Sie sind auf Seite 1von 8

Molecular and Cellular Endocrinology 179 (2001) 39 46

www.elsevier.com/locate/mce

Gonadotropic control of ovarian follicular growth and


development
S.G. Hillier
Reproducti6e Medicine Laboratory, Department of Reproducti6e and De6elopmental Sciences,
Uni6ersity of Edinburgh Centre for Reproducti6e Biology, 37 Chalmers Street, Edinburgh EH3 9EW, UK

Abstract
Development-related paracrine cues that sensitize follicles to follicle stimulating hormone (FSH) and luteinizing hormone (LH)
are crucial to the emergence of a single dominant follicle in each ovulatory menstrual cycle. Sex steroids, insulin-like growth
factors and members of the transforming growth factor-b superfamily are key players in the follicular paracrine system. FSH acts
through membrane-associated granulosa cell receptors (FSHR) to stimulate granulosa cell proliferation and differentiation. The
most responsive follicle at the beginning of the cycle is the first to produce estrogen and express granulosa cell LHR. Paracrine
signalling activated by FSH and LH sustains growth and oestrogen secretion until an ovulation-inducing LH surge is discharged
by the pituitary gland. LH then reprogrammes granulosa cell function, leading to terminal differentiation (luteinization) rupture
of the follicle wall, and release of the fertilizable egg. The genes regulated by the LH surge orchestrate profound changes in sex
steroid production, metabolism and action which are necessary for ovulation. Preovulatory granulosa cells also increase their
ability to metabolise cortisone to cortisol, which may be part of a local anti-inflammatory mechanism to promote rapid healing
of the ruptured ovarian surface. 2001 Elsevier Science Ireland Ltd. All rights reserved.
Keywords: Gonadotropins; Ovary; Ovulation

1. Introduction

2. The new biology of FSH and LH

The ovaries are amongst the most dynamic and


plastic tissues in the body, with the monthly cycle of
follicular maturation, ovulation and (in absence of
pregnancy) resorption of the corpus luteum occurring
300 400 times throughout the average womans reproductive lifespan. The endocrine control of this
process by follicle-stimulating hormone (FSH) and
luteinizing hormone (LH) rests on a network of extracellular and intracellular molecular interactions, the
complexity of which reflects the unique histogenetic
origins and specialized functions of the cell types that
constitute the ovaries. Understanding these interactions is key to defining new targets for pharmaceutical manipulation of ovarian function, towards new
and improved forms of fertility treatment. This review
briefly surveys recent developments in some of these
areas.

Although FSH and LH were first isolated and


purified over 50 years ago, neither gonadotropin was
available in a completely pure form until recently.
Clinicians usually relied on the use of urinary gonadotropin extracts containing FSH with varying
amounts of LH, making it difficult to selectively alter
the dose of either gonadotropin, while experimentalists resorted to scarce and expensive pituitary preparations of varying purity. The breakthrough came
with the preparation of pure, recombinant FSH and
LH by using Chinese Hamster Ovary cells transfected
with expression vectors carrying human cDNAs encoding the common a gonadotropin subunit with one
or other of the b-subunits (Recombinant Human
FSH Product Development Group, 1998). This advance has resulted in the availability of pharmaceutical grade pure FSH and LH preparations that are
finding uses in a clinical setting and which have also
provided powerful new tools in experimental endocrinology (Smyth et al., 1995).

E-mail address: steve.hillier@ed.ac.uk (S.G. Hillier).

0303-7207/01/$ - see front matter 2001 Elsevier Science Ireland Ltd. All rights reserved.
PII: S 0 3 0 3 - 7 2 0 7 ( 0 1 ) 0 0 4 6 9 - 5

40

S.G. Hillier / Molecular and Cellular Endocrinology 179 (2001) 3946

2.1. Gonadotropin receptors


Paralleling this advance in the molecular biology of
FSH and LH was the cloning and expression of the
gonadotropin receptors (R). Long known to be membrane-associated receptors coupled to adenylyl cyclasemediated intracellular signalling, the cloning and
sequencing of gonadotropin-R cDNAs (McFarland et
al., 1989; Sprengel et al., 1990) confirmed that FSHR
and LHR are structurally related members of the seven
transmembrane domain G-protein associated receptor
superfamily that includes TSHR, GHR (Baumann,
1994), b-adrenoreceptor (Collins, 1993) and the
Alzheimer gene STM2 (Levy-Lahad et al., 1996). The
gene encoding the LHR protein contains 11 exons
(Segaloff et al., 1990; Segaloff and Ascoli, 1993). Variable transcription of this gene leads to a complex
pattern of about 13 gene-splice variants. The transmembrane and intracellular G-protein binding domains of
the receptor are encoded by the last exon, therefore
much of the heterogeneity is associated with the extracellular LH binding domain (Ji and Ji, 1991). This may
give rise to encoded products with different functions.
For instance, the 1.8 kb-sized LHR mRNA transcript
codes for a truncated receptor protein corresponding to
the extracellular domain of the receptor, which could
theoretically function as a plasma binding protein. The
extracellular domain of the GHR is secreted and has
been shown to modulate GH action (Baumann, 1994).
Whether this also occurs with LHR is not known. The
extracellular ligand-binding domain of FSHR is encoded by the first nine exons of the FSHR gene, giving
rise to a binding protein with high specificity for FSH.
The gene splice variants transcribed from the FSHR
gene are not as numerous as for the LHR gene (Huhtaniemi, 1994).
The first direct evidence that granulosa cells express
FSHR was obtained by autoradiographic localization
of 125I-labelled FSH binding to immature rat ovarian
sections (Zeleznik et al., 1974). Radioligand-binding
analysis confirmed the presence of specific, saturable,
high-affinity FSH binding sites in granulosa cell membranes (Nimrod et al., 1976). With the isolation of
cDNA clones encoding rat FSHR (Sprengel et al.,
1990), the tissue distribution and developmental regulation of FSHR gene expression was finally mapped
(Camp et al., 1991). That work confirmed gonad-specific expression of an abundant  2.6 kb-sized FSHR
mRNA and a less abundant 5.0 kb transcript. In
ovary, in situ hybridisation showed FSHR mRNA to
be localised exclusively to the granulosa cells of healthy
follicles, persisting throughout preovulatory follicular
development.
The presence of specific human chorionic gonadotropin (hCG)/LH binding sites on thecal/interstital
cells was also revealed by autoradiography, using 125-

hCG as a surrogate LH to specifically label hCG/LH


binding sites in rat ovarian sections (Zeleznik et al.,
1974). Importantly, it was also shown that treatment of
animals with FSH in vivo led to increased binding of
125
I-hCG to granulosa cells, providing the first direct
evidence that FSH induces the granulosa cell hCG/
LHR. Quantitative assessment of the hCG/LH receptor
on thecal-interstitial cells (Magoffin and Erickson,
1982) and induction of the hCG/LH receptor on granulosa cells by FSH (Erickson et al., 1979) were subsequently confirmed in vitro. Following the cloning of the
rat LHR (McFarland et al., 1989), multiple ovarian
LHR transcripts were identified with two major transcripts of 6.5 and  2.6 kb (Segaloff et al., 1990).
Application of in situ hybridisation showed LHR mRNAs to be located exclusively in the thecal cells of
immature follicles but present in both thecal and granulosa cells of mature, antral follicles (Camp et al., 1991).
Direct FSH induction of granulosa cell LHR mRNA
was confirmed in vitro (Piquette et al., 1991) and in
vivo (Segaloff et al., 1990) and shown to involve increased LHR gene transcription (Shi and Segaloff,
1995).

2.2. Post-receptor signaling


The post-receptor signaling systems that relay gonadotropin action into the cell nucleus rest mainly on
adenylyl cyclase, cAMP production, and activation of
protein kinase A (PKA) (Richards, 1994; Richards et
al., 1998). Tonic stimulation of immature granulosa
cells by FSH via FSHR stimulates intracellular cAMP
formation and activation of genes required for proliferation and differentiation. Late during preovulatory follicular growth, the response to FSH includes expression
of LHR, also coupled to PKA (Abell et al., 1998). Prior
to onset of the mid-cycle LH surge that triggers ovulation (see below) tonic stimulation of mature granulosa
cells by LH via LHR mimics the action of FSH. Higher
(surge) concentrations of LH dramatically up-regulate
PKA signaling, also increasing inositol lipid hydrolysis
and activation of protein kinase C, altering the expression panel of other genes that co-ordinate final stages of
follicular development and ovulation (Robker and
Richards, 1998). Little information exists about the
transcription regulators that mediate cellular responses
to these signals. However, C/EBP beta is a critical
downstream target of G-protein-coupled LHR signaling (Pall et al., 1997; Sterneck et al., 1997). Post-receptor signaling pathways that impact on gonadotropin
action include the serine/threonine kinase mothers
against dpp (MAD) cell signaling pathway (Li et al.,
1997), which is activated by members of the transforming growth factorb (TGFb) superfamily (Massague,
1998).

S.G. Hillier / Molecular and Cellular Endocrinology 179 (2001) 3946

3. Relative roles of FSH and LH


During adulthood when ovulatory menstrual cycles
occur, it is estimated to take 2 3 months for a primordial follicle to develop to the point of ovulation (Gougeon, 1996). Although it is a continuum, the life-cycle
of a preovulatory follicle can be broken down into
three successive phases initiation, which occurs from
birth to senescence independent of gondotropic support; FSH-dependent progression, requiring tonic stimulation by FSH; and LH-responsive maturation, when
FSH-induced genes fall under LH control, leading to
oestrogen secretion and ovulation (Zeleznik and Hillier,
1984; Hillier, 1994; McGee and Hsueh, 2000).
Ovulation is triggered by the mid-cycle LH surge,
itself triggered by the sustained high circulating level of
estradiol produced by the preovulatory follicle. The
surge initiates a gene cascade in granulosa cells, the
products of which initiate luteinization, signal the egg
to commence meiotic maturation and lead to rupture of
the follicle wall (Conti et al., 1998; Richards et al.,
1998). The inductive action of FSH on granulosa cell
development that facilitates this response to LH includes alterations in the post-receptor mechanisms
through which granulosa cells respond to gonadotropins and other extracellular factors. Signalling
via Ca2 + /inositol lipid hydrolysis and tyrosine kinase
pathways are involved (Morris et al., 1985, Lamm et
al., 1999) but the primary intracellular drive comes
from cAMP (Richards et al., 1998).
During late granulosa cell maturity induced by FSH,
LH has a relatively stronger effect than FSH on cAMP
formation in vitro (Yong et al., 1992a,b; Richards et
al., 1998), suggesting that LHR density is relatively
greater than FSHR density or that the LHR is more
effectively coupled to cAMP generation. The ovulationinducing LH surge triggers the expression of a panel of
high-tone cAMP response genes that lead to terminal
granulosa cell differentiation and follicular rupture.
Those genes encode pro-inflammatory factors such as
interleukin-1b, interleukin-1bR (Adashi, 1998a) and
prostaglandin endoperoxide synthase-2 (COX-2, Morris
and Richards, 1995); angiogenic factors such as VEGF
(Ravindranath et al., 1992); proteases necessary for
tissue remodelling (Liu et al. 1998); neurotrophin receptors such as TrkA (Mayerhofer et al., 1996); and agents
of progesterone production (steroidogenic acute regulatory protein-StAR) (Ronen-Fuhrmann et al., 1998) and
action (progesterone receptors-PR) (Natraj and
Richards, 1993; Park-Sarge and Sarge, 1995; Duffy et
al., 1996; Clemens et al., 1998). Intriguingly, the LH
surge also induces 11b-hydroxysteroid dehydrogenase
type 1 (11bHSD1) and simultaneously down-regulates
11bHSD2 expression in granulosa cells of preovulatory
follicles (Tetsuka et al., 1997, 1999b). The stimulatory
action of LH on granulosa cell 11bHSD1 gene expres-

41

sion in vitro is mimicked by interleukin-1b (Tetsuka et


al., 1999a). Since 11bHSD1 functions mainly as a C11
reductase (i.e. converts cortisone to cortisol) whereas
11bHSD2 is an dehydrogenase (inactivates cortisol to
cortisone, Stewart, 1996), the changes induced by LH
favour local accumulation of anti-inflammatory cortisol
at a time when rapid healing of the ruptured surface is
required to rapidly restore normal ovarian function
(Hillier and Tetsuka, 1998). LH also simultaneously
suppresses granulosa cell division (Yong et al., 1992a).
Thus, along with the pro/anti-inflammatory, angiogenic
and proteolytic changes that lead to follicular rupture,
the follicle ceases to grow, luteinizes and commences
secretion of progesterone.

4. The follicular paracrine system


A striking feature of the ovaries as endocrine targets
is the shifting pattern of gonadotropin sensitivity displayed by individual ovarian cell types. Much of this
can be explained by the local production of polypeptide
growth/differentiation factors, steroids and other extracellular substances that mediate and modulate gonadotropin action. Some of the better known
participants in this paracrine dialogue will now be
discussed.

4.1. Paracrine growth/differentiation factors


The best characterized ovarian growth factors to-date
are insulin-like growth factors I and II (IGF-I, IGF-II)
(Adashi, 1998b) and members of the TGFb superfamily
(Gaddy-Kurten et al., 1995) such as inhibin and activin
(Hillier, 1991) and growth differentiation factor 9
(GDF-9) (Elvin et al., 1999). Other substances implicated are epidermal growth factor (EGF)/transforming
growth factora/TGFa (Foghi et al., 1998; Lamm et al.,
1999), fibroblast growth factors (FGFs) (Reynolds and
Redmer, 1998), vascular endothelial growth factor
(VEGF, Ravindranath et al., 1992; Laitinen et al.,
1997), nerve growth factor (NGF) (Mayerhofer et al.,
1996), scatter factor (SCF)/hepatocyte growth factor
(HGF, Peluso, 1997; Parrott and Skinner, 1998a), keratinocyte growth factor (KGF, Parrott and Skinner,
1998b), tumour necrosis factora (TNFa, Terranova and
Rice, 1997) and many other cytokines (Adashi, 1998a)
and chemokines (Karstrom-Encrantz et al., 1998).
Platelet-derived growth factor (PDGF), a major
serum mitogen for cells of mesenchymal origin (Heldin,
1992), has been shown to stimulate thecal cell mitosis in
vitro an effect that is augmented by the additional
presence of other growth factors such as EGF (May et
al., 1992). Intriguingly, the three-dimensional structures
of gonadotropins bear structural similarities to PDGF
and other members of the so-called cystine knot family

42

S.G. Hillier / Molecular and Cellular Endocrinology 179 (2001) 3946

of growth factors (Lapthorn et al., 1994, 1995). This


family of growth factors is presently known to include
NGF and TGFbs, and based on sequence homology, at
least thirty other proteins including the three pituitary
glycoprotein hormones (FSH, LH and TSH), activins,
inhibins, anti-Mu llerian hormone (AMH), GDF-9,
VEGF and several bone morphogenetic proteins. Characteristically, they exist as homo- or heterodimers, the
protomers of which predominantly consist of b-strand
secondary structure with a characteristic clustering of
three cystine bridges known as the cystine-knot motif
(Lapthorn et al., 1995). Gonadotropins differ from
most other known cystine-knot regulatory factors in
that they signal through G-protein coupled receptors
instead of receptor serine/tyrosine kinases (Massague,
1998). However, the remarkable structural similarities
between LH, hCG, FSH and other members of the
cystine-knot growth factor family suggests that gonadotrophins have, or in an evolutionary sense once
had, specific growth regulatory functions (Lapthorn et
al., 1995).

4.2. Paracrine regulation of steroid synthesis


During the human menstrual cycle, LH released by
the pituitary gland provides the major endocrine drive
to thecal androgen synthesis, which is fundamental to
folliculogenesis. Among many locally produced factors
capable of modulating this action of LH, the IGFs and
the inhibins/activins ar of particular relevance and will
be considered further here.

4.2.1. IGFs
Ovarian follicular cells varyingly express genes encoding IGF-I, IGF-II and IGF-binding proteins (IGFBPs, Adashi, 1998b), the expression of which are
developmentally regulated (el-Roeiy et al., 1994). Thecal cells from normal human ovaries have been shown
to possess receptors for insulin and IGFs (Poretsky et
al., 1985; Hernandez et al., 1988), and both insulin
(Barbieri et al., 1986) and IGF-I (Hillier et al., 1991c;
Nahum et al., 1995) stimulate thecal/stromal androgen
synthesis in vitro. In the rat ovary, FSH stimulates the
production of IGF-I by granulosa cells, suggesting a
paracrine role for granulosa cell-derived IGF-I in the
regulation of thecal androgen synthesis (Hernandez et
al., 1988; Adashi et al., 1991). The relevance of the
paracrine IGF-I hypothesis to the human ovary was
initially challenged by observations that human granulosa cells do not express the IGF-I gene. On the other
hand, human granulosa cells do express IGF-II (Hernandez et al., 1992; Zhou and Bondy, 1993), which is
equipotent with IGF-I at the level of thecal androgen
synthesis, and therefore likely to have a physiological
paracrine role in regulating androgen production by
human ovaries (Nahum et al., 1995). IGF-BPs pro-

duced by ovarian cells and may also participate in the


local mediation/modulation of gonadotrophin action
(Ling et al., 1993; Piferrer et al., 1997).

4.2.2. Inhibins and acti6ins


Activins and inhibins produced by granulosa cells
have potent modulatory actions on thecal androgen
synthesis: activin being inhibitory and inhibin stimulatory (Hsueh et al., 1987; Hillier et al., 1991b,c). Based
on patterns of inhibin/activin subunit mRNA expression in vitro (Turner et al., 1989; Pei et al., 1991) and in
vivo (Meunier et al., 1989; Schwall et al., 1990; Roberts
et al., 1993) and measurements of immunoreactive inhibin production by granulosa cells in vitro (Hillier et
al., 1991a), activin predominates in immature follicles
where it promotes FSH-induced mitosis and FSH induced steroidogenic differentiation (Miro and Hillier,
1996). Inhibin and the activin-binding protein follistatin
are produced in progressively greater amounts relative
to activin by granulosa cells as follicles mature
(Nakatani et al., 1991). Thereby the stimulatory action
of inhibin on thecal androgen synthesis gains sway
during late preovulatory follicular development, when
androgen is required in increasing amounts as a substrate for oestrogen synthesis (Hillier, 1991). Although
physiological concentrations of insulin, IGF-I and IGFII can stimulate thecal androgen production, inhibin
greatly enhances the actions of all three factors in vitro
(Nahum et al., 1995). Thus regardless of the contributions made by insulin or IGFs to the control of follicular androgen production, paracrine regulation by
inhibin could be of overriding importance during preovulatory follicular development. Antral follicles too
immature to secrete estrogen do not need androgen as
an aromatase precursor (Hillier et al., 1994). At such
early stages of development, androgen synthesis may be
suppressed due to the preponderance of activin relative
to inhibin, possibly aided by granulosa-derived IGFBPs that sequester and inhibit the actions of IGFs
(Ling et al., 1993).
4.3. Paracrine steroid action
Steroids produced by ovarian cells have critically
important roles in the regulation of ovarian function.
As secreted ovarian hormones they participate in longloop feedback regulation of pituitary gonadotropin secretion, while within the ovaries they serve autocrine
and paracrine roles that contribute to normal preovulatory development and function.

4.3.1. Androgens
Androgens are synthesised in thecal cells through
cell-specific expression of P45017a (CYP17), which is
under LH control (Smyth et al., 1993). Granulosa cells
express androgen receptors (AR) throughout antral and

S.G. Hillier / Molecular and Cellular Endocrinology 179 (2001) 3946

early antral development, permitting paracrine androgenic stimulation (Smyth et al., 1995; Tetsuka et al.,
1995; Tetsuka and Hillier, 1996). The main AR-mediated effect in granulosa cells is up-regulation of cAMP
formation, possibly through inhibition of cAMP
metabolism (Hillier and de Zwart, 1982). Hormonally
regulated phosphodiesterase 4A (PDE4A, Conti et al.,
1998) is a potential candidate for regulation by AR but
this remains to be tested. Amplification of FSH-induced
PKA signalling by androgen appears to a means of
sensitising granulosa cells to tonic stimulation by FSH.
During late preovulatory development, transcription of
the granulosa AR gene and AR protein levels decline
(Tetsuka et al., 1995; Tetsuka and Hillier 1996; Hillier
et al., 1997), which may serve to diminish granulosa cell
responsiveness to gonadotrophins and delay terminal
differentiation (luteinization) until timeous exposure to
a full-blown ovulation-inducing LH surge (Hillier and
Tetsuka, 1997).

4.3.2. Estrogens
Estrogen production is the hallmark of preovulatory
follicular development, reflecting FSH induced expression of cytochrome P450arom (CYP19) that converts
androgen to estrogen in granulosa cells (Hillier et al.,
1994). Granulosa cells also express estrogen receptors
(ER), which may mediate autocrine oestrogen action
within the granulosa cell layer. The effects of estrogen
on granulosa cells are similar to those of androgen,
serving to amplify overall actions of FSH (Richards,
1994). Classic ER (ERa) may not fully explain estrogen
action at this level since granulosa cells more abundantly express a second ER isoform, ERb (Enmark et
al., 1997; Sar and Welsch, 1999). Female ERa knockout mice are infertile (Korach et al., 1996), whereas
ERb / females are fertile and exhibit normal
sexual behaviour. However, they have fewer and
smaller litters than wild-type mice, and superovulation
experiments indicate that this reduction in fertility is the
result of reduced ovarian efficiency (Krege et al., 1998).
Developmental regulation of ER is not well studied but
both isoforms are down-regulated by the ovulation
inducing gonadotropin surge (Byers et al., 1997;
Tetsuka et al., 1998b).
4.3.3. Progesterone
Progesterone is produced in increasing amounts by
differentiated granulosa cells through FSH induction of
cytochrome P450scc (CYP11A), which catalyses ratelimiting conversion of cholesterol to pregnenolone
(Richards et al., 1998). Progesterone receptors (PR) do
not appear in granulosa cells until onset of the gonadotrophin surge (Natraj and Richards, 1993; ParkSarge and Mayo, 1994; Park-Sarge and Sarge, 1995).
PR is then transiently expressed before ovulation and
reappears in the corpus luteum. The role of the granu-

43

losa cell PR in ovarian function is uncertain, but


knockout studies in mice demonstrate the gene to be
vital to ovulation (Lydon et al., 1995). Expression of
PR is temporally to expression of COX-2, the inducible
form of prostaglandin synthase that is also essential to
ovulation (Lim et al., 1997).

4.3.4. Glucocorticoids
Ovarian cells do not express cytochromes P450c21
(CYP21B) or P45011b, (CYP11B1), which are necessary
for corticosteroid synthesis (Omura and Morohashi,
1995). Consequently, glucocorticoids are not true
ovarian paracrines. The ovaries do however express
11bHSD isoforms (Tetsuka et al., 1997, 1999b) that
interconvert cortisol and cortisone (Michael et al.,
1997) and thereby gate the access of cortisol to ovarian
corticosteroid receptors (GR). As discussed above, the
pattern of 11bHSD isoform expression induced by the
ovulation-inducing gonadotropin surge favors cortisol
accumulation and hence activation of GR in the preovulatory follicle (Harlow et al., 1997; Yding-Andersen
et al., 1999; Yong et al., 2000). The function of ovarian
cortisol at the time of ovulation remains unknown.
However, as a natural anti-inflammatory agent it is an
obvious potential participant in the ovulation associated injury-repair process (Hillier and Tetsuka, 1998).
References
Abell, A.N., McCormick, D.J., Segaloff, D.L., 1998. Certain activating mutations within helix 6 of the human luteinizing hormone
receptor may be explained by alterations that allow transmembrane regions to activate Gs. Mol. Endocrinol. 12, 1857 1869.
Adashi, E.Y., 1998a. The potential role of interleukin-1 in the ovulatory process: an evolving hypothesis. Mol. Cell Endocrinol. 140,
77 81.
Adashi, E.Y., 1998b. The IGF family and folliculogenesis. J. Reprod.
Immunol. 39, 13 19.
Adashi, E.Y., Resnick, C.E., Hurwitz, A., Ricciarelli, E., Hernandez,
E.R., Roberts, C.T., Leroith, D., Rosenfeld, R., 1991. Insulin-like
growth factors: the ovarian connection. Hum. Reprod. 6, 1213
1219.
Barbieri, R.L., Makris, A., Randall, R.W., Daniels, G., Kistner,
R.W., Ryan, K.J., 1986. Insulin stimulates androgen accumulation in incubations of ovarian stroma obtained from women with
hyperandrogenism. J. Clin. Endocrinol. Metab. 62, 904 910.
Baumann, G., 1994. Growth Hormone-binding protein: state of the
art. J. Endocrinol. 141, 1 6.
Byers, M., Kuiper, G.G., Gustafsson, J.A., Park-Sarge, O.K., 1997.
Estrogen receptor-beta mRNA expression in rat ovary: down-regulation by gonadotropins. Mol. Endocrinol. 11, 172 182.
Camp, T.A., Rahal, J.O., Mayo, K.E., 1991. Cellular localization and
hormonal regulation of follicle-stimulating hormone and luteinizing hormone receptor messenger RNAs in the rat ovary. Mol.
Endocrinol. 5, 1405 1417.
Clemens, J.W., Robker, R.L., Kraus, W.L., Katzenellenbogen, B.S.,
Richards, J.S., 1998. Hormone induction of progesterone receptor
(PR) messenger ribonucleic acid and activation of PR promoter
regions in ovarian granulosa cells: evidence for a role of cyclic
adenosine 3%,5%-monophosphate but not estradiol. Mol. Endocrinol. 12, 1201 1214.

44

S.G. Hillier / Molecular and Cellular Endocrinology 179 (2001) 3946

Collins, S., 1993. Recent perspectives on the molecular structure and


regulation of the beta 2-adrenoceptor. Life Sci. 52, 2083 2091.
Conti, M., Andersen, C.B., Richard, F.J., Shitsukawa, K., Tsafriri,
A., 1998. Role of cyclic nucleotide phosphodiesterases in resumption of meiosis. Mol. Cell Endocrinol. 145, 9 14.
Elvin, J.A., Clark, A.T., Wang, P., Wolfman, N.M., Matzuk, M.M.,
1999. Paracrine actions of growth differentiation factor-9 in the
mammalian ovary. Mol. Endocrinol. 13, 1035 1048.
Duffy, D.M., Molskness, T.A., Stouffer, R.L., 1996. Progesterone
receptor messenger ribonucleic acid and protein in luteinized
granulosa cells of rhesus monkeys are regulated in vitro by
gonadotropins and steroids. Biol. Reprod. 54, 888 895.
el-Roeiy, A., Chen, X., Roberts, V.J., Shimasakai, S., Ling, N.,
LeRoith, D., Roberts, C.T. Jr, Yen, S.S., 1994. Expression of the
genes encoding the insulin-like growth factors (IGF-I and II), the
IGF and insulin receptors, and IGF-binding proteins-1-6 and the
localization of their gene products in normal and polycystic ovary
syndrome ovaries. J. Clin. Endocrinol. Metab. 78, 1488 1496.
Enmark, E., Pelto-Huikko, M., Grandien, K., Lagercrantz, S., Lagercrantz, J., Fried, G., Nordenskjold, M., Gustafsson, J.A., 1997.
Human estrogen receptor b-gene structure, chromosomal localization, and expression pattern. J. Clin. Endocrinol. Metab. 82,
4258 4265.
Erickson, G.F., Wang, C., Hsueh, A.J., 1979. FSH induction of
functional LH receptors in granulosa cells cultured in a chemically
defined medium. Nature 279, 336 338.
Foghi, A., Teerds, K.J., van der Donk, H., Moore, N.C., Dorrington,
J., 1998. Induction of apoptosis in thecal/interstitial cells: action
of transforming growth factor (TGF) alpha plus TGF beta on
bcl-2 and interleukin-1 beta-converting enzyme. J. Endocrinol.
157, 489 494.
Gaddy-Kurten, D., Tsuchida, K., Vale, W., 1995. Activins and the
receptor serine kinase superfamily. Recenti Prog. Horm. Res. 50,
109 129.
Gougeon, A., 1996. Regulation of ovarian follicular development in
primates: facts and hypotheses. Endocr. Rev. 17, 121 155.
Harlow, C.R., Jenkins, J.M., Winston, R.M., 1997. Increased follicular fluid total and free cortisol levels during the luteinizing hormone surge. Fertil. Steril. 68, 48 53.
Heldin, C.H., 1992. Structural and functional studies on plateletderived growth factor. EMBO J. 11, 4251 4259.
Hernandez, E.R., Resnick, C.E., Svoboda, M.E., Van Wyk, J.J.,
Payne, D.W., Adashi, E.Y., 1988. Somatomedin-C/insulin-like
growth factor I as an enhancer of androgen biosynthesis by
cultured rat ovarian cells. Endocrinology 122, 1603 1612.
Hernandez, E.R., Hurwitz, A., Vera, A., Pellicer, A., Adashi, E.Y.,
Leroith, D., Roberts, C.T. Jr, 1992. Expression of the genes
encoding the insulin-like growth factors and their receptors in the
human ovary. J. Clin. Endocrinol. Metab. 74, 419 425 published
erratum appears in J Clin Endocrinol Metab 1993
May;76(5):1343.
Hillier, S.G., 1991. Regulatory functions for inhibin and activin in
human ovaries. J. Endocrinol. 13, 171 175.
Hillier, S.G., 1994. Current concepts of the roles of follicle stimulating hormone and luteinizing hormone in folliculogenesis. Hum.
Reprod. 9, 188 191.
Hillier, S.G., de Zwart, F.A., 1982. Androgen/antiandrogen modulation of cyclic AMP-induced steroidogenesis during granulosa cell
differentiation in tissue culture. Mol. Cell. Endocrinol. 28, 347
361.
Hillier, S.G., Tetsuka, M., 1997. Role of androgens in follicle maturation and atresia. Baillieres. Clin. Obstet. Gynaecol. 11, 249 260.
Hillier, S.G., Tetsuka, M., 1998. An anti-inflammatory role for
glucocorticoids in the ovaries? J. Reprod. Immunol. 39, 21 27.
Hillier, S.G., Wickings, E.J., Illingworth, P.I., Yong, E.L., Reichert,
L.E. Jr, Baird, D.T., McNeilly, A.S., 1991a. Control of immunoactive inhibin production by human granulosa cells. Clin.
Endocrinol. (Oxf) 35, 71 78.

Hillier, S.G., Yong, E.L., Illingworth, P.J., Baird, D.T., Schwall,


R.H., Mason, A.J., 1991b. Effect of recombinant inhibin on
androgen synthesis in cultured human thecal cells. Mol. Cell
Endocrinol. 75, R1 R6 published erratum appears in Mol Cell
Endocrinol 1991 Aug;79(1 3):177.
Hillier, S.G., Yong, E.L., Illingworth, P.J., Baird, D.T., Schwall,
R.H., Mason, A.J., 1991c. Effect of recombinant activin on
androgen synthesis in cultured human thecal cells. J. Clin. Endocrinol. Metab. 72, 1206 1211.
Hillier, S.G., Whitelaw, P.F., Smyth, C.D., 1994. Follicular oestrogen
synthesis: the two-cell, two-gonadotrophin model revisited. Mol.
Cell Endocrinol. 100, 51 54.
Hillier, S.G., Tetsuka, M., Fraser, H.M., 1997. Location and developmental regulation of androgen receptor in primate ovary. Hum.
Reprod. 12, 107 111.
Hsueh, A.J., Dahl, K.D., Vaughan, J., Tucker, E., Rivier, J., Bardin,
C.W., Vale, W., 1987. Heterodimers and homodimers of inhibin
subunits have different paracrine action in the modulation of
luteinizing hormone-stimulated androgen biosynthesis. Proc. Natl.
Acad. Sci. USA 84, 5082 5086.
Huhtaniemi, I., 1994. Fetal testis-a very special endocrine organ. Eur.
J. Endocrinol. 130, 25 31.
Ji, I., Ji, T.H., 1991. Exons 1-10 of the rat LH receptor encode a
high-affinity hormone binding site and exon 11 encodes G-protein
modulation and a potential second hormone binding site. Endocrinology 128, 2648 2650.
Karstrom-Encrantz, L., Runesson, E., Bostrom, E.K., Brannstrom,
M., 1998. Selective presence of the chemokine growth-regulated
oncogene alpha (GROalpha) in the human follicle and secretion
from cultured granulosa lutein cells at ovulation. Mol. Hum.
Reprod. 4, 1077 1083.
Korach, K.S., Couse, J.F., Curtis, S.W., Washburn, T.F., Lindzey, J.,
Kimbro, K.S., Eddy, E.M., Migliaccio, S., Snedeker, S.M.,
Lubahn, D.B., Schomberg, D.W., Smith, E.P., 1996. Estrogen
receptor gene disruption: molecular characterization and experimental and clinical phenotypes. Recent Prog. Horm. Res. 51,
159 186.
Krege, J.H., Hodgin, J.B., Couse, J.F., Enmark, E., Warner, M.,
Mahler, J.F., Sar, M., Korach, K.S., Gustafsson, J.A., Smithies,
O., 1998. Generation and reproductive phenotypes of mice lacking estrogen receptor beta. Proc. Natl. Acad. Sci. USA 95,
15677 15682.
Laitinen, M., Ristimaki, A., Honkasalo, M., Narko, K., Paavonen,
K., Ritvos, O., 1997. Differential hormonal regulation of vascular
endothelial growth factors VEGF, VEGF-B, and VEGF-C messenger ribonucleic acid levels in cultured human granulosa-luteal
cells. Endocrinology 138, 4748 4756.
Lamm, M.L., Rajagopalan-Gupta, R.M., Hunzicker-Dunn, M., 1999.
Epidermal growth factor-induced heterologous desensitization of
the luteinizing hormone/choriogonadotropin receptor in a cell-free
membrane preparation is associated with the tyrosine phosphorylation of the epidermal growth factor receptor. Endocrinology
140, 29 36.
Lapthorn, A.J., Harris, D.C., Littlejohn, A., Lustbader, J.W.,
Canfield, R.E., Machin, K.J., Morgan, F.J., Isaacs, N.W., 1994.
Crystal structure of human chorionic gonadotropin. Nature 369,
455 461 see comments.
Lapthorn, A.J., Janes, R.W., Isaacs, N.W., Wallace, B.A., 1995.
Cystine nooses and protein specificity [letter]. Nat. Struct. Biol. 2,
266 268.
Levy-Lahad, E., Poorkaj, P., Wang, K., Fu, Y.H., Oshima, J.,
Mulligan, J., Schellenberg, G.D., 1996. Genomic structure and
expression of STM2, the chromosome 1 familial Alzheimer disease gene. Genomics. 34, 198 204.
Li, M., Li, J., Hoodless, P.A., Tzukazaki, T., Wrana, J.L., Attisano,
L., Tsang, B.K., 1997. Mothers against decapentaplegic-related
protein 2 expression in avian granulosa cells is up-regulated by

S.G. Hillier / Molecular and Cellular Endocrinology 179 (2001) 3946


transforming growth factor beta during ovarian follicular development. Endocrinology 138, 3659 3665.
Lim, H., Paria, B.C., Das, S.K., Dinchuk, J.E., Langenbach, R.,
Trzaskos, J.M., Dey, S.K., 1997. Multiple female reproductive
failures in cyclooxygenase 2-deficient mice. Cell 91, 197 208.
Ling, N.C., Liu, X.J., Malkowski, M., Guo, Y.L., Erickson, G.F,
Shimasaki, S., 1993. Structural and functional studies of insulinlike growth factor binding proteins in the ovary. Growth Regul. 3,
70 74.
Liu, K., Wahlberg, P., Ny, T., 1998. Coordinated and cell-specific
regulation of membrane type matrix metalloproteinase 1 (MT1MMP) and its substrate matrix metalloproteinase 2 (MMP-2) by
physiological signals during follicular development and ovulation.
Endocrinology 139, 4735 4738.
Lydon, J.P., DeMayo, F.J., Funk, C.R., Mani, S.K., Hughes, A.R.,
Montgomery, C.A. Jr, Shyamala, G., Conneely, O.M., OMalley,
B.W., 1995. Mice lacking progesterone receptor exhibit
pleiotropic reproductive abnormalities. Genes Dev. 9, 2266 2278.
McGee, E.A., Hsueh, A.J., 2000. Initial and cyclic recruitment of
ovarian follicles. Endocr. Rev. 21, 200 214.
Magoffin, D.A., Erickson, G.F., 1982. Primary culture of differentiating ovarian androgen-producing cells in defined medium. J. Biol.
Chem. 257, 4507 4513.
Massague, J., 1998. TGF-b signal transduction. Annu. Rev. Biochem.
67, 753 791.
May, J.V., Bridge, A.J., Gotcher, E.D., Gangrade, B.K., 1992. The
regulation of porcine theca cell proliferation in vitro: synergistic
actions of epidermal growth factor and platelet-derived growth
factor. Endocrinology 131, 689 697.
Mayerhofer, A., Dissen, G.A., Parrott, J.A., Hill, D.F., Mayerhofer,
D., Garfield, R.E., Costa, M.E., Skinner, M.K., Ojeda, S.R.,
1996. Involvement of nerve growth factor in the ovulatory cascade: trkA receptor activation inhibits gap junctional communication between thecal cells. Endocrinology 137, 5662 5670.
McFarland, K.C., Sprengel, R., Phillips, H.S., Kohler, M., Rosemblit, N., Nikolics, K., Segaloff, D.L., Seeburg, P.H., 1989.
Lutropin-choriogonadotropin receptor: an unusual member of the
G protein-coupled receptor family. Science 245, 494 499.
Meunier, H., Roberts, V.J., Sawchenko, P.E., Cajander, S.B., Hsueh,
A.J., Vale, W., 1989. Periovulatory changes in the expression of
inhibin alpha-, beta A-, and beta B-subunits in hormonally induced immature female rats. Mol. Endocrinol. 3, 2062 2069.
Michael, A.E., Evagelatou, M., Norgate, D.P., Clarke, R.J., Antoniw, J.W., Stedman, B.A., Brennan, A., Welsby, R., Bujalska,
I., Stewart, P.M., Cooke, B.A., 1997. Isoforms of 11b-hydroxysteroid dehydrogenase in human granulosa lutein cells. Mol.
Cell Endocrinol. 132, 43 52.
Miro, F., Hillier, S.G., 1996. Modulation of granulosa cell deoxyribonucleic acid synthesis and differentiation by activin. Endocrinology 137, 464 468.
Morris, J.K., Richards, J.S., 1995. Luteinizing hormone induces
prostaglandin endoperoxide synthase-2 and luteinization in vitro
by A-kinase and C-kinase pathways. Endocrinology 136, 1549
1558.
Nahum, R., Thong, K.J., Hillier, S.G., 1995. Metabolic regulation of
androgen production by human thecal cells in vitro. Hum. Reprod. 10, 75 81.
Nakatani, A., Shimasaki, S., Depaolo, L.V., Erickson, G.F., Ling,
N., 1991. Cyclic changes in follistatin messenger ribonucleic acid
and its protein in the rat ovary during the estrous cycle. Endocrinology 129, 603 611.
Natraj, U., Richards, J.S., 1993. Hormonal regulation, localization,
and functional activity of the progesterone receptor in granulosa
cells of rat preovulatory follicles. Endocrinology 133, 761 769.
Nimrod, A., Erickson, G.F., Ryan, K.J., 1976. A specific FSH
receptor in rat granulosa cells: properties of binding in vitro.
Endocrinology 98, 56 64.

45

Omura, T., Morohashi, K., 1995. Gene regulation of steroidogenesis.


J. Steroid. Biochem. Mol. Biol. 53, 19 25.
Pall, M., Hellberg, P., Brannstrom, M., Mikuni, M., Peterson, C.M.,
Sundfeldt, K., Norden, B., Hedin, L., Enerback, S., 1997. The
transcription factor C/EBP-beta and its role in ovarian function;
evidence for direct involvement in the ovulatory process. EMBO
J. 16, 5273 5279.
Park-Sarge, O.K., Mayo, K.E., 1994. Regulation of the progesterone
receptor gene by gonadotropins and cyclic adenosine 3%,5%monophosphate in rat granulosa cells. Endocrinology 134, 709
718.
Park-Sarge, O.K., Sarge, K.D., 1995. Cis-regulatory elements conferring cyclic 3,5-adenosine monophosphate responsiveness of the
progesterone receptor gene in transfected rat granulosa cells.
Endocrinology 136, 5430 5437.
Parrott, J.A., Skinner, M.K., 1998a. Thecal cell-granulosa cell interactions involve a positive feedback loop among keratinocyte
growth factor, hepatocyte growth factor, and Kit ligand during
ovarian follicular development. Endocrinology 139, 2240 2245.
Parrott, J.A., Skinner, M.K., 1998b. Developmental and hormonal
regulation of keratinocyte growth factor expression and action in
the ovarian follicle. Endocrinology 139, 228 235.
Pei, L., Dodson, R., Schoderbek, W.E., Maurer, R.A., Mayo, K.E.,
1991. Regulation of the alpha inhibin gene by cyclic adenosine
3%,5%-monophosphate after transfection into rat granulosa cells.
Mol. Endocrinol. 5, 521 534.
Peluso, J.J., 1997. Putative mechanism through which N-cadherinmediated cell contact maintains calcium homeostasis and thereby
prevents ovarian cells from undergoing apoptosis. Biochem. Pharmacol. 54, 847 853.
Piferrer, F., Li, D., Shimasaki, S., Erickson, G.F., 1997. Transforming growth factor-a stimulates insulin-like growth factor binding
protein-4 (IGFBP-4) expression and blocks follicle-stimulating
hormone regulation of IGFBP-4 production in rat granulosa cells.
Mol. Cell Endocrinol. 133, 9 17.
Piquette, G.N., LaPolt, P.S., Oikawa, M., Hsueh, A.J., 1991. Regulation of luteinizing hormone receptor messengerribonucleic acid
levels by gonadotropins, growthfactors, and gonadotropin-releasing hormone incultured rat granulosa cells. Endocrinology 128,
2449 2456.
Poretsky, L., Grigorescu, F., Seibel, M., Moses, A.C., Flier, J.S.,
1985. Distribution and characterization of insulin and insulin-like
growth factor I receptors in normal human ovary. J. Clin. Endocrinol. Metab. 61, 728 734.
Ravindranath, N., Little-Ihrig, L., Phillips, H.S., Ferrara, N.,
Zeleznik, A.J., 1992. Vascular endothelial growth factor messenger ribonucleic acid expression in the primate ovary. Endocrinology 131, 254 260.
Recombinant Human FSH Product Development Group, 1998. Recombinant follicle stimulating hormon: development of the first
biotechnology product for the treatment of infertility. Hum. Reprod. Update. 4, 862 881.
Reynolds, L.P., Redmer, D.A., 1998. Expression of the angiogenic
factors, basic fibroblast growth factor and vascular endothelial
growth factor, in the ovary. J. Anim. Sci. 76, 1671 1681.
Richards, J.S., 1994. Hormonal control of gene expression in the
ovary. Endocr. Rev. 15, 725 751.
Richards, J.S., Russell, D.L., Robker, R.L., Dajee, M., Alliston,
T.N., 1998. Molecular mechanisms of ovulation and luteinization.
Mol. Cell Endocrinol. 145, 47 54.
Roberts, V.J., Barth, S., el-Roeiy, A., Yen, S.S., 1993. Expression of
inhibin/activin subunits and follistatin messenger ribonucleic acids
and proteins in ovarian follicles and the corpus luteum during the
human menstrual cycle. J. Clin. Endocrinol. Metab. 77, 1402
1410.
Robker, R.L., Richards, J.S., 1998. Hormone-induced proliferation
and differentiation of granulosa cells: a coordinated balance of

46

S.G. Hillier / Molecular and Cellular Endocrinology 179 (2001) 3946

the cell cycle regulators cyclin D2 and p27Kip1. Mol. Endocrinol.


12, 924 940.
Ronen-Fuhrmann, T., Timberg, R., King, S.R., Hales, K.H., Hales,
D.B., Stocco, D.M., Orly, J., 1998. Spatio-temporal expression
patterns of steroidogenic acute regulatory protein (StAR) during
follicular development in the rat ovary. Endocrinology 139, 303
315.
Sar, M., Welsch, F., 1999. Differential expression of estrogen receptor-beta and estrogen receptor- alpha in the rat ovary. Endocrinology 140, 963 971.
Schwall, R.H., Mason, A.J., Wilcox, J.N., Bassett, S.G., Zeleznik,
A.J., 1990. Localization of inhibin/activin subunit mRNAs within
the primate ovary. Mol. Endocrinol. 4, 75 79.
Segaloff, D.L., Ascoli, M., 1993. The lutropin/choriogonadotropin
receptor 4 years later. Endocr. Rev. 14, 324 347.
Segaloff, D.L., Sprengel, R., Nikolics, K., Ascoli, M., 1990. Structure
of the lutropin/choriogonadotropin receptor. Recent. Prog.
Horm. Res. 46, 261 301 discusion.
Shi, H., Segaloff, D.L., 1995. A role for increased lutropin/choriogonadotropin receptor (LHR) gene transcription in the follitropinstimulated induction of the LHR in granulosa cells. Mol.
Endocrinol. 9, 734 744.
Smyth, C.D, Miro, F., Whitelaw, P.F., Howles, C.M., Hillier, S.G.,
1993. Ovarian thecal/interstitial androgen synthesis is enhanced
by a follicle-stimulating hormone-stimulated paracrine mechanism. Endocrinology 133, 1532 1538.
Smyth, C.D., Miro, F., Howles, C.M., Hillier, S.G., 1995. Effect of
luteinizing hormone on follicle stimulating hormone-activated
paracrine signalling in rat ovary. Hum. Reprod. 10, 33 39.
Sprengel, R., Braun, T., Nikolics, K., Segaloff, D.L., Seeburg, P.H.,
1990. The testicular receptor for follicle stimulating hormone:
structure and functional expression of cloned cDNA. Mol. Endocrinol. 4, 525 530.
Sterneck, E., Tessarollo, L., Johnson, P.F., 1997. An essential role for
C/EBPb in female reproduction. Genes Dev. 11, 2153 2162.
Stewart, P.M., 1996. 11 b-Hydroxysteroid dehydrogenase: implications for clinical medicine. Clin. Endocrinol. (Oxf) 44, 493 499.
Terranova, P.F., Rice, V.M., 1997. Review: cytokine involvement in
ovarian processes. Am. J. Reprod. Immunol. 37, 50 63.
Tetsuka, M., Hillier, S.G., 1996. Androgen receptor gene expression
in rat granulosa cells: the role of follicle-stimulating hormone and
steroid hormones. Endocrinology 137, 4392 4397.
Tetsuka, M., Whitelaw, P.F., Bremner, W.J., Millar, M.R., Smyth,
C.D., Hillier, S.G., 1995. Developmental regulation of androgen
receptor in rat ovary. J. Endocrinol. 145, 535 543.

Tetsuka, M., Thomas, F.J., Thomas, M.J., Anderson, R.A., Mason,


J.I., Hillier, S.G., 1997. Differential expression of messenger ribonucleic acids encoding 11beta-hydroxysteroid dehydrogenase
types 1 and 2 in human granulosa cells. J. Clin. Endocrinol.
Metab. 82, 2006 20099.
Tetsuka, M., Milne, M., Hillier, S.G., 1998b. Expression of oestrogen
receptor isoforms in relation to enzymes of oestrogen synthesis in
rat ovary. Mol. Cell Endocrinol. 141, 29 35.
Tetsuka, M., Haines, L.C., Milne, M., Simpson, G.E., Hillier, S.G.,
1999a. Regulation of 11beta-hydroxysteroid dehydrogenase type 1
gene expression by LH and interleukin-1beta in cultured rat
granulosa cells. J. Endocrinol 163, 417 423.
Tetsuka, M., Milne, M., Simpson, G., Hillier, S.G., 1999b. Expression of 11b-hydroxysteroid dehydrogenase, glucocorticoid receptor and mineralocorticoid receptor genes in rat ovary. Biol.
Reprod. 60, 330 335.
Turner, I.M., Saunders, P.T.K., Shimasaki, S., Hillier, S.G., 1989.
Regulation of inhibin subunit gene expression by FSH and estradiol in cultured rat granulosa cells. Endocrinology 125, 2790
2792.
Yding-Andersen, C., Morineau, G., Fukuda, M., Westergaard, L.G.,
Ingerslev, H.J., Fiet, J., Byskov, A.G., 1999. Assessment of the
follicular cortisol:cortisone ratio. Hum. Reprod. 14, 1562 1568.
Yong, E.L., Baird, D.T., Hillier, S.G., 1992a. Mediation of gonadotrophin-stimulated growth and differentiation of human
granulosa cells by adenosine-3,5-monophosphate: one molecule,
two messages. Clin. Endocrinol. (Oxf) 37, 51 58.
Yong, E.L., Baird, D.T., Yates, R., Reichert, L.E. Jr, Hillier, S.G.,
1992b. Hormonal regulation of the growth and steroidogenic
function of human granulosa cells. J. Clin. Endocrinol. Metab.
74, 842 849.
Yong, P.Y.K., Thong, K.J., Andrew, R., Walker, B.R., Hillier, S.G.,
2000. Development related increase in cortisol biosynthesis by
himan granulosa cells. J. Clin. Endocrinol. Metab., 85, 4728
4733.
Zeleznik, A.J., Hillier, S.G., 1984. The role of gonadotropins in the
selection of the preovulatory follicle. Clin. Obstet. Gynecol. 27,
927 940.
Zeleznik, A.J., Midgley, A.R. Jr, Reichert, L.E. Jr, 1974. Granulosa
cell maturation in the rat: increased binding of human chorionic
gonadotropin following treatment with follicle stimulating hormone in vivo. Endocrinology 95, 818 825.
Zhou, J., Bondy, C., 1993. Anatomy of the human ovarian insulinlike growth factor system. Biol. Reprod. 48, 467 482.

Das könnte Ihnen auch gefallen