells of these pri-
mordial follicles. Balanced activation and maintenance sig-
nals result in cyclic recruitment of primordial follicles
throughout the reproductive lifespan. (B) Genetic models
that abrogate the function of maintenance molecules lead
to unrestrained activation of the PI3K/AKT/mTORC path-
way. This results in activation of the entire primordial fol-
licle pool that leads to exhaustion of the follicular reserve
and premature reproductive senescence. (C) Conversely,
genetic models that prevent activation of the PI3K/AKT/
mTORC pathway lack follicular development beyond the
primordial stage. In these models, a permanent block in pri-
mordial follicle activation will eventually lead to atresia
and premature reproductive senescence.

activation. Initial studies revealed premature reproduc- oocytes, generated using a distinct recombination strat-
tive senescence due to unrestricted, global activation of egy, it was shown that the resulting unrestrained activa-
the primordial follicle pool in a Foxo3-/- mouse line tion of the PI3K/AKT pathway leads to
(Fig. 3) [96]. In wild-type ovaries, FOXO3 is localized to hyperphosphorylation, and thus degradation, of FOXO3
both nucleus and cytoplasm of oocytes of primordial [99]. Interestingly, abrogation of AKT activation through
and primary follicles [97]. FOXO3 then becomes unde- conditional deletion of Pdk1 in the oocyte also leads to
tectable in oocytes of secondary follicles and beyond infertility [101]. In this mouse model, long-term blockade
[97]. Conditional Foxo3 ablation from the oocyte [98] of primordial follicle growth proved to be detrimental,
leads to global primordial follicle activation identical to eventually resulting in clearance of these nongrowing fol-
that observed in the conventional Foxo3-/- ovary, confirm- licles and depletion of the primordial follicle pool by early
ing its oocyte-centric function [99]. Identification of adulthood [101]. These studies show that AKT activation
FOXO3 as a likely suppressor of oocyte activation in the oocyte is both necessary and sufficient for activa-
prompted follow-up investigations into the PI3K/AKT tion of the primordial follicle. Somewhat counterintui-
pathway as its potential upstream regulator. tively, however, oocyte-specific constitutive activation
Conditional deletion of the negative regulator of the of PI3K (PI3KCA), which leads to hyperactivation of
PI3K/AKT pathway, Pten, from the oocyte leads to acti- AKT, does not completely phenocopy the loss of Pten
vation of the entire primordial follicle pool, resulting in its from the oocyte [102]. By PND50, the primordial follicle
premature depletion and ovarian failure by early adult- pool is not completely depleted in oocyte-specific Pi3kca
hood [100]. In a similar model of Pten knockout from transgenic mice, yet still shows a reduction by about 50%

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


REGULATION OF PRIMORDIAL FOLLICLE ACTIVATION 31

FIG. 4 Intraoocyte signaling pathways and regulation of granulosa cell differentiation in primordial follicle activation. Through several genetic
models, activation of the PI3K/AKT and mTOR signaling pathways has been shown to promote the exit of the primordial oocyte from quiescence.
The initiating extracellular cues for these pathways still require further investigation, with RTKs such as cKIT as potential candidates. Granulosa cells
of primordial follicles likely provide such oocyte-activating factor(s). mTOR and the JAK/STAT3 pathways have been shown to promote pregra-
nulosa cell activation, while the transcription factors FOXL2 and GATA4/6 are known regulators of granulosa cell differentiation. Activation of the
PI3K/AKT cascade leads to phosphorylation and inactivation of the transcriptional repressor FOXO3. This allows for expression of genes that
promote follicle activation and growth. AKT activation also inhibits the Tsc1/2 complex, which allows for activation of mTORC resulting in the
promotion of translation of proteins regulating oocyte growth.

compared to wild-type littermates [102]. The slower rate neonatal mouse ovaries to a PTEN inhibitor, bpV(pic),
of global primordial follicle activation in oocyte-specific resulted in activation of the PI3K/AKT pathway and
Pi3kca transgenic mice may be attributed to the increased FOXO3 exclusion in oocytes of primordial follicles
survival rate of follicles at all stages of development [102], [105]. Transplantation of treated ovaries into ovariecto-
suggesting a secondary role of AKT in maintaining mized hosts revealed an enhanced rate of follicular
oocyte and follicle survival. growth as compared to controls and in vitro fertilization
Findings regarding the involvement of the PI3K/AKT of the recovered matured oocytes, followed by embryo
pathway in primordial follicle activation have led to transfer, is able to generate viable and fertile offspring
explorations into the potential use of pharmacological [105,106]. Similarly, treatment of human ovarian cortical
modulation of PI3K/AKT in clinical settings. Increas- tissues with bpV(pic) followed by xenograft to immune-
ingly improved outcomes of pediatric cancer cases have deficient mice allows for an increased rate of follicular
led to growing interest in the development of fertility- development to the preovulatory stage and recovery of
preserving regiment against gonadotoxic chemothera- meiotically competent oocytes [105].
peutics. At the forefront is the cryopreservation of cortical Earlier, we discussed the role of KITL and the c-Kit
ovarian tissues consisting mostly of dormant primordial receptor in regulation of germ cell nest breakdown. Early
follicles, followed by in vitro maturation of follicles and observations in ovaries of a naturally occurring mouse
eventually the recovery of fertilizable oocytes [103,104]. line with a mutation (Steel Panda) that lead to severely
Efficient means of promoting activation of cryopreserved reduced expression of KITL (Kitlg) revealed a block in
primordial follicles may greatly improve fertility out- follicle development at the primordial and primary
comes for these cancer survivors. Acute exposure of stages [107]. Abrogation of KITL binding to c-KIT using

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


32 2. REGULATION OF FOLLICLE FORMATION AND DEVELOPMENT BY OVARIAN SIGNALING PATHWAYS

antibody ACK2 was shown to block resumption of pathway positively regulates mTORC1 activity through
growth of primordial follicles in both in vivo and inactivation of the mTORC1 inhibitors TSC1 (tuberous
ex vivo models across several different species sclerosis complex 1) and TSC2.
[26,108,109]. In turn, addition of KITL to ex vivo cultured Characterization of one of the Pten oocyte-specific
ovaries promotes primordial follicle activation [109]. knockout mouse lines revealed increased phosphoryla-
c-KIT is a RTK that is localized to the oocyte, and c-KIT tion of rpS6 as compared to wild-type littermates, impli-
inhibition appears to phenocopy the block to primordial cating the involvement of mTOR signaling in the
follicle activation seen in models of PI3K/AKT inhibition observed global activation of primordial follicles
[101]. This suggests that c-KIT activation may be at least (Fig. 3) [100]. Inactivation of mTORC1 through treat-
partially responsible for PI3K/AKT activation within the ment with rapamycin is able to partially preserve the
oocyte during the primordial to primary follicle transi- primordial follicle pool in the oocyte-specific Pten-
tion. Indeed, short-term culture of oocytes that are col- knockout ovary, suggesting that functions of PTEN in
lected from mouse primordial follicles with KITL leads the oocyte are partially mediated through mTORC1
to nuclear export, and as a consequent inactivation, of [113]. Additional evidence for the importance of the
the transcriptional repressor FOXO3, which is dependent mTOR pathway in primordial follicle activation comes
upon activation of the PI3K/AKT-signaling pathway from conditional deletion of Tsc1 (OoTsc1/) or Tsc2
[110]. An oocyte-specific Kit gain-of-function mutation (OoTsc2/) in oocytes resulting in global primordial
leads to global primordial follicle activation, while follicle activation (Fig. 3) [114,115]. S6K1 and rpS6 are
oocyte-specific Kit inactivation leads to an extended block hyperphosphorylated in OoTsc1/ or OoTsc2/ oocytes,
to follicular development at the primordial stage that indicating increased activation of the mTOR pathway
eventually results in global follicular atresia (Fig. 3) [114,115]. Global primordial follicle activation in
[111]. Hyperactivation of AKT and cytoplasmic localiza- OoTsc1/ ovaries can be attributed entirely to over
tion of Foxo3 are observed in oocyte-specific Kit gain-of- activation of mTORC1, since long-term treatment with
function ovaries, while persistent nuclear localization of rapamycin completely abrogates the observed depletion
Foxo3 is observed in oocyte-specific Kit-inactivated ova- of the primordial follicle pool [114]. While mTORC1 in
ries [111]. While these studies provide strong evidence for the oocytes appears to be sufficient for activating
Kit involvement in primordial follicle activation, the fact primordial follicles, it does not seem to be necessary,
remains that only a limited number of primordial follicles due to the compensatory potential of the PI3K/AKT
are activated at any given time, regardless of c-Kit expres- pathway. Regulatory-associated protein of mTORC1
sion in the oocyte population [112]. Therefore, it is highly (RPTOR) is a part of the mTORC1 complex and is
likely that additional regulators, perhaps spatially or required for mTORC1 assembly. Conditional deletion
temporally modulated cofactors to the c-KIT signaling of Rptor from the oocyte (OoRptor/) results in no repro-
pathway, are involved in the process of primordial folli- ductive phenotypes despite the loss of mTORC1 activity
cle activation. [116]. AKT is hyperphosphorylated in OoRptor/
oocytes, suggesting that AKT signaling without mTORC1
Contribution of mTOR Signaling to Primordial activity, presumably through inactivation of FOXO3,
Follicle Activation is able to support a normal rate of primordial follicle
In addition to inactivation of Foxo3, the PI3K/AKT activation [116].
pathway in the oocyte has also been linked to mamma- Interestingly, mTOR also appears to be important in
lian target of rapamycin (mTOR) signaling in the regula- maintaining primordial follicle quiescence through sig-
tion of primordial follicle activation. The mTOR pathway naling within the granulosa cells. Granulosa cell-specific
(Fig. 4) serves as a regulator of cell metabolism, growth, disruption of Rptor (PfGC-Rptor/) leads to a prevalent
proliferation, and survival by integrating diverse extra- block in follicular activation, resulting in very few grow-
cellular and intracellular cues. Central to mTOR signaling ing follicles by juvenile age (Fig. 3) [117]. In contrast,
is the assembly of the large multidomain mTOR com- granulosa cell knockout of the mTORC1 negative regula-
plexes (mTORC1 and mTORC2) of serine-threonine tor Tsc1 (PfGC-Tsc1/) leads to global activation of pri-
kinases. Instead of induction of gene transcription per mordial follicles, a phenotype that can be reversed by a
se, mTOR signaling promotes protein synthesis through regimen of rapamycin treatment [117]. Granulosa cells
activation of components of the translation machinery of PfGC-Tsc1/ ovaries express elevated levels of KITL,
following phosphorylation by mTORC. Some targets of which leads to hyperactivation of the c-KIT-mediated
mTORC include the eukaryotic initiation factor PI3K/AKT pathway within oocytes [117]. Indeed, phar-
4E-binding protein 1 (4E-BP1) and the p70 ribosomal s6 macological abrogation of the PI3K/AKT pathway is
kinase 1 (S6K1). The ribosomal protein S6 (rpS6) is a sub- able to rescue the global primordial follicle activation
strate of S6K1 and its phosphorylation; thus, rpS6 phos- phenotype of PfGC-Tsc1/ ovaries [117], consistent
phorylation is often used as an experimental indicator with findings that have been discussed earlier in this
of mTOR pathway activation. The activated PI3K/AKT section. This result suggests that granulosa cell-secreted

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


REGULATION OF PRIMORDIAL FOLLICLE ACTIVATION 33
factors, such as KITL, likely act as initiating cues for Sohlh1/ and Sohlh2/ ovaries is comparable to wild-
the oocyte to exit dormancy and for the follicle to enter type littermates, follicles with cuboidal granulosa cells
the growing pool (Fig. 4). are never observed, indicating a failure of primordial fol-
licles to transition into the primary stage [122,127].
SOHLH1 and SOHLH2 homodimerize, as well as hetero-
dimerize, and promote expression of oocyte specific
Oocyte-Specific Transcriptional Networks That
genes such as Nobox and Lhx8 [126,128]. Collectively,
Promote Primordial Follicle Activation these studies begin to clarify a transcriptional network
Several oocyte-specific transcription factors are impor- that regulates the process whereby primordial follicles
tant in the transition from the primordial to the primary remain dormant, or are activated to form primary
follicle stage. The search for master regulators of primor- follicles.
dial follicle activation has been performed by a combina-
tion of in silico analysis, gene expression studies, and
genetic mouse models. Nobox is one of the first oocyte- Granulosa Cell Differentiation During the
specific homeobox genes identified through in silico
analysis of NCBI UniGene database of a neonatal mouse
Primordial to Primary Follicle Transition
ovary cDNA library [118]. Detected in oocytes beginning As discussed in the last section, granulosa cells of pri-
at E15.5, high expression of Nobox remains throughout mordial follicle likely provide ligands or extracellular
folliculogenesis. While the males are completely fertile, cues that initiate the intra-oocyte cascades that lead to fol-
Nobox/ females are infertile due to an almost complete licle activation. Recently, activation of the JAK/STAT3
loss of oocytes by 6 weeks of age [119]. Although primor- pathway in granulosa cells of primordial follicles was
dial follicles are initially observed, no growing follicles shown to enhance primordial follicle recruitment [129].
are ever detected in Nobox/ ovaries, suggesting a failure Further analysis on the downstream consequences, and
in primordial follicle activation. By PND14, Nobox/ particularly of the changes in secreted products following
ovaries contain mostly degenerating follicles which JAK/STAT3 pathway activation, may aid in identifying
lack oocytes. At the molecular level, Nobox binds to additional candidates for primordial oocyte-activating
the promoters of the oocyte specific genes Oct4 and factors.
Gdf9, and expression of these genes is lost in Nobox/ Failure of granulosa cells to undergo proper differen-
ovaries [119,120]. This finding is consistent with the tiation often leads to unsynchronized development
clinical observation of high prevalence of NOBOX between the oocyte and granulosa cells, which is detri-
mutations, leading to compromised ability to bind the mental to the survival of the follicle. Previously, we dis-
Gdf9 promoter, in patients with primary ovarian cussed the failure of pregranulosa cells to interact with
insufficiency [121]. germ cell nests and properly form follicles in Foxl2/
The lim-homeobox protein, LHX8 is localized to mice [130]. A distinct transgenic line of Foxl2 disruption
oocytes throughout follicle development [122]. While (Foxl2lacZ), while appearing to undergo primordial follicle
conventional knockout of Lhx8 results in very similar formation, is infertile due to a block in follicular develop-
phenotypes to those observed in Nobox/ mice, charac- ment during the primordial to primary stage transition
terized by failure of primordial follicles to transition into [23]. Foxl2lacZ squamous granulosa cells never become
the growing pool and eventual global atresia by PND7, cuboidal, and thus remain nonproliferative [23,85]. Inter-
conditional oocyte deletion of Lhx8 reveals a more spe- estingly, in 2-week-old Foxl2lacZ ovaries, Kitl expression is
cific function [123], with global primordial follicle activa- significantly upregulated in the granulosa cells, resulting
tion due to hyperactivation of the PI3K/AKT pathway in stimulation of Gdf9 expression in almost all oocytes, a
[124]. LIN28A, a known positive regulator of the AKT/ hallmark of a growing oocyte population, and thus global
mTOR pathway, is elevated in the oocyte-specific Lhx8 follicular activation [131]. Despite the failure of the gran-
knockout, and indeed, Lhx8 was found to bind the Lin28a ulosa cells to differentiate from the dormant squamous
promoter and suppress its expression [124]. morphology, oocytes do undergo activation in Foxl2lacZ
Sohlh1 and Sohlh2 encode basic helix-loop-helix tran- ovaries, suggesting a failure of the germ cells and somatic
scription factors that are expressed exclusively in female cells to coordinate their developmental transitions [23].
germ cells beginning at E15.5 and E12.5, respectively The importance of proper differentiation of granulosa
[122,125,126]. While expression of Sohlh1 and Sohlh2 is cells to follicle survival and primordial to primary stage
robust in oocytes within germ cell nests and primordial transition is further illustrated by a transgenic mouse tar-
follicles, their expression diminishes in primary follicles geting the gonadal specific transcription factors GATA4
and beyond, suggesting that their functions are restricted and GATA6. GATA proteins are zinc finger transcrip-
to early follicular development. Sohlh1 [122] or Sohlh2 tions factors, and GATA4 and GATA6 are predominantly
[127] knockout females are infertile due to lack of follicu- expressed in granulosa cells beginning at the embryonic
lar growth. While the size of the primordial follicle pool in age in the mammalian ovary and function to regulate

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


34 2. REGULATION OF FOLLICLE FORMATION AND DEVELOPMENT BY OVARIAN SIGNALING PATHWAYS

expression of numerous genes that are important various models of FSH signaling disruption [135–137].
throughout follicular development, including those While gonadotropin-responsive antral follicles are never
involved in follicular growth and steroid biosynthesis observed in genetic models lacking FSH (through abroga-
[132,133]. Deletion of both Gata4 and Gata6 in granulosa tion of the β-subunit of FSH) [135] or lacking the FSH
cells leads to a loss of expression of granulosa cell receptor (FSHR) [136,137], the numbers of primordial,
markers, including Foxl2, indicating compromised gran- primary, and multilayered preantral follicles are unaf-
ulosa cell identity [134]. Consistent with the most prom- fected. This suggests that local signals are predominant
inent phenotype of Foxl2lacZ ovaries, granulosa cells do in regulating the development of follicles through the
not transition from a squamous to cuboidal morphology preantral stages.
following Gata4 and Gata6 deletion, leading to a lack of The acquisition of additional layers of granulosa cells
growing follicles and eventually to clearance of the per- during the transition between the primary and the sec-
manently dormant primordial follicle pool through ondary follicular stages and beyond involves many
wide-spread atresia [134]. While currently less well diverse, and often highly interacting signaling pathways.
understood than the expansive oocyte-specific transcrip- Moreover, during this period, the oocytes undergo size
tional network that is implicated in primordial follicle expansion that needs to be tightly coordinated with the
activation, future investigations into novel FOXL2 and growth of the granulosa cells. Finally, as follicles reach
GATA4/6 targets should provide further important the secondary stage, local signals are required to initiate
insights into the concurrent differentiation of follicular the recruitment and organization of the thecal cell layer, a
granulosa cells. second type of somatic cell that is indispensable for ste-
roidogenesis. Fig. 5 illustrates the diversity of signaling
pathways that are involved in gonadotropin-
CELLULAR SIGNALING DURING independent granulosa cell growth, as well as the multi-
PREANTRAL FOLLICULAR directional communication between the oocyte and
DEVELOPMENT somatic cells during this preantral period. While we rec-
ognize the complexity of many pathways involved in
The growth of follicles between the primary and antral these preantral developmental events, we first focus dis-
stages is accepted to be independent from the pituitary cussion on paracrine signaling through the TGF-β super-
gonadotropin, FSH. This paradigm is derived from the family of peptides, whose members are highly
unaltered preantral follicular development observed in represented among both oocyte-secreted and somatic

FIG. 5 Regulation of gonadotropin-


independent follicular growth. The
growth of ovarian follicles prior to
antrum formation is independent of
FSH and is governed by various modes
of local signaling. Following activation,
oocytes begin secreting GDF9 and
BMP15, which act on granulosa cells to
promote growth and gene expression.
Under the influence of GDF9, granulosa
cells express the Hh ligand that functions
to specify the final somatic cell layer, the
theca cells. Activin is a mitogen that is
secreted and signals within the granulosa
cell layer. Activin promotes expression of
Esr2 and suppresses expression of
Cyp26b1. This results in increased levels
of ERβ signaling and retinoic acid recep-
tor signaling, both of which are growth
promoting within granulosa cells. Pro-
motion of granulosa cell proliferation is
also achieved through cross-regulation
between the activin and Notch signaling
pathways. Notch signaling occurs
between the oocyte and granulosa cells,
as well as between adjacent
granulosa cells.

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


CELLULAR SIGNALING DURING PREANTRAL FOLLICULAR DEVELOPMENT 35
cell-secreted factors in the ovary (reviewed in Ref. [71]). results in infertility due to development of metastatic
In addition, we discuss the role of the juxtacrine Notch granulosa cell tumors by 3 months of age [145]. In con-
signaling pathway that is important for preantral follicu- trast, the double conditional knockout of Smad2/3 is sub-
lar growth, in addition to its roles during germ cell nest fertile, showing decreased numbers of antral follicles and
breakdown. Finally, we summarize recent findings on increased follicular atresia as indicated by a high inci-
the functions of the highly conserved Hedgehog signal- dence of ZP remnants in 3-month-old ovaries [146]. In
ing pathway in regulating the formation of the addition to follicular developmental aberrations, a high
thecal cells. incidence of trapped oocytes in luteinizing follicles and
a reduced expression of cumulus expansion markers
strongly suggest ovulatory defects in Smad2/3 conditional
knockout mice [146]. Granulosa cell conditional knockout
TGF-β Family Signal Transduction in the Ovary
of the coSMAD Smad4 results in similar phenotypes to the
Based on ligand sequence similarity and modes of sig- Smad2/3 conditional knockout, presenting with increased
nal transduction, the TGF-β superfamily is divided into numbers of atretic preantral follicles and ovulatory
two subgroups, the growth differentiation factors/bone defects [147].
morphogenetic proteins (GDFs/BMPs) and the acti-
vins/TGF-βs. TGF-β superfamily peptides form either Control of Follicular Growth by Oocyte-Secreted
homo- or hetero-dimeric ligands. Activins and TGF-βs GDF9 and BMP15
initiate signaling by binding to a serine/threonine kinase Immediately following activation, oocytes of primary
cell surface receptor, known as the type II receptor [138]. follicles begin to produce GDF9, a secreted TGF-β family
Five type-II receptors have been identified in mammals ligand [131,148,149]. Gdf9 null female mice are infertile
[139]. The type-II receptor recruits a type-I receptor, also due to a block in follicular development at the primary
known as an activin-like kinase (ALK), and activates the follicle stage, which appears to result from a failure of
type-1 receptor through trans-phosphorylation [138]. granulosa cell proliferation [131,150,151]. Demise of the
Seven type-1 receptors have been characterized in mam- follicles is initiated by degeneration of the oocytes in
mals [139]. In contrast, BMPs bind first to a type-I recep- Gdf9/ ovaries, which results in surviving follicular rem-
tor, followed by recruitment of a type-II receptor, or to a nants that acquire a luteinized morphology [131]. The
preformed type-1/type-2 receptor complex on the cell actions of GDF9 appear to be specifically directed toward
surface [140]. The signal is transduced intracellularly by somatic cells, since Gdf9/ oocytes can be successfully
type-1 receptor-mediated phosphorylation of a SMAD matured in vitro (with addition of the necessary factors)
protein (receptor-regulated SMAD -rSMAD), which will to resume meiosis [150]. In vivo treatment of PND5 rats
then recruit an obligatory common-mediator SMAD with GDF9 leads to increased activation of primordial fol-
(coSMAD) [141]. Receptor activation by GDFs/BMPs licles and subsequent enhancement of growth of primary
activates SMAD1, SMAD5, or SMAD8, while receptor follicles into multilayered preantral follicles [152]. Gdf9
activation by activins or TGF-β activates SMAD2 or stimulates proliferation in cultured granulosa cells, as
SMAD3 [142]. One exception to this model is the well as growth of cultured primary and secondary folli-
ligand GDF9, which utilizes SMAD2/3 in its signal cles [153,154]. The actions of GDF9 on granulosa cells
transduction [143,144] SMAD4 acts as a coSMAD for are most likely achieved through binding with a complex
receptor activation by both subtypes of ligands [142]. of the type-I receptor ALK5 [143,155] and the bone mor-
Finally, an rSMAD-coSMAD complex translocates to phogenetic protein receptor type II (BMPRII) [156] as dis-
the nucleus and acts as a transcriptional activator of tar- ruption of either leads to abrogation of the proliferative
get genes [141]. The SMAD pathway is negatively regu- effects of GDF9 on granulosa cells [143, 155, 156].
lated by inhibitory SMADs (SMAD6, SMAD7) through BMP15, a TGF-β family ligand highly related to GDF9,
prevention of rSMAD activation at the receptor level or was identified through a homology-based cloning strat-
through competitive binding of activated rSMAD to egy [157]. Like Gdf9, Bmp15 is expressed specifically in
coSMAD [141]. oocytes beginning at the primary follicle stage [157]
In granulosa cells, there appears to be significant func- and exerts proliferative effects on cultured rat granulosa
tional redundancies between SMAD1, 5, and 8, and cells [158]. Several naturally occurring mutations affect-
between SMAD2 and 3. This is demonstrated by the ing BMP15 signaling, and as a consequent ovulation rate,
absence of fertility defects following disruption of any have been characterized in sheep. The Inverdale (FecXI)
of these individual rSmad alleles in female mice mutation is a point mutation that leads to a single amino
[145,146]. Since conventional knockouts of all but Smad8 acid replacement in the mature Bmp15 protein, while a
result in embryonic lethality, a conditional knockout distinct point mutation known as the Hanna (FecXH)
approach has been taken to study these pathways in mutation leads to the creation of premature stop codon
the ovary. Double conditional knockout of Smad1/5 or tri- and thus a truncated Bmp15 protein [159]. Both the FecXI
ple conditional knockout of Smad1/5/8 in granulosa cells and FecXH alleles lead to nonfunctional Bmp15, and

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


36 2. REGULATION OF FOLLICLE FORMATION AND DEVELOPMENT BY OVARIAN SIGNALING PATHWAYS

homozygous females are infertile due to a block in follicle in the GDF9 transcriptional regulatory region [169] and
development at the primary stage [159]. Counterintui- point mutations that lead to reduced levels of mature
tively, ewes heterozygous for either mutation have an GDF9 or to aberrant mature GDF9 that is less potent in
increased ovulation rate, which leads to a higher inci- activating the SMAD2 pathway [170]. These findings sug-
dence of multiple births compared to wild-type ewes gest that the cumulative effects of BMP15 and GDF9 on
[159]. Consistent with the mitogenic role of Bmp15 in granulosa cell proliferation and gene expression are a
granulosa cell growth, antral follicles in heterozygous complex function of the two distinct rSmad pathways
FecXI and FecXH ewes are undersized; yet, the granulosa that they activate.
cells of these antral follicles have increased expression of
Lhcgr, rendering them more responsive to the Regulation of Granulosa Cell Proliferation by
LH-ovulatory signal [160]. Similarly, Booroola (FecB) ewes Granulosa Cell-Derived TGF-β Family Ligands
with a mutation that leads to reduced intracellular kinase In addition to GDF9 and BMP15 originating from the
activity of the type I BMPRIB (ALK6) receptor are hyper- oocyte, granulosa cells also produce multiple TGF-β fam-
prolific, with high rates of multiple births [161]. While the ily ligands that act in a paracrine manner to regulate fol-
precise molecular basis of the dependency of ovulation licular development. There are three mammalian TGF-β
rate on the level of BMP15 signaling remains unclear, it isoforms (TGF-β1–3), and localization studies have iden-
has been postulated that the determining factor may be tified their expression in all three follicular cell types
the ratio of GDF9 to BMP15 in the ovary [162]. beginning at the preantral or early-antral stages in human
Given the similarity in the structures of BMP15 and and rodent ovaries [171,172]. Depending on the species,
GDF9, as well as their expression by the oocyte in a tem- TGF-β1 can assert stimulatory or inhibitory effects on
porally overlapping manner, it is of interest to under- granulosa cell proliferation. While in vitro, TGF-β1 pro-
stand whether they assert their effects on granulosa motes rat granulosa cell proliferation, it exerts inhibitory
cells in any cooperative or redundant manner. When effect on the proliferation of granulosa cells from cattle,
coexpressed in human embryonic kidney 293T (293T) cells, sheep, and pig ovaries (reviewed in Ref. [171]). In con-
recombinant GDF9 and BMP15 are able to form heterodi- trast, negative regulation of thecal cell steroidogenesis
mers, suggesting their potential interaction in vivo by TGF-β1 appears to be conserved across these
[163,164]. In the mouse model, Bmp15/ females are sub- species [171].
fertile, in contrast to the infertile Gdf9/ females, present- Activins and inhibins are granulosa cell-secreted pep-
ing with only minimal morphological defects in follicular tide hormones that were originally identified as a part of
development, albeit having decreased ovulation and fertil- the ovarian feedback loop regulating the release of FSH
ization rates [165]. A more severe fertility defect was by the pituitary gland [71]. Mature activin and inhibin
observed in Bmp15/ Gdf9+/ females compared to are composed of dimers of structurally related peptides
Bmp15/ females [165]. However, Bmp15/ Gdf9/ connected by a disulfide bond [173]. Dimers of the two
double-knockout females have a fertility defect that is no β subunits form mature activins (Activin A—βAβA, Acti-
worse than that of the Gdf9/ single mutant [165]. In vin B—βBβB, or Activin AB—βAβB), while heterodimers of
cultured mouse granulosa cells, GDF9 and BMP15 were the α and β subunits result in mature inhibins (Inhibin
shown to act synergistically to induce proliferation and A—αβA or Inhibin B—αβB). Expression of all three sub-
SMAD3-dependent transcription [166]. The importance units can be detected in granulosa cells and is dynami-
of GDF9:BMP15 heterodimers in regulating granulosa cally regulated throughout the rat estrous cycle [48].
cell function was further demonstrated in a mouse cumulus Activin exerts its actions through binding to its type-II
cell expansion assay, where they were shown to be 10- to receptor (ACTRIIA or ACTRIIB) and transactivation of
30-fold more potent than the GDF9 homodimer in ALK2, 4, or 7, followed by activation of SMAD2/3 and
promoting cumulus expansion through activation of the the co-SMAD SMAD4 (reviewed in Ref. [174]). Inhibin
SMAD2/3 pathway [167]. suppresses the actions of activin through competitive
Several mutations at the BMP15 locus have been asso- binding with the type-II receptor. Finally, as previously
ciated with cases of primary ovarian insufficiency (POI) discussed, the ovary also produces FST, a molecule that
in humans [168]. Biochemical investigations into ten of binds to and neutralizes the biological activity of
these POI-associated BMP15 mutations showed that they activin [175].
resulted in reduced levels of mature BMP15, a decreased In vitro, activin exerts proliferative effects on granulosa
ability of BMP15 to induce SMAD1/5/8-dependent tran- cells of preantral follicles isolated from prepubertal mice,
scription, or a reduced capacity for BMP15 to synergisti- and FST treatment blocks this effect [176]. The effects of
cally induce proliferation and SMAD3-dependent activin on granulosa cell proliferation appear to be age
transcription in the presence of GDF9 [168]. Similarly, dependent, possibly due to additional growth signals
mutations in the GDF9 locus have also been identified by FSH following the onset of puberty. For example, acti-
in cases of POI, including those that lead to aberrations vin treatment of neonatal mice leads to increased

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


CELLULAR SIGNALING DURING PREANTRAL FOLLICULAR DEVELOPMENT 37
proliferation of granulosa cells and to a limited degree the granulosa cells, this study further illustrates the exten-
germ cells [21]. However, while activin stimulates growth sive network of signals with which activin interacts
of preantral follicles isolated from juvenile mice, it during gonadotropin-independent follicle growth.
promotes atresia and abrogates FSH-induced growth in
preantral follicles isolated from adult animals [177,178].
Regardless of this age-specific effect, activin appears to
The Role of Notch Signaling in Preantral Follicle
be necessary for female fertility, as animals lacking any
ovarian expression of either of the β subunits are infertile,
Development
showing decreased expression of factors that are associ- Earlier, we reviewed evidence for a role of Notch sig-
ated with granulosa cell survival and proliferation such naling in promoting germ cell nest breakdown. It is now
as Kitl, Taf4b, and c-Myc [179]. Consistent with a defi- appreciated that Notch signaling continues to be impor-
ciency in activin signaling, juvenile Smad3/ ovaries tant during follicle growth and development, functioning
have decreased numbers of preantral and early antral to promote granulosa cell proliferation and to coordinate
follicles, indicating disrupted follicular growth from the the development of the oocyte and granulosa cells
secondary stage and beyond [180]. (Fig. 5). Inhibition of Notch signaling using the
Overexpression of the inhibin α subunit in transgenic γ-secretase inhibitors, DAPT or L658,458, leads to
mice (MT-alpha) results in increased levels of circulating decreased proliferation of cultured granulosa cells or sec-
mature inhibins and decreased levels of mature activins ondary follicles [189]. Expression of c-Myc, a
[181]. Expression of both β subunits is downregulated proliferation-associated transcription factor, in granulosa
in the MT-alpha ovaries, suggesting a certain degree of cells is suppressed following Notch inhibition, and resto-
autoregulation of gene expression by activin signaling ration of Notch signaling through expression of constitu-
[181]. Consistent with pharmacological and genetic tively active N2ICD leads to recovery of both c-Myc
models of suppressed activin action discussed above, expression and granulosa cell proliferation [189]. Addi-
MT-alpha female mice are subfertile with decreased num- tionally, Notch may also regulate granulosa cell prolifer-
bers of mature antral follicles in the ovary [181,182]. ation through interactions with activin signaling. There is
While inhibin is not currently known to activate any spe- evidence that the Notch target gene Hey2 is coregulated
cific receptor, the genetic model of inhibin deficiency by activin in a SMAD3-dependent manner in preantral
Inha/ has demonstrated the necessity for its presence granulosa cells [190]. Indeed, activin treatment is able
in order to restrain and balance the mitogenic effects of to overcome the effect of DAPT inhibition and restore
activin on granulosa cells [183]. Inha/ ovaries are proliferation in cultured granulosa cells [190].
abnormally large compared to the wild-type littermates In conditional-knockout models of the ligand JAG1
due to granulosa cell hyperproliferation, which eventu- from the oocytes (J1KO) or the receptor NOTCH2 from
ally leads to gonadal tumors and infertility in granulosa cells (N2KO), defects in proliferation of granu-
adulthood [183]. losa cells are first observed by PND19, and are associated
Studies in cultured granulosa cells from preantral with enhanced cell death [74]. J1KO and N2KO ovaries
follicles have revealed the potential for cross-talk also present with a phenotype that indicates disruption
between activin and at least two different types of of developmental coordination between oocytes and
nuclear receptor signaling pathways in regulating gran- granulosa cells, characterized by enlarged oocytes that
ulosa cell proliferation (Fig. 5). Activin treatment is are contained in very early stage growing follicles (i.e.,
found to induce expression of Esr1 and Esr2 in a follicles with a single layer of granulosa cells) [74].
SMAD2/3-dependent manner in proliferating granu- Molecularly, this is supported by elevated expression of
losa cells [184]. Since estrogen is mitogenic, as well as the oocyte-specific factors Gdf9, Figla, and Zp3 as com-
pro-survival, in preantral granulosa cells [185–187], it pared to wild-type littermates [74].
is likely that activin promotes proliferation at least in The Notch target gene Hes1 is expressed in somatic
part through enhancement of estrogen receptor signal- cells embryonically (by E15.5) and its expression
ing. Transcriptomic analysis of activin-treated granu- decreases following birth [71,191]. Conventional, or gran-
losa cells identified Cyp26b1 as downregulated by ulosa cell conditional, knockouts of Hes1 lead not only to
activin [188]. A member of the cytochrome P450 family, increased oocyte death but also increased proliferation of
the gene product of Cyp26b1 functions to degrade RA the pregranulosa cells [191]. Both mouse lines are subfer-
and is expressed in granulosa cells of growing follicles tile, indicating that disruption of early follicular develop-
[188]. RA or Cyp26b1 inhibitor treatments of cultured ment results in detrimental effects on fecundity.
granulosa cells leads to an increase in granulosa cell Increased expression of the βB subunit (Inhbb) of activin
proliferation in a dose-dependent manner, while inhibi- is observed in both models of Hes1 deficiency [191],
tion of retinoic acid receptors abrogates this effect [188]. which makes the enhanced pregranulosa cell prolifera-
Besides identifying a novel mitogenic factor in tion consistent with effects observed in neonatal mice

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


38 2. REGULATION OF FOLLICLE FORMATION AND DEVELOPMENT BY OVARIAN SIGNALING PATHWAYS

treated with exogenous activin [21]. An additional com- follicular development at the preantral stage and blunted
mon phenotype between the two models of Hes1 defi- steroid production [194]. It has been shown that GDF9
ciency is the suppression of c-Kit expression in oocytes from the oocyte stimulates the expression of Dhh and
[191]. As covered in earlier sections, KITL/c-KIT signal- Ihh in granulosa cells, and indeed, expression of both
ing is particularly important for direct communication Hh ligands is severely downregulated in Gdf9/ ovaries
between granulosa cells and oocytes, and it regulates nest [194]. Theca layer recruitment and differentiation is an
breakdown and primordial follicle activation in the neo- elegant example of how three distinct cell types within
natal ovary. Thus, while Notch signaling is mitogenic in the follicle communicate and regulate each other’s func-
preantral granulosa cells, an additional function is per- tions through diverse signaling pathways.
haps to ensure coordinated development between
oocytes and granulosa cells prior to exposure to gonado-
tropins, which will act as the master regulators for follic- REGULATION OF GRANULOSA CELL
ular development beyond the preantral stage. PROLIFERATION AND
DIFFERENTIATION IN ANTRAL AND
PREOVULATORY FOLLICLES
Molecular Mechanisms of Theca Cell
In larger multilayer follicles, small fluid filled spaces
Recruitment and Specification are initiated that will eventually converge to form the
The thecal cell layer forms just outside the basement antral cavity [198]. A hallmark of antral follicle develop-
membrane surrounding the outermost granulosa cells ment is the increased abundance of transcripts for the
when the follicle reaches the secondary stage [192]. FSH receptor (Fshr) within the granulosa cells, which
Through the use of transgenic reporter mouse lines and upon translation into FSHR renders the follicles respon-
lineage tracing, it was shown that theca cells originate sive to circulating FSH [199]. Indeed, FSHR signaling is
from one of two sources, the fibroblast-like precursor cells required for continued development beyond the multi-
indigenous to the ovary (Wt1 expressing cells) or mesen- layered secondary follicle stage, as antral follicles are
chymal cells that migrate into the ovary from the meso- never detected in mice lacking either mature FSH
nephros [193,194]. Theca cells function in ovarian (Fshb/ line) [135] or FSHR [136,137]. Following puberty,
steroidogenesis by providing aromatizable androstenedi- FSH functions to prevent early antral follicles from under-
one to the granulosa cells, where aromatase will convert going atresia [200] by regulating both granulosa cell
the androgen into 17β-estradiol [192]. It was long sus- proliferation and differentiation [201,202]. Under the
pected that preantral follicles secrete certain thecal cell influence of FSH, the cell cycle of granulosa cells is short-
differentiating factors, as conditioned medium collected ened, requiring a doubling period of only about 24 h as
from theca-less preantral follicles stimulated androstene- compared to requiring greater than 7 d during the FSH-
dione production in cultured thecal-interstitial cells [195]. independent period [203]. FSH also functions to induce
This putative theca differentiating factor was thought to gene expression changes in granulosa cells, which differ-
be a small molecule on the order of 20–25 kDa in entiates them toward the LH responsive, preovulatory
size [196]. stage [204,205]. Characteristics of granulosa cell matura-
Recent studies indicate that the developmentally con- tion in response to FSH signaling include the expression
served Hedgehog (Hh) signaling pathway is the chief reg- of genes involved in steroid biosynthesis, such as
ulator of thecal cell specification in the ovary. Hedgehog Cyp19a1, Cyp11a1, and Hsd3b1, which leads to increased
signaling is initiated by binding of one of three HH production of estradiol and progesterone, and the expres-
ligands (Sonic hedgehog—SHH, Indian hedgehog— sion of the membrane receptor for LH (Lhcgr) [204,206].
IHH, Desert hedgehog—DHH) to the canonical receptor The FSHR is a G-protein-coupled receptor (GPCR) that
Patched (PTCH1, 2). Hh binding to PTCH results in dere- stimulates synthesis of the second messenger cyclic AMP
pression of Smoothened (SMO2), an obligate signal trans- (cAMP) when activated [207,208]. The intracellular rise in
ducer in Hh signaling, and finally the translocation of the cAMP leads to activation of the cAMP-dependent protein
Hh transcriptional effector GLI (GLI1 in thecal cell pre- kinase A (PKA) [209], which in turn promotes gene tran-
cursors) into the nucleus. Ihh and Dhh are expressed by scription through phosphorylation and activation of sev-
granulosa cells of growing follicles, while the receptors eral transcription factors, including but not limited to, the
Ptch1, Ptch2, and the transcriptional effector Gli1 are only cAMP response element-binding protein (CREB)
detected in the surrounding pretheca cells, suggesting [210,211] and GATA4 [212,213]. Additionally, PKA
that theca cells are the exclusive target of Hh ligands in downstream of FSHR activation also modifies chromatin
the ovary (Fig. 5) [197]. structure through phosphorylation of histone H3, which
The thecal layer never forms in ovaries with condi- leads to increased nucleosome accessibility [214,215]. It is
tional deletion of both Dhh and Ihh from the granulosa now appreciated that to exert maximum mitogenic and
cells [194]. These animals are infertile, with blocked differentiation effects, FSH/FSHR signaling interacts

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


REGULATION OF GRANULOSA CELL PROLIFERATION AND DIFFERENTIATION IN ANTRAL AND PREOVULATORY FOLLICLES 39
and cooperates with diverse local ovarian signals. In this processes, including proliferation and gene transcription
section, we discuss intraovarian signals that regulate pro- (Fig. 6) [217]. While originally described as an intracellu-
liferation and differentiation in antral to preovulatory lar signaling pathway that is downstream of RTKs, the
granulosa cells and their cross talk with FSHR signaling. MAPK pathway is now also known to function down-
For the purposes of clarity and simplicity, we will refer to stream of GPCR activation [218]. The MAPK/ERK path-
granulosa cells as undifferentiated prior exposure to FSH way is active in proliferating cells, including granulosa
(including those collected from estrogen stimulated cells. ERK1/2 activation is observed in undifferentiated
animals and used in vitro studies) and as differentiating proliferating granulosa cells in culture [219]. An increase
following activation of FSHR, while appreciating that in intracellular cAMP levels was found to prevent activa-
these are points along a continuum [216]. tion of ERK2 in response to growth factors in human arte-
rial smooth muscle cells, adipocytes, and mouse
fibroblast cell lines [220]. Based on these findings, FSHR
Crosstalk Between FSHR Signaling and Kinase activation and its associated rise in cAMP levels were ini-
Cascades in Granulosa Cell Proliferation and tially hypothesized to downregulate ERK1/2 activation
in differentiating granulosa cells. On the contrary, FSH
Differentiation
stimulation of rat granulosa cells in culture leads to an
Control of Granulosa Cell Proliferation and Gene acute activation of ERK1/2, which occurs in a cAMP-
Expression by MAPK/ERK Activation Downstream dependent manner [219]. In addition to proliferation,
of FSHR Signaling activation of the MAPK/ERK pathway following FSH
The mitogen-activated protein kinases (MAPKs) are a stimulation is also important for transcription of genes
highly conserved family of serine/threonine protein associated with the preovulatory phenotype, including
kinases that function to relay extracellular cues by way positive regulation of the Cyp19a1, Cyp11a1, Inha, Lhcgr,
of kinase cascade activation and influence diverse cellular and Egfr genes [221].

FIG. 6 Integration of FSH and paracrine signals in promoting granulosa cell proliferation and differentiation. Following antrum formation, gran-
ulosa cells proliferate and differentiate into the preovulatory stage through upregulation of Lhcgr and genes important for steroidogenesis in
response to FSH. To achieve the maximal proliferative and differentiation effects of FSH, FSHR signaling interacts with kinase cascades downstream
of RTKs such as EGFR and IGFR and cooperates with Smad-dependent signaling pathways downstream of TGFβ and activin family receptors. Inte-
gration of PKA and PI3K/AKT signaling leads to inactivation of the transcriptional repressor FOXO1 through phosphorylation, which targets it for
degradation. In turn, MAPK/ERK and SMAD2/3 activation provide activating signals for the transcription of granulosa cell differentiation-
associated genes. Notch signaling promotes granulosa cell differentiation through positive regulation of steroid biosynthetic enzyme gene expression
and through suppression of proliferative kinase cascades. Additionally, the Notch NICD is known to coregulate expression of common effectors by
interacting with the SMAD complex. Note that distinct subsets of transcriptional regulatory factors will interact at the promoters of different target
genes, and the schematic illustrates generic and summed examples of such regulatory pathway interactions.

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


40 2. REGULATION OF FOLLICLE FORMATION AND DEVELOPMENT BY OVARIAN SIGNALING PATHWAYS

It is still debated whether any specific RTK is involved observation that the ligand insulin-like growth factor-1
in MAPK/ERK pathway activation in FSH-stimulated (Igf1) and Fshr are coexpressed in healthy, steroidogeni-
granulosa cells. In undifferentiated, proliferating granu- cally active large antral follicles from mouse [228]. Igf1
losa cells from estrogen-treated mice, the MAPK pathway null females are infertile, with ovaries lacking follicles
upstream of ERK1/2 is observed to be constitutively beyond the early antral stage, resembling Fshb/ animals
active [222]. In these granulosa cells, ERK1/2 is continu- [228]. In granulosa cells, activation of the IGF1-R or the
ously dephosphorylated by the MAP kinase phosphatase FSHR independently stimulates the PI3K/AKT pathway
3 (MKP3), thus restraining the full activation of the (Fig. 6), and coactivation of both receptors leads to syner-
MAPK/ERK pathway [221]. FSH stimulation and PKA gistic effects on AKT activation [229]. Indeed, inhibition
activation were in turn shown to inhibit MKP3 activity, of the IGF1-R prevents the induction of granulosa cell dif-
and thus function to relieve the inhibition on ERK1/2 ferentiation by FSH, as measured by the lack of an
[221]. Gene expression changes are in turn dependent increase in Cyp19a1 expression and estradiol production
upon phosphorylation of the RNA and DNA binding [229]. Additional support for the importance of IGF1-R
protein YB-1 by the activated ERK1/2 [220]. While epi- signaling in facilitating follicular maturation by FSH
dermal growth factor receptor (EGFR) functions promi- comes from the recently reported Igf1r granulosa cell con-
nently in propagating the LH signal during ovulation, ditional knockout (IGF1Rgcko) mouse line [230].
there is also evidence for its modulation downstream of IGF1Rgcko females are infertile with ovaries lacking antral
FSHR signaling. Radioactively labeled epidermal growth follicles and granulosa cells that are unable to differenti-
factor (EGF) is observed to bind to immature rat follicles, ate in response to pregnant mare serum gonadotropin
and FSH treatment enhances the number of EGF mole- (PMSG) stimulation, which activates the FSHR, despite
cules bound, suggesting modulation of Egfr expression normal levels of Fshr expression compared to wild-type
by FSH in differentiating granulosa cells [223]. More littermates [230].
recently, Egfr expression was shown to be strongly down- Another important interaction between FSHR signal-
regulated in Fshb/ prepubertal ovaries [224]. Treatment ing and a paracrine factor occurs in the regulation of gran-
with FSH was shown to restore Egfr expression (and ulosa cell proliferation by activin. In cultured rat
eventually the ability to ovulate), confirming modulation granulosa cells, the full mitogenic effect of FSH requires
of Egfr by FSHR signaling. In addition to MAPK/ERK the presence of activin [231]. The apparent explanation
activation downstream of FSHR, a known, but minor, is that activation of both AKT and SMAD2/3 is required
product of Fshr alternative splicing (growth factor type1 for expression of the cell cycle regulator Cyclin D2 (Ccnd2)
receptor or oFSH-R3) whose activation rapidly leads to following FSH and activin stimulation (Fig. 6) [231]. It is
phosphorylation of ERK1/2 instead of the rise in cAMP now appreciated that FSH activation of the PI3K/AKT
level can also be detected in the ovary [225]. pathway occurs through a PKA-dependent cascade
Through the use of the TNR mouse line, Notch signal- [232] and targets the transcriptional suppressor Foxo1
ing is observed to remain active in granulosa cells of [233]. Phosphorylation of Foxo1 leads to its translocation
antral and preovulatory follicles [226]. Notch signaling from the nucleus and directs it for degradation. More
molecules are expressed in these later stage follicles than 60% of FSH-regulated genes have now been identi-
and there is evidence that their expression is dynamically fied to be targets for Foxo1, including those involved in
regulated by gonadotropins [72]. Of note, is the ligand proliferation, such as Ccnd2, and those involved in steroid
Jag1 whose localization shifts from being oocyte-specific biosynthesis, such as Star, Cyp11a1, and Cyp19a1
to being present in ovarian somatic cells following expo- [233,234]. Fig. 6 summarizes the integration of FSHR,
sure to exogenous gonadotropins [226,227]. Disruption of RTKs, Notch, and activin signaling pathways in regulat-
the Notch ligand Jag1 in cultured granulosa cells collected ing granulosa cell proliferation and differentiation. These
from PMSG-primed immature mice leads to a compro- data demonstrate the importance of, what are often
mised differentiation response, as measured by suppres- accepted to be ubiquitous, kinase cascades in integrating
sed steroidogenesis [226]. At the expense of suppressed endocrine and paracrine cues to promote the differenti-
differentiation, enhanced proliferation, resembling that of ated preovulatory phenotype in granulosa cells.
less mature granulosa cells, is maintained in Jag1-deficient
cells and this is associated with to increased activation of
the MAPK/ERK pathway. Together, these studies demon- The Roles of Steroid Hormones in Follicular
strate that in differentiating granulosa cells, FSH functions Maturation
to promote proliferation and differentiation in conjunction
with underlying signals that are initiated by local factors. The ovarian follicle is the main source of sex steroid
hormones (estrogens, progestins, and androgens) that
Regulation of Granulosa Cell Differentiation and are indispensable for overall female reproductive physi-
Proliferation by the PI3K/AKT Pathway ology. Sex steroids act on many organ systems, including
Interest in insulin growth factor receptor (IGFR), a but not limited to the nervous system, the skeletal system,
RTK, signaling in follicle maturation began with the the cardiovascular system, metabolism, and last but not
I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS
REGULATION OF GRANULOSA CELL PROLIFERATION AND DIFFERENTIATION IN ANTRAL AND PREOVULATORY FOLLICLES 41
least the reproductive system. Comprehensive discus- ER-β (ESR2) is localized almost exclusively to granulosa
sions of the molecular mechanism of ovarian steroido- cells, and its expression is dynamically regulated by
genesis are found elsewhere in this text and reviewed gonadotropins [41,244]. Thus, models of Esr2 disruption
in Ref. [235]. The two-cell, two-gonadotropin model of have allowed for further understanding of the functions
follicular steroidogenesis [236] is based on localization of estrogen in follicular development.
of the enzymes required for distinct steps in the steroid ER-β-knockout mice are subfertile with ovaries show-
biosynthetic cascade. Theca cells convert cholesterol into ing a block in progression from early antral follicles to the
androstenedione under the influence of Lhcgr signaling. large antral and preovulatory stages [245,246]. Granulosa
In turn, the hydrolyzable androstenedione is utilized cells of ER-β-knockout follicles show attenuated
by granulosa cells as a substrate for estradiol under the responses to the differentiating effect of FSH, including
influence of FSHR signaling. Following ovulation and suboptimal induction of Cyp19a1—therefore reduced
formation of the corpus luteum, progesterone will become steroidogenesis—and Lhcgr expression [246]. As a conse-
the main sex steroid produced by the ovary. In this section, quent of decreased expression of Lhcgr, ER-β-knockout
our focus will be on the role of estrogens and androgens follicles are less responsive to ovulatory LH signals as
as paracrine factors regulating follicular events throughout compared to wild-type controls [247]. These findings sug-
the FSH-dependent and periovulatory-developmental gest that ER-β signaling is important to promote FSH-
stages. While we recognize the equally important role of regulated granulosa cell differentiation and promotion
progesterone, we refer readers to Ref. [237] for a discussion of the preovulatory phenotype. Interestingly, ovarian
on the importance of progesterone receptor signaling phenotypes of double-knockout Esr1 and Esr2 mice
during the ovulatory cascade and pregnancy. (αβERKO) are completely distinct from either single
knockout. αβERKO females are infertile with defective
ovarian follicles that appear to transdifferentiate into
Estrogen and Estrogen Receptor Signaling in Antral
seminiferous tubule-like structures in adulthood [248].
and Preovulatory Follicles
Estrogen increases granulosa cell proliferation in pre-
antral follicles [185–187]. Estrogen is also found to Physiological and Supraphysiological Effects of
increase the presence of FSHR and LHCGR in granulosa Androgens on Follicular Development
cells and enhances the rise in cAMP following cholera While initially thought to function solely as the sub-
toxin stimulation (due to blocked hydrolysis of strate for estrogen synthesis in the ovary [236], the impor-
G-protein-associated GTP, and thus constitutively acti- tance of androgens and androgen receptor (AR) signaling
vated adenylate cyclase), suggesting its ability to aug- in follicular development has now been demonstrated
ment GPCR/cAMP signaling [238]. Ovaries of mice through the generation of granulosa cell-specific knock-
lacking aromatase (Cyp19a1; ArKO), an enzyme that is outs of the AR in mice (ARcKO) [249,250]. Two indepen-
required for the final conversion of androgens into estra- dently generated ARcKO lines are subfertile, with
diol, lack follicles beyond the early antral stages [187]. decreased follicular growth and increased follicular atre-
The phenotypes of the ArKO mouse, however, appear sia observed starting at the preantral stage [249,250].
to be caused by chronic elevated serum gonadotropin Testosterone and dihydrotestosterone (DHT) were
levels due to the lack of estrogen feedback to the pitui- shown to prevent granulosa cell apoptosis and folli-
tary. Upon exogenous estrogen treatment, follicular cular atresia by increasing the expression of miR-125b, a
development is able to resume and ovulation is known suppressor of proapoptotic proteins [251]. Also
restored [187]. in FSH-responsive follicles, androgen increases the level
In the ovary, estrogen exerts its mitogenic and differ- of granulosa cell Fshr mRNA, thus enhancing the FSH
entiative effects through activation of one of two recep- stimulation of follicle growth [251]. Testosterone or DHT
tors (ER-α or ER-β) belonging to the nuclear receptor was also shown to augment FSH-stimulated granulosa
family of transcription factors. While G protein-coupled cell expression of Star [252].
receptor 30 (Gpr30), one of many membrane-bound Clinically, ovarian hyperandrogenism is a feature of
forms of estrogen receptor, can be detected in the mam- the relatively common endocrine and metabolic disorder
malian ovary [239], Gpr30-deficient mice are completely polycystic ovarian syndrome (PCOS), which often leads
fertile [240]. Following ligand binding in the cytoplasm, to anovulation [253,254]. At supraphysiological levels,
ERs dimerize and translocate into the nucleus to activate DHT treatment of prepubertal rats leads to a block in fol-
gene transcription upon recognition of an estrogen- licular development at the early antral stage [255]. The
responsive element (ERE) [241]. Esr1 (ER-α) is expressed abnormally large population of small growing follicles,
throughout the female reproductive tract, including in where dominant follicle(s) fail to emerge, resemble that
the thecal and interstitial cells of the ovary [242]. The of PCOS ovaries where these small follicles localize to
Esr1-knockout mouse is infertile due to reproductive tract the ovarian cortex and resemble cysts in diagnostic ultra-
defects, including thin uteri, lack of preovulatory follicles, sound examination. High levels of DHT were shown to
and hemorrhagic cysts in the ovary [242,243]. In contrast, inhibit FSH-induced granulosa cell proliferation through
I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS
42 2. REGULATION OF FOLLICLE FORMATION AND DEVELOPMENT BY OVARIAN SIGNALING PATHWAYS

dampening of PI3K/AKT pathway activation and Cyclin follicles develop will continue to allow for the identifica-
D1 expression [255]. Excess DHT was also shown to tion of the genetic and molecular basis of reproductive
inhibit proliferation by preventing insulin- and FSH- disorders, including those of often unknown etiology.
stimulated activation of the MAPK/ERK and Cyclin D2 Where relevant, we present examples of clinical findings
expression in cultured granulosa cells [256,257]. These that were informed by these basic studies. The increasing
findings demonstrate that at both physiological and sensitivity and affordability of genome-wide analysis has
supraphysiological levels, androgens influence antral fol- made possible the identification of genetic polymor-
licular development by modulating both the proliferative phisms in complex, often poorly understood diseases,
and differentiating effects of FSHR signaling. including idiopathic cases of infertility. Researchers and
clinicians continue to identify novel infertility-associated
loci, making possible mechanistic studies that are often
Conclusions informed by an understanding of the diverse signaling
pathways discussed here. It is an exciting era of discov-
It is abundantly clear that intraovarian signaling path- ery, with robust opportunities to continue to increase
ways are critical for regulating follicular function during our understanding of the ovary and its central role in
both the gonadotropin-independent and -dependent female reproduction.
stages of follicle development. Through this review, we
hope to have communicated the diversity by which cells
of the follicle communicate with each other to ensure SUPPORTING GRANTS
coordinated development. It is perhaps not surprising
that reproduction, a necessarily robust process to ensure NIH/NICDH P01 HD021921
the continuation of species, requires highly interacting, NIH/NIGMS T32 GM008061
and often overlapping, complex signaling mechanisms.
Disruption of early developmental events that are
gonadotropin-autonomous, such as follicle formation or
control of primordial follicular activation, can directly DISCLOSURE SUMMARY
determine fertility status. However, FSH and LH alone
are also not sufficient to support fertility, as their actions The authors have nothing to disclose
are highly dependent upon interactions with underlying
paracrine factors. It is remarkable how diverse these
intrinsic ovarian regulators are, spanning the gamut of References
evolutionarily conserved developmental pathways such [1] Ginsburg M, Snow MH, McLaren A. Primordial germ cells in the
as Notch and Hedgehog signaling, the large TGF-β family mouse embryo during gastrulation. Development 1990;110
of ligands, many RTKs, and nuclear receptor signaling. (2):521–8.
The advent of transgenic overexpression and reporter [2] Liu C-F, Liu C, Yao HHC. Building pathways for ovary organogen-
esis in the mouse embryo. Curr Top Dev Biol 2010;90:263–90.
mice, as well as conditional deletion strategies using the
[3] Anderson EL, Baltus AE, Roepers-Gajadien HL, Hassold TJ, de
Cre/LoxP system in the mouse, has allowed for the discov- Rooij DG, van Pelt AMM, et al. Stra8 and its inducer, retinoic acid,
ery of molecular mechanisms that contribute to specific regulate meiotic initiation in both spermatogenesis and oogenesis
periods of follicular development. In the coming years, in mice. Proc Natl Acad Sci U S A 2008;105(39):14976–80.
we expect that the maturation of CRISPR/Cas9 genome [4] Koubova J, Menke DB, Zhou Q, Capel B, Griswold MD, Page DC.
Retinoic acid regulates sex-specific timing of meiotic initiation in
editing technology will extend our knowledge of molec-
mice. Proc Natl Acad Sci U S A 2006;103(8):2474–9.
ular endocrinology, and perhaps allow relevant ques- [5] Bowles J, Knight D, Smith C, Wilhelm D, Richman J, Mamiya S,
tions to be asked in mammals beyond rodents [258]. et al. Retinoid signaling determines germ cell fate in mice. Am
Moreover, CRISPR/Cas9 technology promises to Assoc Adv Sci 2006;312(5773):596–600.
increase the efficiency—with respect to timing and [6] Dokshin GA, Baltus AE, Eppig JJ, Page DC. Oocyte differentiation
is genetically dissociable from meiosis in mice. Nat Genet 2013;45
precision—of generation of transgenic and knockout
(8):877–83.
models. In combination with predictive in silico analysis [7] Pepling ME, Spradling AC. Mouse ovarian germ cell cysts undergo
and high-throughput-omics studies, we may soon iden- programmed breakdown to form primordial follicles. Dev Biol
tify molecular regulators of poorly understood aspects 2001;234(2):339–51.
of follicular development, such as follicle formation and [8] Auersperg N, Wong AS, Choi KC, Kang SK, Leung PC. Ovarian
surface epithelium: biology, endocrinology, and pathology. Endocr
primordial follicle activation.
Rev 2001 Apr;22(2):255–88.
Most findings presented in this chapter represent basic [9] Byskov AG, Lintern-Moore S. Follicle formation in the immature
research conducted largely in murine models. We posit mouse ovary: the role of the rete ovarii. J Anat 1973;116
that understanding the molecular basis of how ovarian (Pt 2):207–17.

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


REFERENCES 43
[10] Zheng W, Zhang H, Gorre N, Risal S, Shen Y, Liu K. Two classes of [31] Flaws JA, Hirshfield AN, Hewitt JA, Babus JK, Furth PA. Effect of
ovarian primordial follicles exhibit distinct developmental dynam- bcl-2 on the primordial follicle endowment in the mouse ovary. Biol
ics and physiological functions. Hum Mol Genet 2014;23(4):920–8. Reprod 2001;64(4):1153–9.
[11] Mork L, Maatouk DM, McMahon JA, Guo JJ, Zhang P, [32] Bergeron L, Perez GI, Macdonald G, Shi L, Sun Y, Jurisicova A, et al.
McMahon AP, et al. Temporal differences in granulosa cell specifi- Defects in regulation of apoptosis in caspase-2-deficient mice.
cation in the ovary reflect distinct follicle fates in mice. Biol Reprod Genes Dev 1998;12(9):1304–14.
2012;86(2):37. [33] Montano MM, Welshons WV. Saal vom FS. Free estradiol in serum
[12] Hilscher B, Hilscher W, B€ ulthoff-Ohnolz B, Kr€amer U, Birke A, and brain uptake of estradiol during fetal and neonatal sexual dif-
Pelzer H, et al. Kinetics of gametogenesis. I. Comparative histolog- ferentiation in female rats. Biol Reprod 1995;53(5):1198–207.
ical and autoradiographic studies of oocytes and transitional pros- [34] Weisz J, Ward IL. Plasma testosterone and progesterone titers of
permatogonia during oogenesis and prespermatogenesis. Cell pregnant rats, their male and female fetuses, and neonatal off-
Tissue Res 1974;154(4):443–70. spring. Endocrinology 1980;106(1):306–16.
[13] Pepling ME, Spradling AC. Female mouse germ cells form synchro- [35] Nilsson EE, Skinner MK. Progesterone regulation of primordial fol-
nously dividing cysts. Development 1998 Sep;125(17):3323–8. licle assembly in bovine fetal ovaries. Mol Cell Endocrinol 2009;313
[14] Mork L, Tang H, Batchvarov I, Capel B. Mouse germ cell clusters (1–2):9–16.
form by aggregation as well as clonal divisions. Mech Dev 2012 [36] Fortune JE, Yang MY, Muruvi W. In vitro and in vivo regulation of
Jan;128(11–12):591–6. follicular formation and activation in cattle. Reprod Fertil Dev
[15] Lei L, Spradling AC. Mouse primordial germ cells produce cysts 2011;23(1):15–22.
that partially fragment prior to meiosis. Development 2013;140 [37] Thau R, Lanman JT, Brinson A. Declining plasma progesterone
(10):2075–81. concentration with advancing gestation in blood from umbilical
[16] B€uning J. The insect ovary. Springer Science & Business Media; and uterine veins and fetal heart in monkeys. Biol Reprod
2012. 1 p. 1976;14(4):507–9.
[17] de Cuevas M, Lilly MA, Spradling AC. Germline cyst formation in [38] Kezele P, Skinner MK. Regulation of ovarian primordial follicle
Drosophila. Annu Rev Genet 1997;31(1):405–28. assembly and development by estrogen and progesterone: endo-
[18] Lei L, Spradling AC. Mouse oocytes differentiate through organelle crine model of follicle assembly. Endocrinology 2003;144
enrichment from sister cyst germ cells. Science 2016;352(6281):95–9. (8):3329–37.
[19] Pepling ME, Wilhelm JE, O’Hara AL, Gephardt GW, Spradling AC. [39] Chen Y, Jefferson WN, Newbold RR, Padilla-Banks E, Pepling ME.
Mouse oocytes within germ cell cysts and primordial follicles Estradiol, progesterone, and genistein inhibit oocyte nest break-
contain a Balbiani body. Proc Natl Acad Sci U S A 2007;104 down and primordial follicle assembly in the neonatal mouse
(1):187–92. ovary in vitro and in vivo. Endocrinology 2007;148(8):3580–90.
[20] Greenbaum MP, Iwamori N, Agno JE, Matzuk MM. Mouse TEX14 [40] Chen Y, Breen K, Pepling ME. Estrogen can signal through multiple
is required for embryonic germ cell intercellular bridges but not pathways to regulate oocyte cyst breakdown and primordial folli-
female fertility. Biol Reprod 2009;80(3):449–57. cle assembly in the neonatal mouse ovary. J Endocrinol 2009;202
[21] Bristol-Gould SK, Kreeger PK, Selkirk CG, Kilen SM, Cook RW, (3):407–17.
Kipp JL, et al. Postnatal regulation of germ cells by activin: the estab- [41] Jefferson WN, Couse JF, Banks EP, Korach KS, Newbold RR.
lishment of the initial follicle pool. Dev Biol 2006;298(1):132–48. Expression of estrogen receptor beta is developmentally regulated
[22] Morita Y, Tilly JL. Oocyte apoptosis: like sand through an hour- in reproductive tissues of male and female mice. Biol Reprod
glass. Dev Biol 1999;213(1):1–17. 2000;62(2):310–7.
[23] Schmidt D, Ovitt CE, Anlag K, Fehsenfeld S, Gredsted L, Treier A-C, [42] Iguchi T, Fukazawa Y, Uesugi Y, Takasugi N. Polyovular follicles
et al. The murine winged-helix transcription factor Foxl2 is required in mouse ovaries exposed neonatally to diethylstilbestrol in vivo
for granulosa cell differentiation and ovary maintenance. and in vitro. Biol Reprod 1990;43(3):478–84.
Development 2004;131(4):933–42. [43] Kim H, Nakajima T, Hayashi S, Chambon P, Watanabe H, Iguchi T,
[24] Uda M, Ottolenghi C, Crisponi L, Garcia JE, Deiana M, Kimber W, et al. Effects of diethylstilbestrol on programmed oocyte death and
et al. Foxl2 disruption causes mouse ovarian failure by pervasive induction of polyovular follicles in neonatal mouse ovaries. Biol
blockage of follicle development. Hum Mol Genet 2004;13 Reprod 2009;81(5):1002–9.
(11):1171–81. [44] Jefferson WN, Couse JF, Padilla-Banks E, Korach KS, Newbold RR.
[25] Liang L, Soyal SM, Dean J. FIGalpha, a germ cell specific transcrip- Neonatal exposure to genistein induces estrogen receptor (ER)
tion factor involved in the coordinate expression of the zona pellu- alpha expression and multioocyte follicles in the maturing mouse
cida genes. Development 1997;124(24):4939–47. ovary: evidence for ERbeta-mediated and nonestrogenic actions.
[26] Parrott JA, Skinner MK. Kit-ligand/stem cell factor induces pri- Biol Reprod 2002;67(4):1285–96.
mordial follicle development and initiates folliculogenesis. [45] Jefferson W. Neonatal genistein treatment alters ovarian differenti-
Endocrinology 1999;140(9):4262–71. ation in the mouse: inhibition of oocyte nest breakdown and
[27] Perez GI, Robles R, Knudson CM, Flaws JA, Korsmeyer SJ, Tilly JL. increased oocyte survival. Biol Reprod 2005;74(1):161–8.
Prolongation of ovarian lifespan into advanced chronological age [46] Iguchi T, Kamiya K, Uesugi Y, Sayama K, Takasugi N. In vitro
by Bax-deficiency. Nat Genet 1999;21(2):200–3. fertilization of oocytes from polyovular follicles in mouse ovaries
[28] Greenfeld CR, Pepling ME, Babus JK, Furth PA, Flaws JA. BAX reg- exposed neonatally to diethylstilbestrol. In Vivo 1991;
ulates follicular endowment in mice. Reproduction 2007;133 5(4):359–63.
(5):865–76. [47] Dandekar PV, Martin MC, Glass RH. Polyovular follicles associ-
[29] Myers M, Morgan FH, Liew SH, Zerafa N, Gamage TU, Sarraj M, ated with human in vitro fertilization. Fertil Steril 1988;49(3):483–6.
et al. PUMA regulates germ cell loss and primordial follicle endow- [48] Woodruff TK, D’Agostino J, Schwartz NB, Mayo KE. Dynamic
ment in mice. Reproduction 2014 Aug;148(2):211–9. changes in inhibin messenger RNAs in rat ovarian follicles during
[30] Liew SH, Vaithiyanathan K, Cook M, Bouillet P, Scott CL, Kerr JB, the reproductive cycle. Science 1988;239(4845):1296–9.
et al. Loss of the proapoptotic BH3-only protein BCL-2 modifying [49] Welt C, Sidis Y, Keutmann H, Schneyer A. Activins, inhibins, and
factor prolongs the fertile life span in female mice. Biol Reprod 2014 follistatins: from endocrinology to signaling. A paradigm for the
Apr;90(4):77. new millennium. Exp Biol Med (Maywood) 2002;227(9):724–52.

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


44 2. REGULATION OF FOLLICLE FORMATION AND DEVELOPMENT BY OVARIAN SIGNALING PATHWAYS

[50] Kimura F, Bonomi LM, Schneyer AL. Follistatin regulates germ cell [70] Johnson J, Espinoza T, McGaughey RW, Rawls A, Wilson-Rawls J.
nest breakdown and primordial follicle formation. Endocrinology Notch pathway genes are expressed in mammalian ovarian folli-
2011;152(2):697–706. cles. Mech Dev 2001;109(2):355–61.
[51] Bristol-Gould S. Establishing and maintaining the mammalian [71] Trombly DJ, Woodruff TK, Mayo KE. Roles for transforming
ovarian follicle pool. ProQuest Information and Learning growth factor beta superfamily proteins in early folliculogenesis.
Company Semin Reprod Med 2009;27(1):14–23.
[52] Kimura F, Sidis Y, Bonomi L, Xia Y, Schneyer A. The follistatin-288 [72] Murta D, Batista M, Silva E, Trindade A, Mateus L, Duarte A, et al.
isoform alone is sufficient for survival but not for normal fertility in Differential expression of Notch component and effector genes dur-
mice. Endocrinology 2010;151(3):1310–9. ing ovarian follicle and corpus luteum development during the oes-
[53] Wang Z, Niu W, Wang Y, Teng Z, Wen J, Xia G, et al. Follistatin288 trous cycle. Reprod Fertil Dev 2015;27(7):1038–48.
regulates germ cell cyst breakdown and primordial follicle assem- [73] Duncan AW, Rattis FM, DiMascio LN, Congdon KL, Pazianos G,
bly in the mouse ovary. PLoS One 2015;10(6):e0129643. Zhao C, et al. Integration of Notch and Wnt signaling in hematopoi-
[54] McCoshen JA, McCallion DJ. A study of the primordial germ cells etic stem cell maintenance. Nat Immunol 2005;6(3):314–22.
during their migratory phase in Steel mutant mice. Experientia [74] Vanorny DA, Prasasya RD, Chalpe AJ, Kilen SM, Mayo KE. Notch
1975;31(5):589–90. signaling regulates ovarian follicle formation and coordinates fol-
[55] Jones RL, Pepling ME. KIT signaling regulates primordial follicle licular growth. Mol Endocrinol 2014;28(4):499–511.
formation in the neonatal mouse ovary. Dev Biol 2013;382 [75] Trombly DJ, Woodruff TK, Mayo KE. Suppression of Notch signal-
(1):186–97. ing in the neonatal mouse ovary decreases primordial follicle for-
[56] Asadi-Azarbaijani B, Santos RR, Jahnukainen K, Braber S, van mation. Endocrinology 2009;150(2):1014–24.
Duursen MBM, Toppari J, et al. Developmental effects of imatinib [76] Terauchi KJ, Shigeta Y, Iguchi T, Sato T. Role of Notch signaling in
mesylate on follicle assembly and early activation of primordial fol- granulosa cell proliferation and polyovular follicle induction dur-
licle pool in postnatal rat ovary. Reprod Biol 2017;17(1):25–33. ing folliculogenesis in mouse ovary. Cell Tissue Res 2016;365
[57] Dissen GA, Hirshfield AN, Malamed S, Ojeda SR. Expression of (1):197–208.
neurotrophins and their receptors in the mammalian ovary is [77] Chen C-L, Fu X-F, Wang L-Q, Wang J-J, Ma H-G, Cheng S-F, et al.
developmentally regulated: changes at the time of folliculogenesis. Primordial follicle assembly was regulated by Notch signaling
Endocrinology 1995;136(10):4681–92. pathway in the mice. Mol Biol Rep 2014;41(3):1891–9.
[58] Dissen GA, Romero C, Hirshfield AN, Ojeda SR. Nerve growth fac- [78] Edwards DR, Handsley MM, Pennington CJ. The ADAM metallo-
tor is required for early follicular development in the mammalian proteinases. Mol Asp Med 2008;29(5):258–89.
ovary. Endocrinology 2001;142(5):2078–86. [79] Feng L, Wang Y, Cai H, Sun G, Niu W, Xin Q, et al. ADAM10-Notch
[59] Kerr B, Garcia-Rudaz C, Dorfman M, Paredes A, Ojeda SR. NTRK1 signaling governs the recruitment of ovarian pregranulosa cells
and NTRK2 receptors facilitate follicle assembly and early follicular and controls folliculogenesis in mice. J Cell Sci 2016;129(11)
development in the mouse ovary. Reproduction 2009;138 jcs.184267–2212.
(1):131–40. [80] Xu J, Gridley T. Notch2 is required in somatic cells for breakdown
[60] Schindler R, Nilsson E, Skinner MK. Induction of ovarian primor- of ovarian germ-cell nests and formation of primordial follicles.
dial follicle assembly by connective tissue growth factor CTGF. BMC Biol BioMed 2013;11(1):13.
PLoS One 2010;5(9):e12979. [81] Hahn KL, Johnson J, Beres BJ, Howard S, Wilson-Rawls J. Lunatic
[61] Wahab NA, Weston BS, Mason RM. Connective tissue growth fac- fringe null female mice are infertile due to defects in meiotic mat-
tor CCN2 interacts with and activates the tyrosine kinase receptor uration. Development 2005;132(4):817–28.
TrkA. J Am Soc Nephrol 2005 Feb;16(2):340–51. [82] Zhao L, Du X, Huang K, Zhang T, Teng Z, Niu W, et al. Rac1 mod-
[62] Rajareddy S, Reddy P, Du C, Liu L, Jagarlamudi K, Tang W, et al. ulates the formation of primordial follicles by facilitating STAT3-
p27kip1 (cyclin-dependent kinase inhibitor 1B) controls ovarian directed Jagged1, GDF9 and BMP15 transcription in mice. Sci
development by suppressing follicle endowment and activation Rep 2016;6(1):23972.
and promoting follicle atresia in mice. Mol Endocrinol 2007;21 [83] Guo M, Zhang H, Bian F, Li G, Mu X, Wen J, et al. P4 down-
(9):2189–202. regulates Jagged2 and Notch1 expression during primordial folli-
[63] Perez-Sanz J, Arluzea J, Matorras R, González-Santiago N, Bilbao J, culogenesis. Front Biosci 2012;4:2731–44.
Yeh N, et al. Increased number of multi-oocyte follicles (MOFs) in [84] Dorfman MD, Kerr B, Garcia-Rudaz C, Paredes AH, Dissen GA,
juvenile p27Kip1 mutant mice: potential role of granulosa cells. Ojeda SR. Neurotrophins acting via TRKB receptors activate the
Hum Reprod 2013;28(4):1023–30. JAGGED1-NOTCH2 cell-cell communication pathway to facilitate
[64] Li R, Albertini DF. The road to maturation: somatic cell interaction early ovarian development. Endocrinology 2011;152(12):5005–16.
and self-organization of the mammalian oocyte. Nat. Rev Mol Cell [85] Meredith S, Dudenhoeffer G, Jackson K. Classification of small type
Biol 2013;14(3):141–52. B/C follicles as primordial follicles in mature rats. J Reprod Fertil
[65] Kopan R, Ilagan MXG. The canonical Notch signaling pathway: 2000;119(1):43–8.
unfolding the activation mechanism. Cell 2009;137(2):216–33. [86] Epifano O, Liang LF, Familari M, Moos MC, Dean J. Coordinate
[66] Mumm JS, Schroeter EH, Saxena MT, Griesemer A, Tian X, Pan DJ, expression of the three zona pellucida genes during mouse oogen-
et al. A ligand-induced extracellular cleavage regulates gamma- esis. Development 1995;121(7):1947–56.
secretase-like proteolytic activation of Notch1. Mol Cell 2000; [87] Ueno S, Takahashi M, Manganaro TF, Ragin RC, Donahoe PK. Cel-
5(2):197–206. lular localization of m€ullerian inhibiting substance in the develop-
[67] Kovall RA. More complicated than it looks: assembly of Notch ing rat ovary. Endocrinology 1989;124(2):1000–6.
pathway transcription complexes. Oncogene 2008;27(38):5099–109. [88] Ueno S, Kuroda T, Maclaughlin DT, Ragin RC, Manganaro TF,
[68] Artavanis-Tsakonas S, Rand MD, Lake RJ. Notch signaling: cell fate Donahoe PK. Mullerian inhibiting substance in the adult rat ovary
control and signal integration in development. Science 1999;284 during various stages of the estrous cycle. Endocrinology 1989;
(5415):770–6. 125(2):1060–6.
[69] Lopez-Schier H, St Johnston D. Delta signaling from the germ line [89] van Rooij IAJ, Broekmans FJM, Velde te ER, Fauser BCJM,
controls the proliferation and differentiation of the somatic follicle Bancsi LFJMM, de Jong FH, et al. Serum anti-M€ ullerian hormone
cells during Drosophila oogenesis. Genes Dev 2001;15(11): levels: a novel measure of ovarian reserve. Hum Reprod 2002;17
1393–405. (12):3065–71.

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


REFERENCES 45
[90] de Vet A, Laven JSE, de Jong FH, Themmen APN, Fauser BCJM. [108] Yoshida H, Takakura N, Kataoka H, Kunisada T, Okamura H,
Antim€ ullerian hormone serum levels: a putative marker for ovar- Nishikawa SI. Stepwise requirement of c-kit tyrosine kinase in
ian aging. Fertil Steril 2002;77(2):357–62. mouse ovarian follicle development. Dev Biol 1997;184(1):122–37.
[91] Meduri G, Massin N, Guibourdenche J, Bachelot A, Fiori O, [109] Hutt KJ, McLaughlin EA, Holland MK. KIT/KIT ligand in mam-
Kuttenn F, et al. Serum anti-M€ ullerian hormone expression in malian oogenesis and folliculogenesis: roles in rabbit and murine
women with premature ovarian failure. Hum Reprod 2007;22(1): ovarian follicle activation and oocyte growth. Biol Reprod 2006;75
117–23. (3):421–33.
[92] Durlinger AL, Kramer P, Karels B, de Jong FH, Uilenbroek JT, [110] Reddy P, Shen L, Ren C, Boman K, Lundin E, Ottander U, et al.
Grootegoed JA, et al. Control of primordial follicle recruitment Activation of Akt (PKB) and suppression of FKHRL1 in mouse
by anti-M€ ullerian hormone in the mouse ovary. Endocrinology and rat oocytes by stem cell factor during follicular activation
1999;140(12):5789–96. and development. Dev Biol 2005;281(2):160–70.
[93] Durlinger ALL, Gruijters MJG, Kramer P, Karels B, Ingraham HA, [111] Saatcioglu HD, Cuevas I, Castrillon DH. Control of oocyte reawa-
Nachtigal MW, et al. Anti-M€ ullerian hormone inhibits initiation of kening by kit. PLoS Genet 2016;12(8):e1006215.
primordial follicle growth in the mouse ovary. Endocrinology [112] Horie K, Takakura K, Taii S, Narimoto K, Noda Y, Nishikawa S,
2002;143(3):1076–84. et al. The expression of c-kit protein during oogenesis and early
[94] Visser JA, Durlinger ALL, Peters IJJ, van den Heuvel ER, Rose UM, embryonic development. Biol Reprod 1991;45(4):547–52.
Kramer P, et al. Increased oocyte degeneration and follicular atresia [113] Adhikari D, Risal S, Liu K, Shen Y. Pharmacological inhibition of
during the estrous cycle in Anti-M€ ullerian hormone null mice. mTORC1 prevents over-activation of the primordial follicle pool
Endocrinology 2007;148(5):2301–8. in response to elevated PI3K signaling. PLoS One 2013;8(1):
[95] Arden KC, Biggs WH. Regulation of the FoxO family of transcrip- e53810.
tion factors by phosphatidylinositol-3 kinase-activated signaling. [114] Adhikari D, Zheng W, Shen Y, Gorre N, H€am€al€ainen T,
Arch Biochem Biophys 2002;403(2):292–8. Cooney AJ, et al. Tsc/mTORC1 signaling in oocytes governs
[96] Castrillon DH, Miao L, Kollipara R, Horner JW, DePinho RA. Sup- the quiescence and activation of primordial follicles. Hum Mol
pression of ovarian follicle activation in mice by the transcription Genet 2010;19(3):397–410.
factor Foxo3a. Science 2003;301(5630):215–8. [115] Adhikari D, Flohr G, Gorre N, Shen Y, Yang H, Lundin E, et al.
[97] Liu L, Rajareddy S, Reddy P, Du C, Jagarlamudi K, Shen Y, et al. Disruption of Tsc2 in oocytes leads to overactivation of the entire
Infertility caused by retardation of follicular development in mice pool of primordial follicles. Mol Hum Reprod 2009;15
with oocyte-specific expression of Foxo3a. Development 2007;134 (12):765–70.
(1):199–209. [116] Gorre N, Adhikari D, Lindkvist R, Br€annstr€ om M, Liu K, Shen Y.
[98] Gallardo T, Shirley L, John GB, Castrillon DH. Generation of a germ mTORC1 Signaling in oocytes is dispensable for the survival of
cell-specific mouse transgenic Cre line, Vasa-Cre. Genesis 2007;45 primordial follicles and for female fertility. PLoS One 2014;9(10)
(6):413–7. e110491.
[99] John GB, Gallardo TD, Shirley LJ, Castrillon DH. Foxo3 is a PI3K- [117] Zhang H, Liu K. Cellular and molecular regulation of the activa-
dependent molecular switch controlling the initiation of oocyte tion of mammalian primordial follicles: somatic cells initiate folli-
growth. Dev Biol 2008;321(1):197–204. cle activation in adulthood. Hum Reprod Update 2015;21
[100] Reddy P, Liu L, Adhikari D, Jagarlamudi K, Rajareddy S, Shen Y, (6):779–86.
et al. Oocyte-specific deletion of Pten causes premature activa- [118] Suzumori N, Yan C, Matzuk MM, Rajkovic A. Nobox is a
tion of the primordial follicle pool. Science 2008;319(5863):611–3. homeobox-encoding gene preferentially expressed in primordial
[101] Reddy P, Adhikari D, Zheng W, Liang S, H€am€al€ainen T, and growing oocytes. Mech Dev 2002;111(1-2):137–41.
Tohonen V, et al. PDK1 signaling in oocytes controls reproductive [119] Rajkovic A, Pangas SA, Ballow D, Suzumori N, Matzuk MM.
aging and lifespan by manipulating the survival of primordial fol- NOBOX deficiency disrupts early folliculogenesis and oocyte-
licles. Hum Mol Genet 2009;18(15):2813–24. specific gene expression. Science 2004;305(5687):1157–9.
[102] Kim S-Y, Ebbert K, Cordeiro MH, Romero M, Zhu J, Serna VA, [120] Choi Y, Rajkovic A. Characterization of NOBOX DNA binding
et al. Cell autonomous phosphoinositide 3-kinase activation in specificity and its regulation of Gdf9 and Pou5f1 promoters.
oocytes disrupts normal ovarian function through promoting sur- J Biol Chem 2006;281(47):35747–56.
vival and overgrowth of ovarian follicles. Endocrinology 2015;156 [121] Bouilly J, Bachelot A, Broutin I, Touraine P, Binart N. Novel
(4):1464–76. NOBOX loss-of-function mutations account for 6.2% of cases in
[103] Hovatta O. Cryopreservation and culture of human ovarian cor- a large primary ovarian insufficiency cohort. Hum Mutat
tical tissue containing early follicles. Eur J Obstet Gynecol Reprod 2011;32(10):1108–13.
Biol 2004;113(Suppl 1):S50–4. [122] Pangas SA, Choi Y, Ballow DJ, Zhao Y, Westphal H, Matzuk MM,
[104] Duncan FE, Pavone ME, Gunn AH, Badawy S, Gracia C, et al. Oogenesis requires germ cell-specific transcriptional regula-
Ginsberg JP, et al. Pediatric and teen ovarian tissue removed for tors Sohlh1 and Lhx8. Proc Natl Acad Sci U S A 2006;103
cryopreservation contains follicles irrespective of age, disease (21):8090–5.
diagnosis, treatment history, and specimen processing methods. [123] Choi Y, Ballow DJ, Xin Y, Rajkovic A. Lim homeobox gene, lhx8, is
J Adolesc Young Adult Oncol 2015;4(4):174–83. essential for mouse oocyte differentiation and survival. Biol
[105] Li J, Kawamura K, Cheng Y, Liu S, Klein C, Liu S, et al. Activation Reprod 2008;79(3):442–9.
of dormant ovarian follicles to generate mature eggs. Proc Natl [124] Ren Y, Suzuki H, Jagarlamudi K, Golnoski K, McGuire M,
Acad Sci U S A 2010;107(22):10280–4. Lopes R, et al. Lhx8 regulates primordial follicle activation and
[106] Adhikari D, Gorre N, Risal S, Zhao Z, Zhang H, Shen Y, et al. The postnatal folliculogenesis. BMC Biol 2015;13(1):39.
safe use of a PTEN inhibitor for the activation of dormant mouse [125] Ballow DJ, Xin Y, Choi Y, Pangas SA, Rajkovic A. Sohlh2 is a germ
primordial follicles and generation of fertilizable eggs. PLoS One cell-specific bHLH transcription factor. Gene Expr Patterns 2006;6
2012;7(6):e39034. (8):1014–8.
[107] Huang EJ, Manova K, Packer AI, Sanchez S, Bachvarova RF, [126] Shin YH, Ren Y, Suzuki H, Golnoski KJ, Ahn HW, Mico V, et al.
Besmer P. The murine steel panda mutation affects kit ligand Transcription factors SOHLH1 and SOHLH2 coordinate oocyte
expression and growth of early ovarian follicles. Dev Biol 1993 differentiation without affecting meiosis I. J Clin Invest 2017;127
May;157(1):100–9. (6):2106–17.

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


46 2. REGULATION OF FOLLICLE FORMATION AND DEVELOPMENT BY OVARIAN SIGNALING PATHWAYS

[127] Choi Y, Yuan D, Rajkovic A. Germ cell-specific transcriptional [147] Pangas SA, Li X, Robertson EJ, Matzuk MM. Premature luteiniza-
regulator sohlh2 is essential for early mouse folliculogenesis tion and cumulus cell defects in ovarian-specific Smad4 knockout
and oocyte-specific gene expression. Biol Reprod 2008;79(6): mice. Mol Endocrinol 2006;20(6):1406–22.
1176–82. [148] McGrath SA, Esquela AF, Lee SJ. Oocyte-specific expression of
[128] Suzuki H, Ahn HW, Chu T, Bowden W, Gassei K, Orwig K, et al. growth/differentiation factor-9. Mol Endocrinol 1995;9(1):131–6.
SOHLH1 and SOHLH2 coordinate spermatogonial differentia- [149] Jaatinen R, Laitinen MP, Vuojolainen K, Aaltonen J, Louhio H,
tion. Dev Biol 2012;361(2):301–12. Heikinheimo K, et al. Localization of growth differentiation
[129] Sutherland JM, Keightley RA, Nixon B, Roman SD, Robker RL, factor-9 (GDF-9) mRNA and protein in rat ovaries and cDNA
Russell DL, et al. Suppressor of cytokine signaling 4 (SOCS4): cloning of rat GDF-9 and its novel homolog GDF-9B. Mol Cell
moderator of ovarian primordial follicle activation. J Cell Physiol Endocrinol 1999;156(1–2):189–93.
2012;227(3):1188–98. [150] Carabatsos MJ, Elvin J, Matzuk MM, Albertini DF. Characteriza-
[130] Uda M, Ottolenghi C, Crisponi L, Garcia JE, Deiana M, Kimber W, tion of oocyte and follicle development in growth differentiation
Forabosco A, Cao A, Schssinger D, Pilia G. Foxl2 disruption factor-9-deficient mice. Dev Biol 1998;204(2):373–84.
causes mouse ovarian failure by pervasive blockage of follicle [151] Elvin JA, Yan C, Wang P, Nishimori K, Matzuk MM. Molecular
development. Hum Mol Genet 2004;13(11):1171–81. characterization of the follicle defects in the growth differentiation
[131] Dong J, Albertini DF, Nishimori K, Kumar TR, Lu N, Matzuk MM. factor 9-deficient ovary. Mol Endocrinol 1999;13(6):1018–34.
Growth differentiation factor-9 is required during early ovarian [152] Vitt UA, McGee EA, Hayashi M, Hsueh AJ. In vivo treatment with
folliculogenesis. Nature 1996;383(6600):531–5. GDF-9 stimulates primordial and primary follicle progression and
[132] Viger RS, Guittot SM, Anttonen M, Wilson DB, Heikinheimo M. theca cell marker CYP17 in ovaries of immature rats.
Role of the GATA family of transcription factors in endocrine Endocrinology 2000;141(10):3814–20.
development, function, and disease. Mol Endocrinol 2008;22(4): [153] Vitt UA, Hayashi M, Klein C, Hsueh AJ. Growth differentiation
781–98. factor-9 stimulates proliferation but suppresses the follicle-
[133] Zaytouni T, Efimenko EE, Tevosian SG. GATA transcription fac- stimulating hormone-induced differentiation of cultured granu-
tors in the developing reproductive system. Adv Genet losa cells from small antral and preovulatory rat follicles. Biol
2011;76:93–134. Reprod 2000;62(2):370–7.
[134] Padua MB, Fox SC, Jiang T, Morse DA, Tevosian SG. Simulta- [154] Cook-Andersen H, Curnow KJ, Su HI, Chang RJ, Shimasaki S.
neous gene deletion of gata4 and gata6 leads to early disruption Growth and differentiation factor 9 promotes oocyte growth at
of follicular development and germ cell loss in the murine ovary. the primary but not the early secondary stage in three-dimensional
Biol Reprod 2014;91(1):24. follicle culture. J Assist Reprod Genet 2016;33(8):1067–77.
[135] Kumar TR, Wang Y, Lu N, Matzuk MM. Follicle stimulating hor- [155] Fenwick MA, Mora JM, Mansour YT, Baithun C, Franks S,
mone is required for ovarian follicle maturation but not male fer- Hardy K. Investigations of TGF-β signaling in preantral follicles
tility. Nat Genet 1997;15(2):201–4. of female mice reveal differential roles for bone morphogenetic
[136] Dierich A, Sairam MR, Monaco L, Fimia GM, Gansmuller A, protein 15. Endocrinology 2013;154(9):3423–36.
LeMeur M, et al. Impairing follicle-stimulating hormone (FSH) [156] Vitt UA, Mazerbourg S, Klein C, Hsueh AJW. Bone morphoge-
signaling in vivo: targeted disruption of the FSH receptor leads netic protein receptor type II is a receptor for growth differentia-
to aberrant gametogenesis and hormonal imbalance. Proc Natl tion factor-9. Biol Reprod 2002;67(2):473–80.
Acad Sci U S A 1998;95(23):13612–7. [157] Dube JL, Wang P, Elvin J, Lyons KM, Celeste AJ, Matzuk MM. The
[137] Abel MH, Wootton AN, Wilkins V, Huhtaniemi I, Knight PG, bone morphogenetic protein 15 gene is X-linked and expressed in
Charlton HM. The effect of a null mutation in the follicle- oocytes. Mol Endocrinol 1998;12(12):1809–17.
stimulating hormone receptor gene on mouse reproduction. [158] Otsuka F, Shimasaki S. A negative feedback system between
Endocrinology 2000;141(5):1795–803. oocyte bone morphogenetic protein 15 and granulosa cell kit
[138] Massague J. TGF-beta signal transduction. Annu Rev Biochem ligand: its role in regulating granulosa cell mitosis. Proc Natl Acad
1998;67(1):753–91. Sci U S A 2002;99(12):8060–5.
[139] Shimasaki S, Moore RK, Otsuka F, Erickson GF. The bone mor- [159] Galloway SM, KP MNATTY, Cambridge LM, Laitinen MP,
phogenetic protein system in mammalian reproduction. Endocr Juengel JL, Jokiranta TS, et al. Mutations in an oocyte-derived
Rev 2004;25(1):72–101. growth factor gene (BMP15) cause increased ovulation rate and
[140] Miyazono K, Kusanagi K, Inoue H. Divergence and convergence infertility in a dosage-sensitive manner. Nat Genet 2000;25
of TGF-beta/BMP signaling. J Cell Physiol 2001;187(3):265–76. (3):279–83.
[141] Kaivo-Oja N, Jeffery LA, Ritvos O, Mottershead DG. Smad signal- [160] KP MN, Smith P, Hudson NL, Heath DA, Tisdall DJ, WS O, et al.
ling in the ovary. Reprod Biol 2006;4(1):21. Development of the sheep ovary during fetal and early neonatal
[142] Miyazawa K, Shinozaki M, Hara T, Furuya T, Miyazono K. Two life and the effect of fecundity genes. J Reprod Fertil Suppl
major Smad pathways in TGF-beta superfamily signalling. Genes 1995;49:123–35.
Cells 2002;7(12):1191–204. [161] Wilson T, Wu XY, Juengel JL, Ross IK, Lumsden JM, Lord EA,
[143] Mazerbourg S, Klein C, Roh J, Kaivo-Oja N, Mottershead DG, et al. Highly prolific Booroola sheep have a mutation in the intra-
Korchynskyi O, et al. Growth differentiation factor-9 signaling cellular kinase domain of bone morphogenetic protein IB receptor
is mediated by the type I receptor, activin receptor-like kinase 5. (ALK-6) that is expressed in both oocytes and granulosa cells. Biol
Mol Endocrinol 2004;18(3):653–65. Reprod 2001;64(4):1225–35.
[144] Roh J-S, Bondestam J, Mazerbourg S, Kaivo-Oja N, Groome N, [162] Crawford JL, KP MN. The ratio of growth differentiation factor 9:
Ritvos O, et al. Growth differentiation factor-9 stimulates inhibin bone morphogenetic protein 15 mRNA expression is tightly
production and activates Smad2 in cultured rat granulosa cells. co-regulated and differs between species over a wide range of
Endocrinology 2003;144(1):172–8. ovulation rates. Mol Cell Endocrinol 2012;348(1):339–43.
[145] Pangas SA, Li X, Umans L, Zwijsen A, Huylebroeck D, [163] Liao WX, Moore RK, Otsuka F, Shimasaki S. Effect of intracellular
Gutierrez C, et al. Conditional deletion of Smad1 and Smad5 in interactions on the processing and secretion of bone morphoge-
somatic cells of male and female gonads leads to metastatic tumor netic protein-15 (BMP-15) and growth and differentiation factor-
development in mice. Mol Cell Biol 2008;28(1):248–57. 9. Implication of the aberrant ovarian phenotype of BMP-15
[146] Li Q, Pangas SA, Jorgez CJ, Graff JM, Weinstein M, Matzuk MM. mutant sheep. J Biol Chem 2003;278(6):3713–9.
Redundant roles of SMAD2 and SMAD3 in ovarian granulosa [164] Mottershead DG, Sugimura S, Al-Musawi SL, Li J-J, Richani D,
cells in vivo. Mol Cell Biol 2008;28(23):7001–11. White MA, et al. Cumulin, an oocyte-secreted heterodimer of

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


REFERENCES 47
the transforming growth factor-β family, is a potent activator of [183] Matzuk MM, Finegold MJ, Su JG, Hsueh AJ, Bradley A. Alpha-
granulosa cells and improves oocyte quality. J Biol Chem inhibin is a tumour-suppressor gene with gonadal specificity in
2015;290(39):24007–20. mice. Nature 1992;360(6402):313–9.
[165] Yan C, Wang P, DeMayo J, DeMayo FJ, Elvin JA, Carino C, et al. [184] Kipp JL, Kilen SM, Woodruff TK, Mayo KE. Activin regulates
Synergistic roles of bone morphogenetic protein 15 and growth estrogen receptor gene expression in the mouse ovary. J Biol Chem
differentiation factor 9 in ovarian function. Mol Endocrinol 2007;282(50):36755–65.
2001;15(6):854–66. [185] Rao MC, Midgley AR, Richards JS. Hormonal regulation of ovar-
[166] Mottershead DG, Ritter LJ, Gilchrist RB. Signalling pathways ian cellular proliferation. Cell 1978;14(1):71–8.
mediating specific synergistic interactions between GDF9 and [186] Bendell JJ, Dorrington J. Estradiol-17 beta stimulates DNA synthe-
BMP15. Mol Hum Reprod 2012 Mar;18(3):121–8. sis in rat granulosa cells: action mediated by transforming growth
[167] Peng J, Li Q, Wigglesworth K, Rangarajan A, Kattamuri C, factor-beta. Endocrinology 1991;128(5):2663–5.
Peterson RT, et al. Growth differentiation factor 9: bone mor- [187] Britt KL, Drummond AE, Cox VA, Dyson M, Wreford NG,
phogenetic protein 15 heterodimers are potent regulators of Jones ME, et al. An age-related ovarian phenotype in mice with
ovarian functions. Proc Natl Acad Sci U S A 2013;110(8): targeted disruption of the Cyp 19 (aromatase) gene.
E776–85. Endocrinology 2000;141(7):2614–23.
[168] Patiño LC, Walton KL, Mueller TD, Johnson KE, Stocker W, [188] Kipp JL, Golebiowski A, Rodriguez G, Demczuk M, Kilen SM,
Richani D, et al. BMP15 mutations associated with primary ovar- Mayo KE. Gene expression profiling reveals Cyp26b1 to be an
ian insufficiency reduce expression, activity, or synergy with activin regulated gene involved in ovarian granulosa cell prolifer-
GDF9. J Clin Endocrinol Metabol 2017;102(3):1009–19. ation. Endocrinology 2011;152(1):303–12.
[169] Norling A, Hirschberg AL, Rodriguez-Wallberg KA, Iwarsson E, [189] Zhang C-P, Yang J-L, Zhang J, Li L, Huang L, Ji S-Y, et al. Notch
Wedell A, Barbaro M. Identification of a duplication within the signaling is involved in ovarian follicle development by regulating
GDF9 gene and novel candidate genes for primary ovarian insuf- granulosa cell proliferation. Endocrinology 2011;152(6):2437–47.
ficiency (POI) by a customized high-resolution array comparative [190] Sun X-F, Sun X-H, Cheng S-F, Wang J-J, Feng Y-N, Zhao Y, et al.
genomic hybridization platform. Hum Reprod 2014;29 Interaction of the transforming growth factor-β and Notch signal-
(8):1818–27. ing pathways in the regulation of granulosa cell proliferation.
[170] Wang T-T, Ke Z-H, Song Y, Chen L-T, Chen X-J, Feng C, et al. Reprod Fertil Dev 1873;28(12):3.
Identification of a mutation in GDF9 as a novel cause of dimin- [191] Manosalva I, González A, Kageyama R. Hes1 in the somatic cells
ished ovarian reserve in young women. Hum Reprod 2013;28 of the murine ovary is necessary for oocyte survival and matura-
(9):2473–81. tion. Dev Biol 2013;375(2):140–51.
[171] Juengel JL, McNATTY KP. The role of proteins of the trans- [192] Magoffin DA. Ovarian theca cell. Int J Biochem Cell Biol 2005
forming growth factor-beta superfamily in the intraovarian reg- Jul;37(7):1344–9.
ulation of follicular development. Hum Reprod Update 2005;11 [193] Honda A, Hirose M, Hara K, Matoba S, Inoue K, Miki H, et al. Iso-
(2):143–60. lation, characterization, and in vitro and in vivo differentiation of
[172] Christopher B. Immunolocalization of transforming growth putative thecal stem cells. Proc Natl Acad Sci U S A 2007;104
factor-beta1 during follicular development and atresia in the (30):12389–94.
mouse ovary. Endocr J 2000;47(4):475–80. [194] Liu C, Peng J, Matzuk MM. Yao HHC. Lineage specification of
[173] Woodruff TK, Meunier H, Jones PB, Hsueh AJ, Mayo KE. Rat ovarian theca cells requires multicellular interactions via oocyte
inhibin: molecular cloning of alpha- and beta-subunit comple- and granulosa cells. Nat Commun 2015;6:6934.
mentary deoxyribonucleic acids and expression in the ovary. [195] Magoffin DA, Magarelli PC. Preantral follicles stimulate luteiniz-
Mol Endocrinol 1987;1(8):561–8. ing hormone independent differentiation of ovarian theca-
[174] Makanji Y, Zhu J, Mishra R, Holmquist C, Wong WPS, interstitial cells by an intrafollicular paracrine mechanism.
Schwartz NB, et al. Inhibin at 90: from discovery to clinical appli- Endocrine 1995;3(2):107–12.
cation, a historical review. Endocr Rev 2014;35(5):747–94. [196] Magarelli PC, Zachow RJ, Magoffin DA. Developmental and hor-
[175] Nakamura T, Takio K, Eto Y, Shibai H, Titani K, Sugino H. Acti- monal regulation of rat theca-cell differentiation factor secretion in
vin-binding protein from rat ovary is follistatin. Science 1990;247 ovarian follicles. Biol Reprod 1996;55(2):416–20.
(4944):836–8. [197] Wijgerde M, Ooms M, Hoogerbrugge JW, Grootegoed JA.
[176] Li R, Phillips DM, Mather JP. Activin promotes ovarian follicle Hedgehog signaling in mouse ovary: Indian hedgehog and
development in vitro. Endocrinology 1995;136(3):849–56. desert hedgehog from granulosa cells induce target gene ex-
[177] Yokota H, Yamada K, Liu X, Kobayashi J, Abe Y, pression in developing theca cells. Endocrinology 2005;146(8):
Mizunuma H, et al. Paradoxical action of activin A on follicu- 3558–66.
logenesis in immature and adult mice. Endocrinology 1997;138 [198] Edson MA, Nagaraja AK, Matzuk MM. The mammalian ovary
(11):4572–6. from genesis to revelation. Endocr Rev 2009;30(6):624–712.
[178] Mizunuma H, Liu X, Andoh K, Abe Y, Kobayashi J, Yamada K, [199] Camp TA, Rahal JO, Mayo KE. Cellular localization and hormonal
et al. Activin from secondary follicles causes small preantral folli- regulation of follicle-stimulating hormone and luteinizing hor-
cles to remain dormant at the resting stage. Endocrinology mone receptor messenger RNAs in the rat ovary. Mol Endocrinol
1999;140(1):37–42. 1991;5(10):1405–17.
[179] Pangas SA, Jorgez CJ, Tran M, Agno J, Li X, Brown CW, et al. [200] Chun SY, Eisenhauer KM, Minami S, Billig H, Perlas E, Hsueh AJ.
Intraovarian activins are required for female fertility. Mol Endo- Hormonal regulation of apoptosis in early antral follicles: follicle-
crinol 2007;21(10):2458–71. stimulating hormone as a major survival factor. Endocrinology
[180] Tomic D, Brodie SG, Deng C, Hickey RJ, Babus JK, Malkas LH, 1996;137(4):1447–56.
et al. Smad 3 may regulate follicular growth in the mouse ovary. [201] Richards JS, Russell DL, Ochsner S, Hsieh M, Doyle KH,
Biol Reprod 2002;66(4):917–23. Falender AE, et al. Novel signaling pathways that control ovarian
[181] McMullen ML, Cho BN, Yates CJ, Mayo KE. Gonadal pathologies follicular development, ovulation, and luteinization. Recent Prog
in transgenic mice expressing the rat inhibin alpha-subunit. Horm Res 2002;57:195–220.
Endocrinology 2001;142(11):5005–14. [202] Hunzicker-Dunn M, Maizels ET. FSH signaling pathways in
[182] Cho BN, McMullen ML, Pei L, Yates CJ, Mayo KE. Reproductive immature granulosa cells that regulate target gene expression:
deficiencies in transgenic mice expressing the rat inhibin alpha- branching out from protein kinase A. Cell Signal 2006;18
subunit gene. Endocrinology 2001;142(11):4994–5004. (9):1351–9.

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


48 2. REGULATION OF FOLLICLE FORMATION AND DEVELOPMENT BY OVARIAN SIGNALING PATHWAYS

[203] Hirshfield AN. Development of follicles in the mammalian ovary. binding protein 1 (YB-1) enhances gene expression in granulosa
Int Rev Cytol 1991;124:43–101. cells in response to follicle-stimulating hormone (FSH). J Biol
[204] Richards JS. Maturation of ovarian follicles: actions and interac- Chem 2016;291(23):12145–60.
tions of pituitary and ovarian hormones on follicular cell differen- [222] Cottom J, Salvador LM, Maizels ET, Reierstad S, Park Y, Carr DW,
tiation. Physiol Rev 1980;60(1):51–89. et al. Follicle-stimulating hormone activates extracellular signal-
[205] Hsueh AJ, Rauch R. Ovarian Kaleidoscope database: ten years regulated kinase but not extracellular signal-regulated kinase
and beyond. Biol Reprod 2012;86(6):192. kinase through a 100-kDa phosphotyrosine phosphatase. J Biol
[206] Hsueh AJ, Adashi EY, Jones PB, Welsh TH. Hormonal regulation Chem 2003;278(9):7167–79.
of the differentiation of cultured ovarian granulosa cells. Endocr [223] Fujinaga H, Yamoto M, Shikone T, Nakano R. FSH and LH
Rev 1984;5(1):76–127. up-regulate epidermal growth factor receptors in rat granulosa
[207] Knecht M, Amsterdam A, Catt K. The regulatory role of cyclic cells. J Endocrinol 1994;140(2):171–7.
AMP in hormone-induced of granulosa cell differentiation. [224] El-Hayek S, Demeestere I, Clarke HJ. Follicle-stimulating hor-
J Biol Chem 1981;256(20):10628–33. mone regulates expression and activity of epidermal growth fac-
[208] Knecht M, Ranta T, Catt KJ. Granulosa cell differentiation in vitro: tor receptor in the murine ovarian follicle. Proc Natl Acad Sci U S
induction and maintenance of follicle-stimulating hormone recep- A 2014;111(47):16778–83.
tors by adenosine 30 ,50 -monophosphate. Endocrinology 1983;113 [225] Babu PS, Krishnamurthy H, Chedrese PJ, Sairam MR. Activation
(3):949–56. of extracellular-regulated kinase pathways in ovarian granulosa
[209] Taylor SS, Knighton DR, Zheng J, Eyck Ten LF, Sowadski JM. cells by the novel growth factor type 1 follicle-stimulating hor-
cAMP-dependent protein kinase and the protein kinase family. mone receptor. Role in hormone signaling and cell proliferation.
Faraday Discuss 1992;93:143–52. J Biol Chem 2000;275(36):27615–26.
[210] Mukherjee A, Park-Sarge OK, Mayo KE. Gonadotropins induce [226] Prasasya RD, Mayo KE. Notch signaling regulates differentiation
rapid phosphorylation of the 30 ,50 -cyclic adenosine monopho- and steroidogenesis in female mouse ovarian granulosa cells.
sphate response element binding protein in ovarian granulosa Endocrinology 2018;159(1):184–98.
cells. Endocrinology 1996;137(8):3234–45. [227] Wang J, Liu S, Peng L, Dong Q, Bao R, Lv Q, et al. Notch signaling
[211] Carlone DL, Richards JS. Functional interactions, phosphoryla- pathway regulates progesterone secretion in murine luteal cells.
tion, and levels of 30 ,50 -cyclic adenosine monophosphate- Reprod Sci 2015;22(10):1243–51.
regulatory element binding protein and steroidogenic factor-1 [228] Zhou J, Kumar TR, Matzuk MM, Bondy C. Insulin-like growth
mediate hormone-regulated and constitutive expression of aro- factor I regulates gonadotropin responsiveness in the murine
matase in gonadal cells. Mol Endocrinol 1997;11(3):292–304. ovary. Mol Endocrinol 1997;11(13):1924–33.
[212] Tremblay JJ, Viger RS. Transcription factor GATA-4 is activated [229] Zhou P, Baumgarten SC, Wu Y, Bennett J, Winston N, Hirshfeld-
by phosphorylation of serine 261 via the cAMP/protein kinase Cytron J, et al. IGF-I signaling is essential for FSH stimulation of
a signaling pathway in gonadal cells. J Biol Chem 2003;278 AKT and steroidogenic genes in granulosa cells. Mol Endocrinol
(24):22128–35. 2013;27(3):511–23.
[213] Tremblay JJ, Viger RS. GATA factors differentially activate multi- [230] Baumgarten SC, Armouti M, Ko C, Stocco C. IGF1R expression in
ple gonadal promoters through conserved GATA regulatory ele- ovarian granulosa cells is essential for steroidogenesis, follicle sur-
ments. Endocrinology 2001;142(3):977–86. vival, and fertility in female mice. Endocrinology 2017;158
[214] DeManno DA, Cottom JE, Kline MP, Peters CA, Maizels ET, (7):2309–18.
Hunzicker-Dunn M. Follicle-stimulating hormone promotes his- [231] Park Y, Maizels ET, Feiger ZJ, Alam H, Peters CA, Woodruff TK,
tone H3 phosphorylation on serine-10. Mol Endocrinol 1999;13 et al. Induction of cyclin D2 in rat granulosa cells requires FSH-
(1):91–105. dependent relief from FOXO1 repression coupled with positive
[215] Salvador LM, Park Y, Cottom J, Maizels ET, Jones JC, Schillace RV, signals from Smad. J Biol Chem 2005;280(10):9135–48.
et al. Follicle-stimulating hormone stimulates protein kinase [232] Hunzicker-Dunn ME, Lopez-Biladeau B, Law NC, Fiedler SE,
A-mediated histone H3 phosphorylation and acetylation leading Carr DW, Maizels ET. PKA and GAB2 play central roles in the
to select gene activation in ovarian granulosa cells. J Biol Chem FSH signaling pathway to PI3K and AKT in ovarian granulosa
2001;276(43):40146–55. cells. Proc Natl Acad Sci U S A 2012;109(44):E2979–88.
[216] Hunzicker-Dunn M, Mayo K. Chapter 20—gonadotropin signal- [233] Herndon MK, Law NC, Donaubauer EM, Kyriss B, Hunzicker-
ing in the ovary. In: Plant TM, Zeleznik AJ, editors. Knobil and Dunn M. Forkhead box O member FOXO1 regulates the majority
Neill’s physiology of reproduction. 4th ed. Elsevier; 2014. of follicle-stimulating hormone responsive genes in ovarian gran-
p. 895–946. ulosa cells. Mol Cell Endocrinol 2016;434:116–26.
[217] Marshall CJ. Specificity of receptor tyrosine kinase signaling: tran- [234] Liu Z, Rudd MD, Hernandez-Gonzalez I, Gonzalez-Robayna I,
sient versus sustained extracellular signal-regulated kinase activa- Fan H-Y, Zeleznik AJ, et al. FSH and FOXO1 regulate genes in
tion. Cell 1995;80(2):179–85. the sterol/steroid and lipid biosynthetic pathways in granulosa
[218] Dikic I, Blaukat A. Protein tyrosine kinase-mediated pathways in cells. Mol Endocrinol 2009;23(5):649–61.
G protein-coupled receptor signaling. Cell Biochem Biophys [235] Miller WL, Auchus RJ. The molecular biology, biochemistry, and
1999;30(3):369–87. physiology of human steroidogenesis and its disorders. Endocr
[219] Das S, Maizels ET, DeManno D, St Clair E, Adam SA, Hunzicker- Rev 2011;32(1):81–151.
Dunn M. A stimulatory role of cyclic adenosine 30 ,50 - [236] Hillier SG, Whitelaw PF, Smyth CD. Follicular oestrogen synthe-
monophosphate in follicle-stimulating hormone-activated sis: the “two-cell, two-gonadotrophin” model revisited. Mol Cell
mitogen-activated protein kinase signaling pathway in rat ovar- Endocrinol 1994;100(1–2):51–4.
ian granulosa cells. Endocrinology 1996;137(3):967–74. [237] Conneely OM, Mulac-Jericevic B, Lydon JP, De Mayo FJ.
[220] Burgering BM, Bos JL. Regulation of Ras-mediated signalling: Reproductive functions of the progesterone receptor isoforms: les-
more than one way to skin a cat. Trends Biochem Sci 1995;20(1): sons from knock-out mice. Mol Cell Endocrinol 2001;179
18–22. (1–2):97–103.
[221] Donaubauer EM, Hunzicker-Dunn ME. Extracellular signal- [238] Knecht M, Darbon JM, Ranta T, Baukal AJ, Catt KJ. Estrogens
regulated kinase (ERK)-dependent phosphorylation of Y-box- enhance the adenosine 30 ,50 -monophosphate-mediated induction

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS


REFERENCES 49
of follicle-stimulating hormone and luteinizing hormone recep- [249] Sen A, Hammes SR. Granulosa cell-specific androgen receptors
tors in rat granulosa cells. Endocrinology 1984;115(1):41–9. are critical regulators of ovarian development and function. Mol
[239] Wang C, Prossnitz ER, Roy SK. Expression of G protein-coupled Endocrinol 2010;24(7):1393–403.
receptor 30 in the hamster ovary: differential regulation by gonad- [250] Walters KA, Middleton LJ, Joseph SR, Hazra R, Jimenez M,
otropins and steroid hormones. Endocrinology 2007;148 Simanainen U, et al. Targeted loss of androgen receptor signaling
(10):4853–64. in murine granulosa cells of preantral and antral follicles causes
[240] Otto C, Fuchs I, Kauselmann G, Kern H, Zevnik B, Andreasen P, female subfertility. Biol Reprod 2012;87(6):151.
et al. GPR30 does not mediate estrogenic responses in reproduc- [251] Sen A, Prizant H, Light A, Biswas A, Hayes E, Lee H-J, et al.
tive organs in mice. Biol Reprod 2009;80(1):34–41. Androgens regulate ovarian follicular development by increasing
[241] Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Sch€ utz G, follicle stimulating hormone receptor and microRNA-125b
Umesono K, et al. The nuclear receptor superfamily: the second expression. Proc Natl Acad Sci U S A 2014;111(8):3008–13.
decade. Cell 1995;83(6):835–9. [252] Laird M, Thomson K, Fenwick M, Mora J, Franks S, Hardy K.
[242] Lubahn DB, Moyer JS, Golding TS, Couse JF, Korach KS, Androgen stimulates growth of mouse preantral follicles
Smithies O. Alteration of reproductive function but not prenatal in vitro: interaction with follicle-stimulating hormone and with
sexual development after insertional disruption of the mouse growth factors of the TGFβ superfamily. Endocrinology
estrogen receptor gene. Proc Natl Acad Sci U S A 1993;90 2017;158(4):920–35.
(23):11162–6. [253] Franks S. Polycystic ovary syndrome. N Engl J Med 1995;333
[243] Schomberg DW, Couse JF, Mukherjee A, Lubahn DB, Sar M, (13):853–61.
Mayo KE, et al. Targeted disruption of the estrogen receptor-alpha [254] Ehrmann DA. Polycystic ovary syndrome. N Engl J Med 2005;352
gene in female mice: characterization of ovarian responses and (12):1223–36.
phenotype in the adult. Endocrinology 1999;140(6):2733–44. [255] Chen M-J, Chou C-H, Chen S-U, Yang W-S, Yang Y-S, Ho H-N.
[244] Britt KL, Findlay JK. Regulation of the phenotype of ovarian The effect of androgens on ovarian follicle maturation: dihydro-
somatic cells by estrogen. Mol Cell Endocrinol 2003;202(1–2):11–7. testosterone suppress FSH-stimulated granulosa cell proliferation
[245] Krege JH, Hodgin JB, Couse JF, Enmark E, Warner M, Mahler JF, by upregulating PPARγ-dependent PTEN expression. Sci Rep
et al. Generation and reproductive phenotypes of mice lacking 2015;5(1):18319.
estrogen receptor beta. Proc Natl Acad Sci U S A 1998;95 [256] Kayampilly PP, Menon KMJ. Dihydrotestosterone inhibits
(26):15677–82. insulin-stimulated cyclin D2 messenger ribonucleic acid expres-
[246] Couse JF, Yates MM, Deroo BJ, Korach KS. Estrogen receptor-beta sion in rat ovarian granulosa cells by reducing the phosphoryla-
is critical to granulosa cell differentiation and the ovulatory tion of insulin receptor substrate-1. Endocrinology 2006;147
response to gonadotropins. Endocrinology 2005;146(8):3247–62. (1):464–71.
[247] Deroo BJ, Rodriguez KF, Couse JF, Hamilton KJ, Collins JB, [257] Kayampilly PP, Menon KMJ. AMPK activation by dihydrotestos-
Grissom SF, et al. Estrogen receptor beta is required for optimal terone reduces FSH-stimulated cell proliferation in rat granulosa
cAMP production in mouse granulosa cells. Mol Endocrinol cells by inhibiting ERK signaling pathway. Endocrinology
2009;23(7):955–65. 2012;153(6):2831–8.
[248] Couse JF, Hewitt SC, Bunch DO, Sar M, Walker VR, Davis BJ, et al. [258] Wang Y, Du Y, Shen B, Zhou X, Li J, Liu Y, et al. Efficient gener-
Postnatal sex reversal of the ovaries in mice lacking estrogen ation of gene-modified pigs via injection of zygote with Cas9/
receptors alpha and beta. Science 1999;286(5448):2328–31. sgRNA. Sci Rep 2015;5(1):8256.

I. THE OVARIAN FOLLICULAR APPARATUS: OPERATIONAL CHARACTERISTICS

You might also like