You are on page 1of 9

CSIRO PUBLISHING

www.publish.csiro.au/journals/rfd Reproduction, Fertility and Development, 2011, 23, 23–31

Recent insights into oocyte]follicle cell interactions


provide opportunities for the development of new
approaches to in vitro maturation

Robert B. Gilchrist

Robinson Institute, Research Centre for Reproductive Health, and School of Paediatrics
and Reproductive Health, Discipline of Obstetrics and Gynaecology, Medical School,
University of Adelaide, Adelaide, SA 5005, Australia.
Email: robert.gilchrist@adelaide.edu.au

Abstract. The last 5–10 years of research in ovarian and oocyte biology has delivered some major new advances in
knowledge of the molecular and cellular processes regulating oocyte maturation and oocyte developmental competence.
These new insights include, among others: (1) the knowledge that oocytes regulate granulosa and cumulus cell
differentiation, ovulation rate and fertility via the secretion of soluble paracrine growth factors; (2) new perspectives
on the participation of cyclic nucleotides, phosphodiesterases and gap junctions in the regulation of oocyte meiotic arrest
and resumption; and (3) the new appreciation of the mechanisms of LH-induced oocyte maturation and ovulation mediated
by the follicular cascade of epidermal growth factor (EGF)-like peptides, the EGF receptor and their intracellular second
messengers. These recent insights into oocyte–follicle cell interactions provide opportunities for the development of new
approaches to oocyte in vitro maturation (IVM). Laboratory IVM methodologies have changed little over the past 20–
30 years and IVM remains notably less efficient than hormone-stimulated IVF, limiting its wider application in
reproductive medicine and animal breeding. The challenge for oocyte biologists and clinicians practicing IVM is to
modernise clinical IVM systems to benefit from these new insights into oocyte–follicle cell interactions in vivo.

Additional keywords: artificial reproductive technology (ART), cAMP, embryo production, GDF9, oocyte IVM,
simulated physiological oocyte maturation (SPOM).

Background normal offspring (Schroeder and Eppig 1984; Shu-Chi et al.


Oocyte quantity and quality are fundamentally rate-limiting in 2006; Eppig et al. 2009). IVM has the advantage over conven-
female fertility; however, the nature of the molecular and cel- tional IVF as it eliminates or drastically reduces the use of
lular processes that regulate oocyte quality is not well defined. gonadotrophins for ovarian hyperstimulation, thereby reducing
Oocyte quality or oocyte developmental competence refers to the contraindications and cost to patients, and avoiding the
an oocyte’s intrinsic capacity to undergo meiosis and support potential for hyperstimulation-induced abnormalities in the
fertilisation, preimplantation embryo development and fetal developing embryo (Baart et al. 2007; Market-Velker et al.
development to term to produce a healthy offspring. Oocyte 2010). The notable disadvantage of IVM is its significantly
developmental competence is gradually acquired in the devel- lower efficiency compared with hormone-driven IVF, in terms
oping ovarian follicle. Oogenesis and folliculogenesis occur in of embryos and offspring generated per oocyte collected (Child
unison and are orchestrated by a highly complex interplay of et al. 2002; Eppig et al. 2009). This factor alone has greatly
endocrine, paracrine and autocrine signals that are exchanged restricted the use of IVM in human reproductive medicine, where
between the oocyte and somatic compartment of the follicle. it is estimated that just 1300 IVM babies have been born over
Improving our understanding of the intricate bidirectional the past 30 years (Suikkari 2008). By contrast, in 2005 alone,
communication axes between the oocyte and follicular somatic ,133 000 viable cattle IVM offspring were produced (Thibier
cells is opening new opportunities in artificial reproductive 2006) and currently the use of IVM and in vitro embryo produc-
technologies (ART). tion is increasing dramatically in this sector. Hence, for human
One such ART is oocyte in vitro maturation (IVM), which is a and veterinary applications, there is great incentive to improve
technique where immature oocytes are collected from mid-sized the efficiency of IVM. This review will: (1) briefly examine the
antral follicles, generally from unstimulated ovaries, and cul- substantial advances made in recent years in our understanding
tured, matured and fertilised in vitro (Edwards 1965) to produce of oocyte–follicle cell interactions and the mechanisms regulat-
embryos that, after transfer, appear to generate developmentally ing oocyte maturation in vivo and (2) discuss how this new

Ó IETS 2011 10.1071/RD10225 1031-3613/11/010023


24 Reproduction, Fertility and Development R. B. Gilchrist

knowledge might be used to modernise oocyte IVM methodol- enhanced fecundity or ovarian disease in a highly species-
ogies specifically to improve embryo yield from IVM. specific manner (McNatty et al. 2004).
Pertinent to the subject of this review is the knowledge
that a principal function of OSFs during the antral phase of
New insights into oocyte]follicle cell interactions
folliculogenesis is to direct the differentiation of granulosa
Oocyte control of granulosa and cumulus cell function cells into cumulus cells and to maintain this distinct phenotype
An important new paradigm in ovarian and oocyte biology to in the follicle (Eppig et al. 1997; Li et al. 2000). This appears
emerge over the past decade is the concept of oocyte-secreted to be achieved by OSFs establishing a localised concentration
factor (OSF)-regulation of folliculogenesis. This topic has or morphogenic gradient emanating from the oocyte (Hussein
already been the subject of several reviews and so will not be et al. 2005). The consequences of OSF signalling to cumulus
dealt with extensively here (some reviews on this topic: Eppig cell function are numerous (Gilchrist et al. 2008) and this
2001; Gilchrist et al. 2004, 2008; McNatty et al. 2006; remains an active area of research. Some of the actions of
Scaramuzzi et al. 2010). Oocytes regulate granulosa cell growth OSFs on CCs include stimulation of growth and prevention of
and differentiation through the production of soluble paracrine death (Vanderhyden et al. 1992; Gilchrist et al. 2001, 2006;
growth factors. The two most important OSFs are growth dif- Hussein et al. 2005), inhibition of luteinisation (Vanderhyden
ferentiation factor 9 (GDF9) and bone morphogenetic protein 15 et al. 1993; Eppig et al. 1997; Li et al. 2000), regulation of
(BMP15), as both are essential for folliculogenesis and female energy metabolism (Sutton et al. 2003a; Sugiura et al. 2005)
fertility in a species-specific manner (Dong et al. 1996; and sterol biosynthesis (Su et al. 2008) and the regulation of
Galloway et al. 2000). Oocyte-secreted GDF9 and BMP15 act in CC expansion (Buccione et al. 1990; Salustri et al. 1990;
a paracrine mode on granulosa and cumulus cells (CCs) through Fig. 1). By means of OSFs, the oocyte appears to direct the
their receptors, bone morphogenetic protein receptor type-II CCs to perform functions for which the oocyte itself has a poor
(BMPRII) and activin receptor-like kinases (ALKs), to activate capacity (e.g. glycolysis; Sutton-McDowall et al. 2010).
Sma- and Mad-related (SMAD) intracellular signal transducers Hence, the capacity of an oocyte to control such CC functions
(Kaivo-oja et al. 2006). Aberrant expression of any of these is likely to be a crucial function required for acquisition of its
ligands, receptors or second messengers causes sterility, own developmental competence.

EGFR
AREG
EGFR
EREG
BTC
PGE2 PTGER2

ERK1/2 p38 ERK1/2


AREG GPR3 PKA
EREG
BTC X ? MPF
Cx43 ↓ cAMP
?
cAMP
p38 ↓ cGMP ↑ PDE3
GVBD
HAS2 Sterol biosynthesis ? 5-AMP
cAMP ↓ cGMP TSG6
PTX3 Metabolism
GDF9
GC Luteinisation BMP15
? GC
BMPRII
SMAD2/3
LHR NO FSHR ALK5/6
ANP
ERK1/2
?
SRC
LH FSH
EGFR

Granulosa cell Cumulus cell Oocyte

Fig. 1. Model of new perspectives on oocyte–follicle cell interactions and how these may interact to regulate
oocyte maturation in vivo. The resumption of oocyte meiosis in vivo is a highly orchestrated, complex process that
integrates oocyte paracrine signals with an array of maternal endocrine, granulosa/cumulus paracrine and gap-
junctional signals. This highly integrated interaction between oocyte and follicular somatic cells is unlikely
to occur during spontaneous IVM, as it is clinically practiced at present. This hypothetical model of meiotic
induction in vivo is not intended to be complete but rather to highlight the new appreciation of three processes:
(1) interaction of GDF9/BMP15–SMAD2/3 signalling and EGFR; (2) LH-induction of the EGF-like peptide
cascade in granulosa and CCs; and (3) the complex interplay between gap junctions, cGMP, PDE and cAMP.
Dotted arrows indicate signals of diminishing intensity. ‘X’ represents a hypothetical meiotic-inducing factor. See
text for other abbreviations.
Recent insights provide new opportunities for IVM Reproduction, Fertility and Development 25

Two very recent studies demonstrated an important relation- the idea that a transient pulse in cAMP produces a meiosis-
ship between OSF and epidermal growth factor receptor (EGFR) inducing signal (Dekel 1988; Downs and Chen 2008). Two
signalling in CCs (Sasseville et al. 2010; Su et al. 2010; Fig. 1). important recent studies have revealed that the ovulatory
It has been known for some time that there is an important gonadotrophin surge causes a drop in follicular and oocyte
interaction between signals from the oocyte and signals from the cGMP levels leading to upregulated oocyte PDE activity, which
follicle that intersect in CCs. This was initially demonstrated causes a fall in intraoocyte cAMP and meiotic resumption
by the dual requirement for OSFs and FSH for mouse cumulus– (Norris et al. 2009; Vaccari et al. 2009; Fig. 1). The above is a
oocyte complex (COC) expansion (Buccione et al. 1990; substantial oversimplification of the participation of cAMP,
Vanderhyden et al. 1990). Subsequently it was demonstrated cGMP and PDEs in the process of oocyte meiotic resumption
that the follicular signal is mediated by ERK1/2 in CCs, which and the detailed complexity of these signalling pathways
could be activated by FSH or epidermal growth factor (EGF; remains a topical area of investigation.
Su et al. 2002, 2003) and that the oocyte paracrine signal is The second major recent breakthrough is that the preovula-
mediated by GDF9/BMP15 activating SMAD2/3 in CCs tory LH surge induces a rapid secondary cascade of the EGF-
(Dragovic et al. 2005, 2007; Yoshino et al. 2006; Diaz et al. like peptides – amphiregulin (AREG), epiregulin (EREG) and
2007). It is now known, in the mouse at least, that participation b-cellulin (BTC) – in the somatic cells of the follicle (Park et al.
of these two signalling pathways is a prerequisite not only for 2004). These peptides are initially produced by the MGCs in
CC expansion but for many of the OSF-regulated CC functions response to LH and have autocrine effects on MGCs and
listed above. Recently we have shown that cross-talk between paracrine actions on CCs via their EGF receptors (Fig. 1). An
these two pathways is mediated by the EGFR–ERK1/2 pathway important emerging concept is that the EGF receptor is a central
enabling GDF9–SMAD3 signalling in granulosa and cumulus nexus in propagating the ovulatory LH signal from the granulosa
cells (Sasseville et al. 2010). This appears to be mediated by cells, through the cumulus cells to the oocyte (Hsieh et al. 2007;
activation of ERK1/2 through EGFR, which, in turn, phosphor- Reizel et al. 2010). Central to MGC/CC signal transduction
ylates SMAD3 in its linker region, enabling SMAD2/3- are the EGF-like peptides that activate the EGF receptor and
mediated gene transcription. Another recent study has shown ERK1/2 that transmits the intracellular signals (Park et al.
that oocytes, via GDF9/BMP15 signalling, promote EGFR 2004; Fan et al. 2009). Activation of ERK1/2 induces a cascade
expression in CCs via a mechanism requiring SMAD2/3 (Su of prostaglandin E2 (PGE2) and p38MAPK induction, which,
et al. 2010). Hence, it appears that there are complex, mutually in turn, amplifies EGF-like peptide production by MGCs and
dependent interactions between GDF9/BMP15–SMAD2/3 and CCs (Park et al. 2004; Ashkenazi et al. 2005; Shimada et al.
EGFR–ERK1/2 signalling in CCs and that these are at the nexus 2006; Downs and Chen 2008). The EGF-like peptides are potent
of oocyte–somatic communication (Fig. 1). This autoregulatory inducers of oocyte meiotic resumption via a mechanism requir-
loop between the oocyte and its supporting somatic cells is likely ing ERK1/2 (Su et al. 2002; Hsieh et al. 2007; Fan et al. 2009).
to be crucial to oocyte development. Details of cumulus cell ERK1/2-induced meiotic resumption are
not yet fully characterised, but may involve phosphorylation of
Follicular control of oocyte maturation connexin 43 (Cx43), leading to closure of granulosa–cumulus
In terms of follicle cell control of oocyte maturation, major and cumulus–cumulus gap junctions (Sela-Abramovich et al.
advances in understanding have been achieved in just the past 2005; Norris et al. 2008) and subsequent loss of the inhibitory
few years. While it has long been known that the preovulatory effects of follicular cAMP and cGMP (Sela-Abramovich et al.
LH surge induces oocyte maturation, the exact molecular 2006; Norris et al. 2009), or alternatively, production of a
mechanisms regulating this process have been the subject of stimulatory meiosis-inducing factor by the CCs, or some com-
debate for decades. It is evident that the follicular environment bination of the two (reviewed by Downs 2010; Fig. 1).
is responsible for both meiotic arrest of the oocyte at prophase I
(GV stage) and for meiotic resumption. A focal point of oocyte
meiotic regulation and resumption is cyclic AMP (cAMP), New opportunities to modernise oocyte IVM
which is synthesised in the oocyte by constitutively active Given the substantial advances made in recent years in our
G-protein-coupled receptors (GPR3; Mehlmann et al. 2002) understanding of oocyte–CC communication and the molecular
and is also supplied to the oocyte by mural granulosa cells cascades regulating the reinitiation of oocyte maturation in vivo,
(MGCs) and adjacent CCs through gap junctions (Anderson and an important challenge for oocyte biologists and clinicians
Albertini 1976; Fig. 1). High levels of intra-oocyte cAMP keep practicing IVM is to capitalise on these new insights to develop
the oocyte meiotically arrested by suppressing maturation pro- new IVM systems. Ideally, a new-generation IVM system
moting factor (MPF) activity by stimulating cAMP-dependent should recapitulate the key events that occur during oocyte
protein kinase A (PKA). The oocyte possesses a potent type 3 maturation in vivo; among others these should include:
phosphodiesterase (PDE3), the enzyme that degrades cAMP (1) appropriate OSF regulation of cumulus cell SMAD2/3 sig-
(Tsafriri et al. 1996). Importantly, the somatic cells of the fol- nalling and associated EGFR function; (2) careful management
licle also supply cyclic GMP (cGMP) to the oocyte (Törnell of fluxes of cAMP and cGMP; (3) appropriate induction of the
et al. 1991), which inhibits PDE activity. The paradox of cAMP EGF-like peptide/ERK1/2 signalling cascade; and (4) controlled
in the oocyte is that high levels maintain meiotic arrest loss of oocyte–cumulus cell gap-junctional communication.
throughout folliculogenesis yet it also mediates the meiotic- Furthermore, designing new approaches to IVM needs to be
inducing effects of gonadotrophins. This may be explained by undertaken with the explicit purpose in mind of maximising
26 Reproduction, Fertility and Development R. B. Gilchrist

New insights New opportunities resumption of meiosis in spontaneous IVM is hormone-


Oocyte–follicle
IVM
independent, even though hormones such as FSH are typically
interactions added. Critically, oocytes that undergo meiosis spontaneously
in vitro are likely to do so in the absence of the elaborate cascade
of endocrine and paracrine molecular signals that usually induce
1. Oocyte control of follicle cell 1. Appropriate cumulus cell
maturation in vivo (Park et al. 2004; Ashkenazi et al. 2005;
differentiation function during IVM Shimada et al. 2006; Norris et al. 2009; Vaccari et al. 2009;
Fig. 1). The spontaneous resumption of meiosis during standard
• Oocyte-secreted factors • Exogenous GDF9 and/or
BMP15 in IVM IVM techniques is unphysiological and we propose that this
compromises subsequent oocyte developmental competence
2. Follicular control of oocyte meiotic 2. Simulated physiological oocyte
(Gilchrist and Thompson 2007).
arrest and resumption maturation (SPOM)
Application of OSFs to improve IVM
• cGMP, cAMP, PDEs • cAMP/PDE-regulated
• EGFR, EGF-like peptides meiotic resumption Based on the premises that OSFs are fundamental regulators
• Cumulus peptide of CC differentiation and function, and that CC processes that
induction of maturation
facilitate oocyte developmental competence are likely to be
perturbed under conditions of standard spontaneous IVM, we
Fig. 2. New insights into oocyte–follicle cell interactions provide oppor-
tunities for the development of new approaches to IVM. A challenge for
hypothesised that treatment of COCs during IVM with exo-
oocyte biologists and IVM clinicians is to modernise clinical IVM systems to genous OSFs would enhance oocyte developmental competence
benefit from this new knowledge of oocyte–follicle cell interactions in vivo. (Fig. 3). We tested this by treating COCs during IVM with either
native OSFs (COC co-cultured with denuded oocytes) or par-
tially purified recombinant GDF9 or BMP15, or both. Both
IVM efficiency in terms of the production of developmentally approaches improved the capacity of bovine oocytes to proceed
competent healthy embryos (Fig. 2). to the blastocyst stage of embryo development; from 40%
(control) up to as high as ,60% (Hussein et al. 2006). Exposure
Problems with current IVM methodologies of oocytes to GDF9 or BMP15 also improved subsequent
The central limiting problem with current IVM practices is the embryo quality as evidenced by increased total and trophecto-
lower efficiency of IVM, in terms of embryo development, derm cell numbers. Treating mouse COCs during IVM with
implantation and live birth rates, compared with conventional recombinant GDF9 significantly increased the number of sur-
IVF where oocytes mature in vivo (Gilchrist et al. 2010). For viving fetuses after blastocyst transfer to foster mothers without
example, the live birth rate in mice from IVM was 21% com- adverse effects on fetal or placental morphology (Yeo et al.
pared with 52% using IVF (Eppig et al. 2009). Notably, this 2008). Furthermore, Romaguera et al. (2010) have recently
discrepancy in reproductive efficiency between IVM and IVF shown that native OSFs are able to improve the competence
has existed for decades and is common across mammalian of a cohort of oocytes that otherwise would have poor develop-
species. For example, embryo development or implantation mental potential.
rates are notably lower using IVM than IVF in mice, sheep, The precise molecular mechanisms underlying the improved
cattle and women (Leibfried-Rutledge et al. 1987; Thompson oocyte developmental competence with OSF-supplemented
et al. 1995; Child et al. 2002; Rizos et al. 2002; Nogueira et al. IVM are currently unclear. Attempts to elucidate this mechan-
2003). Moreover, the miscarriage rate in women using IVM ism have been hampered by (1) the technical complexities of
is significantly higher than in gonadotrophin-stimulated IVF conducting large-scale COC plus denuded oocyte co-culture
(Buckett et al. 2008). This lower efficiency of IVM compared experiments and (2) the relatively poor quality of recombinant
with hormone-stimulated IVF is the principal factor limiting GDF9 and BMP15 preparations available for research. Until
widespread use of IVM in human reproductive medicine. In very recently, the only GDF9/BMP15 preparations available
contrast, IVM and associated in vitro production of embryos is were in-house generated proteins that varied widely between
in widespread use in cattle artificial breeding and its use is rising research laboratories in terms of construction and purity.
dramatically in South America (Thibier 2006). Hence both of Recently, commercially available purified GDF9 and BMP15
these ART sectors would benefit significantly from a notable have become available from R&D Systems, although these
increase in IVM efficiency. preparations are notable in that they lack their respective
Essentially all human and veterinary ART clinics offering proregions. Current and future efforts to generate recombinant
commercial IVM use a simple, standard system of IVM called GDF9 and BMP15 proteins that are highly purified with appro-
spontaneous oocyte maturation. Using this approach, COCs priate proregion–mature region interactions (Mottershead et al.
are aspirated from unstimulated or mildly stimulated ovaries 2008; Li et al. 2009), are important if clinical IVM is to benefit
and cultured for 24–48 h in vitro, typically in complex medium from the applications of these proteins.
supplemented with serum or albumin (Sutton et al. 2003b). By Although the molecular mechanisms require further investi-
rapidly removing COCs from the meiotic-inhibiting influence of gation, it seems clear that OSFs participate in the regulation of
the follicle and the follicular fluid, oocytes irrevocably reinitiate oocyte developmental competence by directing fundamental
and complete meiosis in vitro (Pincus and Enzmann 1935; aspects of CC differentiation and function during folliculogen-
Edwards 1965); hence the term ‘spontaneous IVM’. The esis and oocyte maturation (Gilchrist et al. 2008; Fig. 3). Across
Recent insights provide new opportunities for IVM Reproduction, Fertility and Development 27

I P
PRI SMAD2/3
BM
L K 5 P
A
F9
GD P
II
BMPR SMAD1/5/8

BMP15 ALK6 P

? P
Ga arac
p rine
jun
cti Regulation of cumulus cell function
on
al Proliferation Metabolism
Apoptosis Expansion
Oocyte quality Luteinisation

Embryo development
Fetal viability

Fig. 3. Bidirectional communication between the oocyte and cumulus cells is required for oocyte
developmental competence. Oocytes control the differentiation of CCs through the secretion of GDF9
and BMP15. These act via CC receptors BMPRII, ALK5 and ALK6 to activate SMAD intracellular
transducer molecules, which, in concert with maternal signals such as FSH and the EGF-like peptides,
regulate a large range of CC functions. CCs in turn make substantial contributions to oocyte growth and
development via paracrine and gap-junctional-mediated processes. This oocyte–CC feedback loop is
likely to be a crucial determinant of oocyte developmental competence.

mammalian species, activation of the GDF9 signalling cascade clinical scenario in mind, whereby at pick-up, COCs are
in CCs by oocytes, including ERK1/2-mediated SMAD2/3 removed from the follicular environment and typically placed in
activation (Sasseville et al. 2010) and subsequent EGFR regula- a simple buffered collection medium that leads to a rapid and
tion (Su et al. 2010), is probably fundamental to the regulation of precipitous drop in oocyte cAMP levels (Vivarelli et al. 1983).
oocyte quality (Figs 1, 3). Indeed, oocyte developmental com- Accordingly, in SPOM we designed a pre-IVM phase to be used
petence or embryo post-transfer fetal survival is compromised in at oocyte pick-up, containing cAMP-modulating agents (for-
mutant mice with perturbed GDF9 and BMP15 expression (Su skolin þ IBMX), that generate a large rapid increase (4100-fold)
et al. 2004) or when GDF9 signalling is inhibited during IVM in COC cAMP levels, as occurs in COCs immediately after the
(Yeo et al. 2009). Considerable recent effort to identify pre- preovulatory gonadotrophin surge in vivo (Albuz et al. 2010).
dictors of oocyte quality has identified a considerable number of This approach is analogous to the use of invasive adenylate
CC genes that are specifically under OSF control (see review cyclase that has previously been used in bovine oocyte collection
Li et al. 2008). These include, among others, OSF-regulated medium (Aktas et al. 1995; Luciano et al. 1999; Guixue et al.
genes such as Has2, Ptgs2, Ptx3, Grem, Areg, Star and Ereg 2001). The pre-IVM phase was designed to cause an immediate
(McKenzie et al. 2004; Feuerstein et al. 2007; Assidi et al. 2008; increase to both CC and oocyte cAMP levels. This has the effect of
Fig. 1). These studies provide further weight to the notion that preventing immediate loss of oocyte–cumulus gap-junctional
GDF9/BMP15 regulation of CC function regulates oocyte communication at pick-up (Albuz et al. 2010) and simultaneously
quality (Gilchrist et al. 2008) and provide the basis for devel- loads the oocyte with cAMP, thereby preventing precocious
oping a rapid quantitative test of oocyte quality using a biopsy of spontaneous resumption of meiosis.
CCs (Smitz et al. 2010). The extended IVM phase of SPOM attenuates meiotic
resumption, which is overridden or induced by FSH. An
Simulated physiological oocyte maturation (SPOM): a new important feature of the IVM system is that oocytes are
IVM system based on induced oocyte maturation in vivo continuously exposed throughout maturation to a low to
Building on the significant new insights into mechanisms moderate concentration of a type 3-specific PDE3 inhibitor.
mediating oocyte maturation in vivo – specifically the roles of As PDE3s are principally expressed in the oocyte and not in
cAMP/cGMP/PDEs and the induction of maturation via EGFR- CCs (Tsafriri et al. 1996; Sasseville et al. 2009), this approach
ERK1/2 signalling (Fig. 1) – we have recently developed a new specially targets intraoocyte cAMP. The PDE3 inhibitor con-
approach to oocyte IVM called ‘simulated physiological oocyte centration is low but sufficient to notably impair meiotic
maturation’ (SPOM; Albuz et al. 2010). SPOM is an integrated maturation in the absence of a meiotic-inducing hormone. In
IVM system that includes a short pre-IVM phase (1–2 h) and an SPOM a relatively high concentration of FSH (100 mIU mL1)
extended IVM phase that synergise to generate high embryo and is crucial to override or ‘induce’ oocyte maturation in the
fetal yields following embryo transfer (Fig. 4). The pre-IVM presence of the PDE3 inhibitor. We have previously used a
phase is specifically designed with the human and veterinary similar approach to improve the developmental potential of
28 Reproduction, Fertility and Development R. B. Gilchrist

Fetal
yield

Conventional IVF Ovarian hyperstimulation


22%
(in vivo matured)
IVF/ET

Standard IVM 8%
IVM (FSH) IVF/ET
Simulated physiological
oocyte maturation 26%
(SPOM) Pre-IVM Extended IVM IVF/ET
(forskolin  IBMX) (cilostamide  FSH)

Fig. 4. Schematic illustration of the SPOM IVM system and how it differs from standard IVM and
conventional IVF. SPOM is characterised by a short pre-IVM phase (COC collection) where COCs are
exposed to agents that cause a spike in cAMP, and by an extended IVM phase where FSH induces oocyte
maturation in the continuous presence of an oocyte-specific PDE inhibitor. SPOM generates murine
fertilisation, blastocyst and implantation rates and fetal yield comparable to hormone-driven conventional
IVF. Figure modified from Albuz et al. (2010).

bovine oocytes (Thomas et al. 2004). In the SPOM system, the peptide–ERK1/2 cascade, have now been characterised in
pre-IVM and IVM phases synergise to substantially improve detail. It is now incumbent on oocyte IVM scientists and clin-
oocyte developmental competence relative to standard sponta- icians to incorporate this new knowledge into new IVM systems
neous IVM, to levels that match hormone-stimulated conven- for genetic improvement in domestic animals and clinical
tional IVF (Albuz et al. 2010; Fig. 4). infertility treatment that are designed to improve oocyte
It is notable that a model of hormone-dependent (using FSH, developmental competence and produce a high yield of healthy
EGF, FF-MAS, etc.) induced oocyte maturation has been used embryos and offspring.
for decades by mouse oocyte biologists to study the fine detail
of meiotic regulation (Downs et al. 1988), but curiously this Acknowledgements
mode of oocyte maturation has rarely been used as a means of I would like to acknowledge collaborators who have worked closely with me
generating embryos. SPOM differs from ‘biphasic IVM’, which over many years on these topics; Jeremy Thompson, Firas Albuz, David
also typically exploits PDE inhibitors. Biphasic IVM differs Armstrong, David Mottershead, François Richard, Lesley Ritter, Maxime
notably in that PDE inhibitors or other meiotic inhibitors are Sasseville, Johan Smitz and Melanie Sutton-McDowall. Critical review
used at sufficient concentration to block meiotic resumption of the manuscript by Maxime Sasseville, Karine Reynaud and Jeremy
Thompson is greatly appreciated. I would like to thank Graeme Martin for
for a defined period (usually ,24 h), after which the inhibitor is
the artwork provided in Fig. 3. This research area was funded by the National
washed out so that the oocyte then undergoes spontaneous Health and Medical Research Council of Australia (Grants 519235, 453556
maturation (reviews Smitz et al. 2004; Gilchrist and Thompson and Fellowship 465415), by the Australian Research Council (Grant
2007). SPOM is a form of hormone-dependent oocyte matura- LP0562536) and by Cook Medical Pty Ltd.
tion as sufficient FSH is required to drive meiotic induction in
the presence of the type 3 PDE inhibitor (Fig. 4). Furthermore, References
FSH induction of meiotic resumption occurs by a mechanism Aktas, H., Wheeler, M. B., Rosenkrans, C. F., Jr, First, N. L., and Leibfried
that requires the EGF receptor (Albuz et al. 2010), which is Rutledge, M. L. (1995). Maintenance of bovine oocytes in prophase
likely to involve FSH/cAMP-induced expression of EGF-like of meiosis I by high [cAMP]i. J. Reprod. Fertil. 105(2), 227–235.
peptides by CCs (Downs and Chen 2008). By generating high doi:10.1530/JRF.0.1050227
levels of cAMP in the oocyte and the induced nature of oocyte Albuz, F. K., Sasseville, M., Lane, M., Armstrong, D. T., Thompson, J. G.,
maturation, SPOM mimics some of the key, newly characterised and Gilchrist, R. B. (2010). Simulated physiological oocyte maturation
molecular signals that occur during oocyte maturation in vivo. (SPOM): a novel in vitro maturation system that substantially improves
embryo yield and pregnancy outcomes. Hum. Reprod. doi:10.1093/
Continued development of IVM methodologies that build on our
HUMREP/DEQ246
new knowledge of mechanisms regulating oocyte maturation Anderson, E., and Albertini, D. F. (1976). Gap junctions between the oocyte
in vivo should lead to further increases in IVM efficiency and and companion follicle cells in the mammalian ovary. J. Cell Biol. 71(2),
embryo production in vitro. 680–686. doi:10.1083/JCB.71.2.680
Ashkenazi, H., Cao, X., Motola, S., Popliker, M., Conti, M., and Tsafriri, A.
Conclusions (2005). Epidermal growth factor family members: endogenous media-
tors of the ovulatory response. Endocrinology 146(1), 77–84.
Substantial progress has been made in recent years, largely
doi:10.1210/EN.2004-0588
using the mouse model, into long-standing controversies over Assidi, M., Dufort, I., Ali, A., Hamel, M., Algriany, O., Dielemann, S.,
the detailed cellular and molecular processes regulating and Sirard, M. A. (2008). Identification of potential markers of oocyte
oocyte maturation in vivo. New signalling systems that were competence expressed in bovine cumulus cells matured with follicle-
largely unknown 10 years ago, in particular, the oocyte signal- stimulating hormone and/or phorbol myristate acetate in vitro. Biol.
ling system using GDF9/BMP15–SMAD2/3 and the EGF-like Reprod. 79(2), 209–222. doi:10.1095/BIOLREPROD.108.067686
Recent insights provide new opportunities for IVM Reproduction, Fertility and Development 29

Baart, E. B., Martini, E., Eijkemans, M. J., Van Opstal, D., Beckers, N. G., cells are essential for female fertility. Science 324(5929), 938–941.
Verhoeff, A., Macklon, N. S., and Fauser, B. C. (2007). Milder ovarian doi:10.1126/SCIENCE.1171396
stimulation for in vitro fertilization reduces aneuploidy in the human Feuerstein, P., Cadoret, V., Dalbies-Tran, R., Guerif, F., Bidault, R., and
preimplantation embryo: a randomized controlled trial. Hum. Reprod. Royere, D. (2007). Gene expression in human cumulus cells: one
22(4), 980–988. doi:10.1093/HUMREP/DEL484 approach to oocyte competence. Hum. Reprod. 22(12), 3069–3077.
Buccione, R., Vanderhyden, B. C., Caron, P. J., and Eppig, J. J. (1990). FSH- doi:10.1093/HUMREP/DEM336
induced expansion of the mouse cumulus oophorus in vitro is dependent Galloway, S. M., McNatty, K. P., Cambridge, L. M., Laitinen, M. P.,
upon a specific factor(s) secreted by the oocyte. Dev. Biol. 138(1), Juengel, J. L., et al. (2000). Mutations in an oocyte-derived growth
16–25. doi:10.1016/0012-1606(90)90172-F factor gene (BMP15) cause increased ovulation rate and infertility in a
Buckett, W. M., Chian, R. C., Dean, N. L., Sylvestre, C., Holzer, H. E., and dosage-sensitive manner. Nat. Genet. 25(3), 279–283. doi:10.1038/
Tan, S. L. (2008). Pregnancy loss in pregnancies conceived after in vitro 77033
oocyte maturation, conventional in vitro fertilization, and intracyto- Gilchrist, R. B., and Thompson, J. G. (2007). Oocyte maturation: emerging
plasmic sperm injection. Fertil. Steril. 90, 546–550. doi:10.1016/ concepts and technologies to improve developmental potential in vitro.
J.FERTNSTERT.2007.06.107 Theriogenology 67(1), 6–15. doi:10.1016/J.THERIOGENOLOGY.
Child, T. J., Phillips, S. J., Abdul-Jalil, A. K., Gulekli, B., and Tan, S. L. 2006.09.027
(2002). A comparison of in vitro maturation and in vitro fertilization Gilchrist, R. B., Ritter, L. J., and Armstrong, D. T. (2001). Mouse oocyte
for women with polycystic ovaries. Obstet. Gynecol. 100(4), 665–670. mitogenic activity is developmentally coordinated throughout follicu-
doi:10.1016/S0029-7844(02)02193-2 logenesis and meiotic maturation. Dev. Biol. 240(1), 289–298.
Dekel, N. (1988). Regulation of oocyte maturation. The role of cAMP. doi:10.1006/DBIO.2001.0451
Ann. N. Y. Acad. Sci. 541, 211–216. doi:10.1111/J.1749-6632.1988. Gilchrist, R. B., Ritter, L. J., and Armstrong, D. T. (2004). Oocyte–somatic
TB22258.X cell interactions during follicle development in mammals. Anim.
Diaz, F. J., Wigglesworth, K., and Eppig, J. J. (2007). Oocytes determine Reprod. Sci. 82]83, 431–446. doi:10.1016/J.ANIREPROSCI.2004.
cumulus cell lineage in mouse ovarian follicles. J. Cell Sci. 120(8), 05.017
1330–1340. doi:10.1242/JCS.000968 Gilchrist, R. B., Ritter, L. J., Myllymaa, S., Kaivo-Oja, N., Dragovic, R. A.,
Dong, J., Albertini, D. F., Nishimori, K., Kumar, T. R., Lu, N., and Matzuk, Hickey, T. E., Ritvos, O., and Mottershead, D. G. (2006). Molecular
M. M. (1996). Growth differentiation factor-9 is required during basis of oocyte-paracrine signalling that promotes granulosa cell pro-
early ovarian folliculogenesis. Nature 383(6600), 531–535. doi:10.1038/ liferation. J. Cell Sci. 119(18), 3811–3821. doi:10.1242/JCS.03105
383531A0 Gilchrist, R. B., Lane, M., and Thompson, J. G. (2008). Oocyte-secreted
Downs, S. M. (2010). Regulation of the G2/M transition in rodent oocytes. factors: regulators of cumulus cell function and oocyte quality. Hum.
Mol. Reprod. Dev. 77(7), 566–585. doi:10.1002/MRD.21175 Reprod. Update 14(2), 159–177. doi:10.1093/HUMUPD/DMM040
Downs, S. M., and Chen, J. (2008). EGF-like peptides mediate FSH-induced Gilchrist, R. B., Smitz, J. E. J., and Thompson, J. G. (2010). Current
maturation of cumulus cell-enclosed mouse oocytes. Mol. Reprod. Dev. status and future trends of the clinical practice of human oocyte
75(1), 105–114. doi:10.1002/MRD.20781 in vitro maturation. In ‘Human Assisted Reproductive Technology:
Downs, S. M., Daniel, S. A., and Eppig, J. J. (1988). Induction of maturation Future Trends in Laboratory & Clinical Practice’. (Eds D. K. Gardner,
in cumulus cell-enclosed mouse oocytes by follicle-stimulating B. R. M. B. Rizk and T. Falcone.) (Cambridge University Press:
hormone and epidermal growth factor: evidence for a positive stimulus Cambridge.) (in press)
of somatic cell origin. J. Exp. Zool. 245(1), 86–96. doi:10.1002/JEZ. Guixue, Z., Luciano, A. M., Coenen, K., Gandolfi, F., and Sirard, M. A.
1402450113 (2001). The influence of cAMP before or during bovine oocyte matura-
Dragovic, R. A., Ritter, L. J., Schulz, S. J., Amato, F., Armstrong, D. T., and tion on embryonic developmental competence. Theriogenology 55(8),
Gilchrist, R. B. (2005). Role of oocyte-secreted growth differentiation 1733–1743. doi:10.1016/S0093-691X(01)00516-7
factor 9 in the regulation of mouse cumulus expansion. Endocrinology Hsieh, M., Lee, D., Panigone, S., Horner, K., Chen, R., Theologis, A., Lee,
146(6), 2798–2806. doi:10.1210/EN.2005-0098 D. C., Threadgill, D. W., and Conti, M. (2007). Luteinizing hormone-
Dragovic, R. A., Ritter, L. J., Schulz, S. J., Amato, F., Thompson, J. G., dependent activation of the epidermal growth factor network is essential
Armstrong, D. T., and Gilchrist, R. B. (2007). Oocyte-secreted for ovulation. Mol. Cell. Biol. 27(5), 1914–1924. doi:10.1128/MCB.
factor activation of SMAD 2/3 signalling enables initiation of mouse 01919-06
cumulus cell expansion. Biol. Reprod. 76(5), 848–857. doi:10.1095/ Hussein, T. S., Froiland, D. A., Amato, F., Thompson, J. G., and Gilchrist,
BIOLREPROD.106.057471 R. B. (2005). Oocytes prevent cumulus cell apoptosis by maintaining a
Edwards, R. G. (1965). Maturation in vitro of mouse, sheep, cow, pig, rhesus morphogenic paracrine gradient of bone morphogenetic proteins. J. Cell
monkey and human ovarian oocytes. Nature 208(5008), 349–351. Sci. 118(22), 5257–5268. doi:10.1242/JCS.02644
doi:10.1038/208349A0 Hussein, T. S., Thompson, J. G., and Gilchrist, R. B. (2006). Oocyte-secreted
Eppig, J. J. (2001). Oocyte control of ovarian follicular development and factors enhance oocyte developmental competence. Dev. Biol. 296(2),
function in mammals. Reproduction 122(6), 829–838. doi:10.1530/REP. 514–521. doi:10.1016/J.YDBIO.2006.06.026
0.1220829 Kaivo-oja, N., Jeffery, L. A., Ritvos, O., and Mottershead, D. G. (2006).
Eppig, J. J., Wigglesworth, K., Pendola, F., and Hirao, Y. (1997). Murine Smad signalling in the ovary. Reprod. Biol. Endocrinol. 4, 21–34.
oocytes suppress expression of luteinizing hormone receptor messenger doi:10.1186/1477-7827-4-21
ribonucleic acid by granulosa cells. Biol. Reprod. 56(4), 976–984. Leibfried-Rutledge, M. L., Critser, E. S., Eyestone, W. H., Northey, D. L.,
doi:10.1095/BIOLREPROD56.4.976 and First, N. L. (1987). Development potential of bovine oocytes
Eppig, J. J., O’Brien, M. J., Wigglesworth, K., Nicholson, A., Zhang, W., and matured in vitro or in vivo. Biol. Reprod. 36(2), 376–383. doi:10.1095/
King, B. A. (2009). Effect of in vitro maturation of mouse oocytes on the BIOLREPROD36.2.376
health and lifespan of adult offspring. Hum. Reprod. 24(4), 922–928. Li, Q., McKenzie, L. J., and Matzuk, M. M. (2008). Revisiting oocyte–
doi:10.1093/HUMREP/DEN466 somatic cell interactions: in search of novel intrafollicular predictors and
Fan, H. Y., Liu, Z., Shimada, M., Sterneck, E., Johnson, P. F., Hedrick, S. M., regulators of oocyte developmental competence. Mol. Hum. Reprod.
and Richards, J. S. (2009). MAPK3/1 (ERK1/2) in ovarian granulosa 14(12), 673–678. doi:10.1093/MOLEHR/GAN064
30 Reproduction, Fertility and Development R. B. Gilchrist

Li, Q., Rajanahally, S., Edson, M. A., and Matzuk, M. M. (2009). Stable cumulus expansion. Mol. Endocrinol. 24(2), 402–411. doi:10.1210/
expression and characterization of N-terminal tagged recombinant ME.2009-0267
human bone morphogenetic protein 15. Mol. Hum. Reprod. 15(12), Rizos, D., Ward, F., Duffy, P., Boland, M. P., and Lonergan, P. (2002).
779–788. doi:10.1093/MOLEHR/GAP062 Consequences of bovine oocyte maturation, fertilization or early embryo
Li, R., Norman, R. J., Armstrong, D. T., and Gilchrist, R. B. (2000). development in vitro versus in vivo: implications for blastocyst yield
Oocyte-secreted factor(s) determine functional differences between and blastocyst quality. Mol. Reprod. Dev. 61(2), 234–248. doi:10.1002/
bovine mural granulosa cells and cumulus cells. Biol. Reprod. 63(3), MRD.1153
839–845. doi:10.1095/BIOLREPROD63.3.839 Romaguera, R., Morato, R., Jimenez-Macedo, A. R., Catala, M., Roura, M.,
Luciano, A. M., Pocar, P., Milanesi, E., Modina, S., Rieger, D., Lauria, A., Paramio, M. T., Palomo, M. J., Mogas, T., and Izquierdo, D. (2010).
and Gandolfi, F. (1999). Effect of different levels of intracellular cAMP Oocyte-secreted factors improve embryo developmental competence of
on the in vitro maturation of cattle oocytes and their subsequent devel- COCs from small follicles in prepubertal goats. Theriogenology 74(6),
opment following in vitro fertilization. Mol. Reprod. Dev. 54(1), 1050–1059. doi:10.1016/J.THERIOGENOLOGY.2010.04.035
86–91. doi:10.1002/(SICI)1098-2795(199909)54:1o86::AID-MRD134 Salustri, A., Yanagishita, M., and Hascall, V. C. (1990). Mouse oocytes
3.0.CO;2-C regulate hyaluronic acid synthesis and mucification by FSH-stimulated
Market-Velker, B. A., Zhang, L., Magri, L. S., Bonvissuto, A. C., and Mann, cumulus cells. Dev. Biol. 138(1), 26–32. doi:10.1016/0012-1606(90)
M. R. (2010). Dual effects of superovulation: loss of maternal and 90173-G
paternal imprinted methylation in a dose-dependent manner. Hum. Mol. Sasseville, M., Albuz, F. K., Cote, N., Guillemette, C., Gilchrist, R. B., and
Genet. 19(1), 36–51. doi:10.1093/HMG/DDP465 Richard, F. J. (2009). Characterization of novel phosphodiesterases in
McKenzie, L. J., Pangas, S. A., Carson, S. A., Kovanci, E., Cisneros, P., the bovine ovarian follicle. Biol. Reprod. 81(2), 415–425. doi:10.1095/
Buster, J. E., Amato, P., and Matzuk, M. M. (2004). Human cumulus BIOLREPROD.108.074450
granulosa cell gene expression: a predictor of fertilization and embryo Sasseville, M., Ritter, L. J., Nguyen, T. M., Liu, F., Mottershead, D. G.,
selection in women undergoing IVF. Hum. Reprod. 19(12), 2869–2874. Russell, D. L., and Gilchrist, R. B. (2010). Growth differentiation factor
doi:10.1093/HUMREP/DEH535 9 signalling requires ERK1/2 activity in mouse granulosa and cumulus
McNatty, K. P., Moore, L. G., Hudson, N. L., Quirke, L. D., Lawrence, S. B., cells. J. Cell Sci. 123(18), 3166–3176. doi:10.1242/JCS.063834
et al. (2004). The oocyte and its role in regulating ovulation rate: Scaramuzzi, R. J., Baird, D. T., Campbell, B. K., Driancourt, M. A., Dupont,
a new paradigm in reproductive biology. Reproduction 128(4), 379–386. J., et al. (2010). Regulation of folliculogenesis and the determination of
doi:10.1530/REP.1.00280 ovulation rate: a review. Reprod. Fertil. Dev., in press.
McNatty, K. P., Lawrence, S., Groome, N. P., Meerasahib, M. F., Hudson, Schroeder, A. C., and Eppig, J. J. (1984). The developmental capacity of
N. L., Whiting, L., Heath, D. A., and Juengel, J. L. (2006). Meat and mouse oocytes that matured spontaneously in vitro is normal. Dev. Biol.
livestock association plenary lecture 2005. Oocyte signalling molecules 102(2), 493–497. doi:10.1016/0012-1606(84)90215-X
and their effects on reproduction in ruminants. Reprod. Fertil. Dev. Sela-Abramovich, S., Chorev, E., Galiani, D., and Dekel, N. (2005).
18(4), 403–412. doi:10.1071/RD05104 Mitogen-activated protein kinase mediates luteinizing hormone-
Mehlmann, L. M., Jones, T. L., and Jaffe, L. A. (2002). Meiotic arrest induced breakdown of communication and oocyte maturation in rat
in the mouse follicle maintained by a Gs protein in the oocyte. Science ovarian follicles. Endocrinology 146(3), 1236–1244. doi:10.1210/
297(5585), 1343–1345. doi:10.1126/SCIENCE.1073978 EN.2004-1006
Mottershead, D. G., Pulkki, M. M., Muggalla, P., Pasternack, A., Tolonen, Sela-Abramovich, S., Edry, I., Galiani, D., Nevo, N., and Dekel, N.
M., et al. (2008). Characterization of recombinant human growth (2006). Disruption of gap junctional communication within the ovarian
differentiation factor-9 signalling in ovarian granulosa cells. Mol. Cell. follicle induces oocyte maturation. Endocrinology 147(5), 2280–2286.
Endocrinol. 283(1–2), 58–67. doi:10.1016/J.MCE.2007.11.007 doi:10.1210/EN.2005-1011
Nogueira, D., Cortvrindt, R., De Matos, D. G., Vanhoutte, L., and Smitz, J. Shimada, M., Hernandez-Gonzalez, I., Gonzalez-Robayna, I., and Richards,
(2003). Effect of phosphodiesterase type 3 inhibitor on developmental J. S. (2006). Paracrine and autocrine regulation of epidermal
competence of immature mouse oocytes in vitro. Biol. Reprod. 69, growth factor-like factors in cumulus oocyte complexes and granulosa
2045–2052. doi:10.1095/BIOLREPROD.103.021105 cells: key roles for prostaglandin synthase 2 and progesterone
Norris, R. P., Freudzon, M., Mehlmann, L. M., Cowan, A. E., Simon, receptor. Mol. Endocrinol. 20(6), 1352–1365. doi:10.1210/ME.2005-
A. M., Paul, D. L., Lampe, P. D., and Jaffe, L. A. (2008). Luteinizing 0504
hormone causes MAP kinase-dependent phosphorylation and closure of Shu-Chi, M., Jiann-Loung, H., Yu-Hung, L., Tseng-Chen, S., Ming, I., and
connexin 43 gap junctions in mouse ovarian follicles: one of two paths Tsu-Fuh, Y. (2006). Growth and development of children conceived by
to meiotic resumption. Development 135(19), 3229–3238. doi:10.1242/ in vitro maturation of human oocytes. Early Hum. Dev. 82(10), 677–682.
DEV.025494 doi:10.1016/J.EARLHUMDEV.2006.01.012
Norris, R. P., Ratzan, W. J., Freudzon, M., Mehlmann, L. M., Krall, J., Smitz, J., Nogueira, D., Vanhoutte, L., De Matos, D. G., and Cortvrindt,
Movsesian, M. A., Wang, H., Ke, H., Nikolaev, V. O., and Jaffe, L. A. R. N. (2004). Oocyte in vitro maturation. In ‘Textbook of Assisted
(2009). Cyclic GMP from the surrounding somatic cells regulates cyclic Reproductive Techniques: Laboratory and Clinical Perspectives’. 2nd
AMP and meiosis in the mouse oocyte. Development 136(11), 1869– edn. (Eds D. K. Gardner, A. Weissman, C. Howles and Z. Shoham.)
1878. doi:10.1242/DEV.035238 pp. 125–61. (Martin Dunitz Ltd: London.)
Park, J. Y., Su, Y. Q., Ariga, M., Law, E., Jin, S. L., and Conti, M. (2004). Smitz, J., Thompson, J. G., and Gilchrist, R. B. (2010). The promise of in
EGF-like growth factors as mediators of LH action in the ovulatory vitro maturation in assisted reproduction and fertility preservation.
follicle. Science 303(5658), 682–684. doi:10.1126/SCIENCE.1092463 Semin. Reprod. Med., in press.
Pincus, G., and Enzmann, E. V. (1935). The comparative behaviour of Su, Y. Q., Wigglesworth, K., Pendola, F. L., O’Brien, M. J., and Eppig, J. J.
mammalian eggs in vivo and in vitro: I. The activation of ovarian eggs. (2002). Mitogen-activated protein kinase activity in cumulus cells is
J. Exp. Med. 62(5), 665–675. doi:10.1084/JEM.62.5.665 essential for gonadotrophin-induced oocyte meiotic resumption and
Reizel, Y., Elbaz, J., and Dekel, N. (2010). Sustained activity of the EGF cumulus expansion in the mouse. Endocrinology 143(6), 2221–2232.
receptor is an absolute requisite for LH-induced oocyte maturation and doi:10.1210/EN.143.6.2221
Recent insights provide new opportunities for IVM Reproduction, Fertility and Development 31

Su, Y. Q., Denegre, J. M., Wigglesworth, K., Pendola, F. L., O’Brien, M. J., Thompson, J. G., Gardner, D. K., Pugh, P. A., McMillan, W. H., and
and Eppig, J. J. (2003). Oocyte-dependent activation of mitogen- Tervit, H. R. (1995). Lamb birth-weight is affected by culture system
activated protein kinase (ERK1/2) in cumulus cells is required for utilized during in vitro pre-elongation development of ovine embryos.
the maturation of the mouse oocyte–cumulus cell complex. Dev. Biol. Biol. Reprod. 53(6), 1385–1391. doi:10.1095/BIOLREPROD53.6.1385
263(1), 126–138. doi:10.1016/S0012-1606(03)00437-8 Törnell, J., Billig, H., and Hillensjo, T. (1991). Regulation of oocyte
Su, Y. Q., Wu, X., O’Brien, M. J., Pendola, F. L., Denegre, J. N., Matzuk, maturation by changes in ovarian levels of cyclic nucleotides. Hum.
M. M., and Eppig, J. J. (2004). Synergistic roles of BMP15 and GDF9 Reprod. 6(3), 411–422.
in the development and function of the oocyte–cumulus cell complex Tsafriri, A., Chun, S. Y., Zhang, R., Hsueh, A. J., and Conti, M. (1996).
in mice: genetic evidence for an oocyte–granulosa cell regulatory loop. Oocyte maturation involves compartmentalization and opposing
Dev. Biol. 276(1), 64–73. doi:10.1016/J.YDBIO.2004.08.020 changes of cAMP levels in follicular somatic and germ cells: studies
Su, Y. Q., Sugiura, K., Wigglesworth, K., O’Brien, M. J., Affourtit, J. P., using selective phosphodiesterase inhibitors. Dev. Biol. 178(2),
Pangas, S. A., Matzuk, M. M., and Eppig, J. J. (2008). Oocyte regulation 393–402. doi:10.1006/DBIO.1996.0226
of metabolic cooperativity between mouse cumulus cells and oocytes: Vaccari, S., Weeks, J. L., 2nd, Hsieh, M., Menniti, F. S., and Conti, M.
BMP15 and GDF9 control cholesterol biosynthesis in cumulus cells. (2009). Cyclic GMP signalling is involved in the luteinizing hormone-
Development 135(1), 111–121. doi:10.1242/DEV.009068 dependent meiotic maturation of mouse oocytes. Biol. Reprod. 81(3),
Su, Y. Q., Sugiura, K., Li, Q., Wigglesworth, K., Matzuk, M. M., and Eppig, 595–604. doi:10.1095/BIOLREPROD.109.077768
J. J. (2010). Mouse oocytes enable LH-induced maturation of the Vanderhyden, B. C., Caron, P. J., Buccione, R., and Eppig, J. J. (1990).
cumulus–oocyte complex via promoting EGF receptor-dependent signal- Developmental pattern of the secretion of cumulus expansion-enabling
ling. Mol. Endocrinol. 24(6), 1230–1239. doi:10.1210/ME.2009-0497 factor by mouse oocytes and the role of oocytes in promoting granulosa
Sugiura, K., Pendola, F. L., and Eppig, J. J. (2005). Oocyte control of cell differentiation. Dev. Biol. 140(2), 307–317. doi:10.1016/0012-1606
metabolic cooperativity between oocytes and companion granulosa (90)90081-S
cells: energy metabolism. Dev. Biol. 279(1), 20–30. doi:10.1016/ Vanderhyden, B. C., Telfer, E. E., and Eppig, J. J. (1992). Mouse oocytes
J.YDBIO.2004.11.027 promote proliferation of granulosa cells from preantral and antral
Suikkari, A. M. (2008). In vitro maturation: its role in fertility treatment. follicles in vitro. Biol. Reprod. 46(6), 1196–1204. doi:10.1095/
Curr. Opin. Obstet. Gynecol. 20(3), 242–248. doi:10.1097/GCO. BIOLREPROD46.6.1196
0B013E3282F88E33 Vanderhyden, B. C., Cohen, J. N., and Morley, P. (1993). Mouse oocytes
Sutton, M. L., Cetica, P. D., Beconi, M. T., Kind, K. L., Gilchrist, R. B., regulate granulosa cell steroidogenesis. Endocrinology 133(1),
and Thompson, J. G. (2003a). Influence of oocyte-secreted factors and 423–426. doi:10.1210/EN.133.1.423
culture duration on the metabolic activity of bovine cumulus cell Vivarelli, E., Conti, M., De Felici, M., and Siracusa, G. (1983). Meiotic
complexes. Reproduction 126(1), 27–34. doi:10.1530/REP.0.1260027 resumption and intracellular cAMP levels in mouse oocytes treated
Sutton, M. L., Gilchrist, R. B., and Thompson, J. G. (2003b). Effects of with compounds which act on cAMP metabolism. Cell Differ. 12(5),
in vivo and in vitro environments on the metabolism of the cumulus– 271–276. doi:10.1016/0045-6039(83)90023-4
oocyte complex and its influence on oocyte developmental capacity. Yeo, C. X., Gilchrist, R. B., Thompson, J. G., and Lane, M. (2008).
Hum. Reprod. Update 9(1), 35–48. doi:10.1093/HUMUPD/DMG009 Exogenous growth differentiation factor 9 in oocyte maturation media
Sutton-McDowall, M. L., Gilchrist, R. B., and Thompson, J. G. (2010). The enhances subsequent embryo development and fetal viability in mice.
pivotal role of glucose metabolism in determining oocyte developmental Hum. Reprod. 23(1), 67–73. doi:10.1093/HUMREP/DEM140
competence. Reproduction 139(4), 685–695. doi:10.1530/REP-09-0345 Yeo, C. X., Gilchrist, R. B., and Lane, M. (2009). Disruption of bidirectional
Thibier, M. (2006). Transfers of both in vivo-derived and in vitro-produced oocyte–cumulus paracrine signalling during in vitro maturation reduces
embryos in cattle still on the rise and contrasted trends in other species in subsequent mouse oocyte developmental competence. Biol. Reprod.
2005. International Embryo Transfer Society Newsletter 24(4), 12–18. 80(5), 1072–1080. doi:10.1095/BIOLREPROD.108.073908
Thomas, R. E., Thompson, J. G., Armstrong, D. T., and Gilchrist, R. B. Yoshino, O., McMahon, H. E., Sharma, S., and Shimasaki, S. (2006). A
(2004). Effect of specific phosphodiesterase isoenzyme inhibitors during unique preovulatory expression pattern plays a key role in the physio-
in vitro maturation of bovine oocytes on meiotic and developmental logical functions of BMP-15 in the mouse. Proc. Natl. Acad. Sci. USA
capacity. Biol. Reprod. 71(4), 1142–1149. doi:10.1095/BIOLREPROD. 103(28), 10 678–10 683. doi:10.1073/PNAS.0600507103
103.024828

http://www.publish.csiro.au/journals/rfd

You might also like